Fact-checked by Grok 2 weeks ago

Platelet plug

The platelet plug, also known as the hemostatic plug or primary hemostatic plug, is a temporary aggregation of platelets that rapidly forms at the site of a vascular to the damaged and prevent excessive blood loss during the initial phase of , referred to as primary . This process occurs within seconds of and serves as a critical first line of defense, temporarily blocking the breach in the vessel wall much like a in a , while also helping to exclude and pathogens from entering the bloodstream. Unlike the more stable clot formed in secondary , the platelet plug is fragile and relies on subsequent mechanisms for reinforcement. The formation of the platelet plug involves a coordinated sequence of events beginning with vascular injury. Upon damage to the , immediate occurs to reduce flow and minimize initial loss at the injury site, exposing the underlying subendothelial , including fibers. Circulating platelets, which are small, anucleate cell fragments derived from megakaryocytes, then adhere to this exposed matrix primarily through the interaction of platelet Ib-IX-V receptors with (vWF), a protein that bridges platelets to the . This step is essential for localizing platelets at the injury site and initiating the response. Once adhered, platelets undergo , a transformative process triggered by contact with and signaling molecules like . Activated platelets change shape, extending to increase surface area, and release contents from their cytoplasmic granules, including (ADP), thromboxane A2 (TXA2), serotonin, and other mediators that amplify the response. TXA2, in particular, promotes further and recruits additional platelets to the site. Platelet aggregation follows, where activated platelets bind to one another via glycoprotein IIb/IIIa (GPIIb/IIIa) receptors that interact with fibrinogen or additional vWF, forming a dense, multilayered plug that effectively occludes the vessel defect. This aggregation is further enhanced by ADP binding to P2Y1 and receptors on nearby platelets, creating a self-perpetuating . Disruptions in platelet plug formation can lead to bleeding disorders, such as or , highlighting its indispensable role in maintaining vascular integrity. Conversely, excessive or inappropriate plug formation contributes to thrombotic conditions like . Understanding this mechanism has informed therapeutic strategies, including antiplatelet drugs like aspirin, which inhibit TXA2 synthesis to prevent pathological .

Introduction

Definition and components

The platelet plug, also known as the , is a temporary aggregate of activated platelets that forms rapidly at the site of vascular injury to seal small breaches in blood vessels, thereby initiating primary and preventing excessive blood loss. This structure is distinct from the more stable clot formed during secondary , as it relies primarily on platelet interactions rather than extensive factor activation. Key components of the platelet plug include platelets themselves, which serve as the primary cellular elements, along with adhesive proteins such as (vWF) that acts as a bridging molecule between platelets and the subendothelial matrix, fibrinogen that functions as a cross-linker to bind platelets together, and subendothelial that provides the initial binding site for platelet adhesion. vWF, secreted by endothelial cells and stored in platelet alpha-granules, facilitates platelet tethering under high shear conditions, while fibrinogen binds to activated platelet receptors to promote aggregation. exposed upon endothelial damage further recruits and activates platelets via glycoprotein VI and α2β1 receptors. Resting platelets are small, discoid fragments derived from megakaryocytes, typically measuring 2–4 μm in diameter and 0.5–1 μm in thickness, with a smooth, oval shape that allows efficient circulation. Upon activation, platelets undergo a dramatic morphological change, extending pseudopods and to form a spherical or irregular shape up to 3–5 μm in , enhancing their and aggregatory properties essential for plug formation. These activated platelets release granule contents and express surface receptors to consolidate the plug structure. The initial platelet plug typically comprises a small aggregate of platelets sufficient to occlude small injuries, serving as a provisional that requires reinforcement by in secondary .

Role in primary

Primary represents the initial phase of the hemostatic response to vascular injury, characterized by rapid followed by the formation of a platelet plug to achieve immediate control of bleeding, in contrast to the slower secondary involving the cascade and stabilization. This process is essential for sealing small breaches in the wall, preventing excessive blood loss through the swift recruitment and assembly of platelets at the injury site. The sequence of events in primary hemostasis begins with vascular spasm, or , which occurs almost immediately upon injury, typically within seconds, and lasts for several minutes, reducing blood flow to the damaged area and minimizing hemorrhage. This is rapidly succeeded by platelet plug formation, which occurs within seconds to a few minutes, where circulating platelets adhere to the exposed subendothelium and aggregate to create a temporary barrier. Subsequent reinforcement by strands from secondary hemostasis stabilizes the plug, ensuring durable . In physiological contexts, the platelet plug is particularly effective in arterioles and capillaries, where vessel diameters are small enough for the aggregated platelets to span and occlude the breach, providing sufficient immediate sealing without relying solely on . However, in larger vessels such as arteries or veins, the higher forces and greater size necessitate the full integration of secondary to prevent rebleeding. From an evolutionary perspective, the rapid formation of the platelet plug confers a significant advantage by enabling quick cessation of bleeding from minor injuries, thereby preventing and supporting in environments prone to frequent vascular , such as those encountered by early vertebrates. This underscores the adaptive progression of , balancing immediate response with long-term vascular integrity.

Historical development

Early observations

The earliest observations of what would later be identified as platelets date back to the mid-19th century, amid rudimentary advancements in that limited clear visualization of components. In 1842, French histologist Alfred Donné first described small, granular elements in , which he termed "globulin du " and interpreted as fat globules derived from , marking the initial recognition of these structures as a potential third element beyond erythrocytes and leukocytes. Subsequent investigators built on this vague sighting but struggled with interpretation due to technical constraints. In 1865, German biologist Max Schultze provided a more detailed account in his study of , noting abundant, irregular, colorless granular particles in normal blood—likely platelets—using improved microscopic techniques like a heated stage to observe living samples. Around the same period, in 1878, French physician Georges Hayem offered further insights by observing these "plaques" or small corpuscles in and blood, linking them tentatively to and providing the first approximate counts, though he viewed them as possible fragments of white cells. These early descriptions were marred by misconceptions, with the particles often dismissed as cellular debris, degenerated leukocytes, or artifacts from fragmentation, rather than distinct entities. The prevailing focus in 19th-century remained on gross clotting phenomena observable to the , as lacked the resolution for dynamics, hindering definitive classification until later innovations. A pivotal breakthrough came in 1882 with Italian pathologist Giulio Bizzozero's seminal work, which provided the first clear, systematic description of platelets—termed "blood dust" or "plaquettes"—as independent, disc-shaped elements (2-3 micrometers in diameter) circulating in living animal blood. Using advanced intravital microscopy on mesenteric vessels of guinea pigs and rabbits, Bizzozero demonstrated their rapid adhesion to injured , aggregation into plugs, and essential role in and , dispelling prior doubts and establishing platelets as key players in primary clot formation.

Key advancements in mechanisms

In the 1940s, Louis M. Tocantins advanced the understanding of by distinguishing primary hemostasis, mediated by platelet plug formation, from secondary hemostasis involving clot stabilization, based on experimental observations of times and clot dynamics in animal models. This differentiation highlighted the distinct roles of platelets in initial vascular sealing versus coagulation factors in clot reinforcement. In 1948, Kalman Laki and László Lorand identified a factor, now known as factor XIII or -stabilizing factor, that cross-links to enhance clot insolubility and stability, thereby integrating platelet plugs with durable networks. The mid-20th century marked a revival of platelet following decades of relative neglect since their initial description in the , spurred by improved and biochemical techniques that enabled detailed studies of platelet . A pivotal breakthrough came in when Gustav V. R. developed the platelet aggregometer, a photometric that allowed of platelet aggregation in by detecting changes in light transmission as platelets clump. In the same study, demonstrated that adenosine diphosphate (ADP), released from platelet-dense granules, acts as a key mediator of irreversible platelet aggregation, confirming its central role in amplifying primary . During the late 20th century, molecular characterizations of platelet receptors elucidated the mechanisms of and aggregation underlying plug formation. In the 1970s, Harvey J. Weiss and colleagues identified the role of (vWF) in mediating platelet to subendothelial surfaces under high , showing that vWF binds to a specific platelet receptor to tether platelets at injury sites. Concurrently, the glycoprotein Ib-IX-V (GPIb-IX-V) complex was recognized as the primary vWF receptor on platelets, with its absence in Bernard-Soulier syndrome linking GPIbα deficiencies to impaired and bleeding disorders. By the 1980s, the (GPIIb/IIIa, or αIIbβ3 ) was established as the fibrinogen receptor essential for platelet aggregation, as demonstrated by direct binding studies showing its activation-dependent conformational change enables cross-linking of platelets via fibrinogen bridges. In the 1990s, research on signaling pathways revealed bidirectional (inside-out and outside-in) mechanisms regulating GPIIb/IIIa in platelets, where agonist-induced intracellular signals promote ligand binding, while ligand engagement triggers cytoskeletal reorganization for plug consolidation. These insights, derived from studies of talin and kindlin interactions with tails, underscored how signaling cascades amplify platelet responses to ensure stable hemostatic plugs without excessive .

Mechanisms of formation

Platelet activation

Platelet activation follows and is a critical step in the formation of the platelet plug, initiated when circulating platelets encounter vascular injury and are exposed to subendothelial components or soluble agonists. Primary triggers include exposed in the damaged subendothelium, which binds to glycoprotein VI (GPVI) receptors on the platelet surface, and generated at the injury site, which interacts with protease-activated receptors (PAR1 and PAR4). Additional stimuli encompass (ADP) and (ATP) released from damaged cells or platelet-dense granules, acting via P2Y1, , and P2X1 receptors, as well as (TXA2), a potent synthesized de novo in activated platelets through the action of cyclooxygenase-1 and thromboxane synthase, binding to the TP receptor. These agonists collectively induce rapid intracellular signaling cascades essential for platelet priming. Upon stimulation, platelet involves complex intracellular signaling pathways that lead to cytoskeletal reorganization and functional transformations. of agonists to their receptors activates G-protein-coupled signaling (via Gαq, Gαi, and Gα12/13 subunits) and (ITAM)-linked pathways, culminating in () —either β for G-protein pathways or γ2 for ITAM signaling—which hydrolyzes to produce inositol 1,4,5-trisphosphate (IP3) and diacylglycerol (DAG). IP3 triggers calcium release from intracellular stores, elevating cytosolic calcium levels that drive rearrangement through and myosin light chain , transforming the discoid platelet into an ameboid shape with extended for enhanced motility and interaction potential. This shape change is further modulated by Rho and (), ensuring efficient platelet responsiveness. A hallmark of activation is the release of granule contents, which amplifies the response through autocrine and . Alpha granules discharge proteins such as P-selectin (promoting leukocyte interactions) and (vWF, aiding adhesion), while dense granules secrete , ATP, serotonin, and calcium, further recruiting and activating nearby platelets. Lysosomal granules release hydrolytic enzymes like cathepsins, contributing to matrix degradation at the injury site. Concurrently, surface membrane changes occur, including the externalization of (PS) on the outer leaflet via activation, providing a procoagulant surface for generation and formation. αIIbβ3 (GPIIb/IIIa) undergoes conformational activation from a low-affinity to high-affinity state, mediated by talin-1 and kindlin-3 binding to the β3 tail, enabling fibrinogen bridging in subsequent steps. These events are tightly regulated to prevent aberrant activation in intact vessels. The entire process unfolds rapidly, with initial shape change and commencing within seconds of exposure, followed by granule release and activation within 10-30 seconds under physiological conditions. Full amplification via released mediators sustains activation for minutes, ensuring timely formation without excessive delay.

Platelet adhesion

Platelet is the initial step in primary where circulating platelets attach to the exposed subendothelial matrix following vascular injury, which triggers subsequent platelet activation and prepares the cells for further interactions with the vessel wall. Under physiological conditions, adhesion occurs rapidly under blood flow, particularly in high-shear environments such as arteries. Primary adhesion involves the glycoprotein complex GPIb-IX-V on the platelet surface binding to multimeric (vWF) immobilized on the subendothelium. This interaction is shear-dependent and crucial under high flow rates, where vWF unfolds to expose its A1 domain, enabling transient tethering and rolling of platelets along the damaged site. Secondary adhesion provides firmer attachment as platelets interact directly with exposed fibers via the GPVI receptor, which recognizes the , and the integrin α₂β₁, which binds to specific collagen motifs like the glycine-proline-hydroxyproline repeats. These interactions stabilize the initial contact, allowing platelets to spread and prepare for subsequent aggregation. The efficiency of platelet adhesion is highly dependent on shear rates, being most effective at arterial levels of $1000-5000 \, \mathrm{s}^{-1}, where GPIb-IX-V-vWF bonds form catch-bonds that strengthen under force. Initial rolling can also be facilitated by P-selectin expressed on activated , interacting with platelet ligands to slow downflowing platelets near the injury site. vWF, a large multimeric , is synthesized by endothelial cells and megakaryocytes and stored in Weibel-Palade bodies or alpha-granules, respectively. To regulate adhesion and prevent pathological thrombosis, the metalloprotease cleaves ultra-large vWF multimers into smaller, less adhesive forms shortly after secretion.

Platelet aggregation

Platelet aggregation involves the assembly of activated platelets into a cohesive mass through intercellular bridges, primarily mediated by the activation of the (GPIIb/IIIa, or αIIbβ3 ) receptor. Upon platelet activation, this undergoes a conformational change that exposes its fibrinogen-binding site, allowing soluble fibrinogen or multimers in to bind and cross-link adjacent platelets, thereby facilitating the formation of stable platelet-platelet interactions. This process is amplified by autocrine and paracrine signals released from activated platelets. (ADP), secreted from platelet dense granules, binds to the receptor on the platelet surface, which is a G_i-coupled receptor that inhibits adenylate cyclase, reduces cyclic AMP levels, and sustains GPIIb/IIIa activation to promote ongoing aggregation. Similarly, (TXA2), generated via the pathway following activation, binds to the TP receptor, inducing further platelet shape change, , and reinforcement of aggregation through coordinated signaling with ADP receptors and . The aggregation begins with the formation of an initial of platelets at the injury site, which rapidly expands into a multilayer structure comprising dozens to hundreds of platelets, driven by these amplifying signals. This growth becomes irreversible within 30-60 seconds, as stable fibrinogen bridges form and outside-in signaling through GPIIb/IIIa strengthens the . Early aggregation involves reversible weak bonds, primarily through transient interactions, but transitions to irreversible strong bonds following intracellular calcium influx, which mobilizes from stores and extracellular sources to enhance affinity and cytoskeletal reorganization.

Stabilization and maturation

Integration with secondary hemostasis

The platelet plug integrates with secondary hemostasis by providing a procoagulant surface that facilitates the assembly of factor complexes. Upon activation, platelets expose () on their outer , a negatively charged that serves as a catalytic surface for the binding of factors Va and Xa, thereby accelerating the formation of tenase and prothrombinase complexes essential for generation. This exposure, which increases from approximately 2% to 12% on the platelet surface, enhances prothrombin conversion to by up to 150,000-fold compared to soluble factor Xa alone, enabling localized amplification of the cascade. Thrombin generated through this platelet-supported process creates a positive feedback loop that further reinforces the plug. Thrombin activates additional platelets, promoting their aggregation and PS exposure, while simultaneously cleaving fibrinogen to form fibrin monomers that polymerize into a fibrous network surrounding and stabilizing the platelet plug. This fibrin deposition not only entraps platelets but also captures thrombin, concentrating its activity at the injury site to sustain hemostasis. Factor XIII activation by thrombin contributes to the biochemical integration by cross-linking fibrin and platelet-derived proteins. Activated factor XIIIa (FXIIIa) introduces covalent ε-(γ-glutamyl)lysine bonds between fibrin α- and γ-chains, as well as between fibrin and platelet surface proteins like actin and α2-antiplasmin, enhancing clot mechanical stability and resistance to fibrinolysis. Platelet-derived FXIIIa, exposed on the activated platelet surface, contributes to stability through cross-linking and promotes platelet-fibrin adhesion, ensuring a robust hybrid structure. Temporally, the platelet plug forms rapidly during primary within seconds to a minute following vascular injury, providing an initial seal, while secondary overlays the fibrin clot within 3-5 minutes to achieve durable stability. This sequential integration allows the transient platelet aggregate to recruit coagulation factors efficiently without premature fibrin formation.

Plug consolidation and retraction

Following the initial aggregation of platelets and reinforcement by strands from secondary , the platelet plug undergoes consolidation, a process that enhances its structural integrity through molecular interactions between platelets and the network. Platelet , particularly the αIIbβ3 receptor, bind directly to , enabling platelets to exert contractile forces that compact the plug and expel excess fluid, thereby increasing its density and stability. Additionally, proteins secreted from platelet α-granules, including and thrombospondin, are incorporated into the matrix, further reinforcing the scaffold and promoting cross-linking that resists mechanical stress. The key mechanism driving plug retraction is the actin-myosin contractile apparatus within activated platelets, where non-muscle myosin IIA interacts with filaments to generate pulling forces on bound strands. This contraction is regulated by (MLCK), which phosphorylates the regulatory myosin light chain in a calcium-dependent manner, activating the motor activity necessary for filament sliding and force transmission. As a result, the plug shrinks substantially, approximately 50-60% in volume, compacting the structure and squeezing out to form a firmer hemostatic seal. Retraction plays a critical role in by drawing the edges of the injured vessel closer together, which minimizes gaps, reduces permeability to blood components, and facilitates repair. This mechanical approximation supports subsequent cellular and at the site. The process occurs primarily within the first 20-60 minutes after plug formation, after which the consolidated plug maintains its integrity to sustain .

Physiological and pathological roles

Hemostatic benefits

The platelet plug serves as the primary mechanism for rapid , effectively sealing breaches in the vascular to prevent loss, particularly in the microvasculature where small vessel diameters limit the margin for hemorrhage. Upon vascular , circulating platelets adhere to exposed subendothelial matrix components like and , rapidly recruiting additional platelets to form a dynamic aggregate that plugs the defect within seconds to minutes. This process is especially crucial in capillaries and postcapillary venules, where even minor disruptions can lead to significant leakage, and platelets maintain vascular integrity by physically occluding these sites without requiring full activation in low-shear environments. Beyond immediate sealing, the platelet plug contributes to tissue repair through the release of bioactive molecules from platelet granules, including transforming factor-β (TGF-β) from α-granules, which modulates and promotes . TGF-β inhibits excessive immune cell infiltration while stimulating , synthesis, and , thereby facilitating resolution and reducing formation at the injury site. This action ensures that the hemostatic response transitions smoothly into regenerative processes, enhancing overall vascular stability. The hemostatic efficacy of the platelet plug is maintained by precise regulatory mechanisms that prevent overextension, primarily through endothelial-derived inhibitors such as (NO) and (PGI₂). These molecules, produced by intact adjacent to the injury, elevate cyclic GMP and AMP levels in platelets, respectively, inhibiting , , and aggregation to confine the plug to the damaged area and avoid of healthy vessels. Additionally, the plug's formation scales adaptively to injury severity: single platelets can seal pinhole leaks via receptor-mediated spreading on exposed , whereas larger cuts induce amplified aggregation for a robust barrier. In the long term, sustained release of growth factors like TGF-β supports endothelial cell regeneration, promoting re-endothelialization and restoration of barrier function to prevent recurrent bleeding.

Thrombotic and contrary effects

While platelet plugs are essential for at sites of vascular , their formation in intact vessels can lead to pathological . In arterial circulation, inappropriate platelet adhesion and aggregation on disrupted atherosclerotic plaques or eroded promote occlusive thrombi, resulting in conditions such as . In venous systems, platelet plugs contribute to primarily through stasis-induced accumulation, where reduced blood flow allows platelets to interact with the and initiate clot formation, often manifesting as deep vein thrombosis. Several mechanisms underlie these contrary effects. High in stenotic arteries (>5000 s⁻¹) facilitates initial platelet tethering via and glycoprotein Ib-IX-V but often results in unstable plugs characterized by sparse platelet-platelet contacts and porous networks, increasing the risk of fragmentation and . Additionally, inflammation in triggers inappropriate platelet activation; cytokines and exposed subendothelial stimulate platelet receptors like GPVI and αIIbβ3, amplifying aggregation and growth even without overt injury. Representative examples illustrate the clinical impact. In deep vein thrombosis, immobilized platelet-rich plugs form under low-flow conditions, propagating along venous valves and potentially leading to if dislodged. Similarly, embolized platelet fragments from unstable arterial thrombi can occlude cerebral vessels, causing ischemic , with platelet-rich emboli comprising up to 55% of large vessel occlusions linked to atherosclerotic sources. To maintain balance and prevent excessive spread, pathways regulate platelet plug dynamics. For instance, ectodomain shedding of Ibα by ADAM17 metalloprotease reduces surface receptor availability, limiting further platelet and propagation under inflammatory or conditions. This mechanism helps confine plugs to physiological roles while mitigating pathological extension.

Clinical significance

Associated disorders

Disorders associated with impaired platelet plug formation primarily involve hypo-function of platelets, leading to inadequate hemostasis and bleeding tendencies. Thrombocytopenia, characterized by a low platelet count (typically below 150 × 10^9/L), disrupts primary hemostasis by reducing the number of platelets available for adhesion and aggregation at injury sites, resulting in symptoms such as petechiae, easy bruising, and mucosal bleeding, particularly when counts fall below 20 × 10^9/L. Glanzmann thrombasthenia, an autosomal recessive disorder caused by mutations in genes encoding the platelet integrin GPIIb/IIIa (αIIbβ3), impairs fibrinogen-mediated platelet aggregation, leading to failure in stable plug formation and clinical manifestations including mucocutaneous bleeding, epistaxis, and gingival hemorrhage from infancy. This rare condition affects platelet function despite normal platelet counts. Bernard-Soulier syndrome, another inherited disorder, arises from defects in the GPIb-IX-V complex, which mediates platelet adhesion to (vWF) at sites of vascular injury, causing adhesion failure and prolonged . Patients exhibit with giant platelets and symptoms such as severe epistaxis, menorrhagia, and easy bruising, with an estimated of 1 in 1 million individuals. (vWD), the most common inherited disorder with a of approximately 1% in the general population, involves quantitative or qualitative deficiencies in vWF, which is essential for platelet adhesion to subendothelial , thereby weakening initial plug formation. Clinical features include mucosal , prolonged epistaxis, and menorrhagia, often exacerbated by low vWF levels that prolong . In contrast, hyperactive platelet plug formation contributes to thrombotic disorders. , a featuring persistently elevated platelet counts (often >450 × 10^9/L), promotes excessive platelet aggregation and formation, increasing risks of arterial and . Symptoms may include headache, dizziness, (burning pain in extremities), and ischemic events such as or . (HIT), an immune-mediated condition triggered by autoantibodies against platelet factor 4-heparin complexes, paradoxically activates platelets despite causing , leading to hypercoagulability and widespread . This results in symptoms like skin necrosis at injection sites, limb ischemia, and , with platelet contributing to clot formation in up to 50% of cases. These hypo- and hyper-function states highlight the critical balance required for effective platelet plug dynamics, where defects prolong and excesses predispose to ischemic complications.

Diagnostics

The assessment of platelet plug function primarily involves laboratory tests that evaluate platelet adhesion, aggregation, and overall hemostatic competence. The bleeding time test, once used to measure primary hemostasis by incising the skin and timing cessation of bleeding, is now considered outdated and no longer recommended due to its poor reproducibility, insensitivity, and lack of specificity for platelet disorders. In its place, the Platelet Function Analyzer (PFA-100) provides a more standardized simulation of high-shear conditions, where citrated whole blood flows through a membrane coated with collagen and epinephrine or ADP, measuring closure time to assess adhesion and aggregation defects relevant to primary hemostasis. Light transmission aggregometry (LTA) remains the gold standard for evaluating platelet aggregation responses to agonists such as ADP and collagen, quantifying the extent of platelet clumping in platelet-rich plasma to identify functional impairments in plug formation. Flow cytometry offers a sensitive method to detect abnormalities in platelet receptor expression, such as glycoprotein Ib or IIb/IIIa, by labeling surface markers and analyzing activation states, aiding in the diagnosis of qualitative platelet defects.

Treatments

Clinical interventions to modulate platelet plug formation focus on antiplatelet therapies that inhibit key pathways in primary hemostasis, primarily to prevent excessive while preserving balance. Aspirin irreversibly inhibits cyclooxygenase-1 (COX-1) in platelets, blocking production and thereby reducing platelet activation and aggregation essential for plug stability. Clopidogrel, a converted to an active metabolite, selectively antagonizes the ADP receptor on platelets, preventing amplification of the aggregation signal and diminishing plug consolidation. Glycoprotein IIb/IIIa antagonists, such as , bind to the fibrinogen receptor on activated platelets, inhibiting the final common pathway of aggregation and effectively disrupting irreversible plug formation during acute settings like percutaneous coronary interventions. For conditions impairing platelet adhesion, such as (vWD), (DDAVP) is administered to stimulate endothelial release of (vWF), enhancing platelet tethering to the subendothelium and improving primary .

Emerging Therapies and Monitoring

In cases of thrombocytopenia leading to inadequate platelet plug formation, platelet transfusions provide immediate support by replenishing functional platelets to restore hemostatic capacity, particularly in patients or those undergoing . approaches, including lentiviral vectors targeting hematopoietic stem cells, have advanced to phase 2 clinical trials for Glanzmann thrombasthenia involving GPIIb/IIIa deficiencies, showing promising results in improving platelet function and as of 2025. For (HIT), universal heparin reversal agents (UHRAs) are under preclinical investigation; these agents disrupt immune complexes to suppress platelet activation and thrombosis without increasing risk. Emerging diagnostics include microfluidic systems and the Total Thrombus-Formation Analysis System (T-TAS), which simulate conditions to assess platelet plug formation in real-time using minimal volumes. To responses to antiplatelet therapies, the VerifyNow serves as a point-of-care that measures platelet reactivity in using light transmission through fibrinogen-coated beads, providing rapid quantification of inhibition levels (e.g., in P2Y12 reaction units) to guide dose adjustments and assess non-response.

Current research

Experimental models

In vitro experimental models have been instrumental in elucidating the mechanisms of platelet plug formation by simulating key physiological conditions such as and agonist-induced . Parallel plate flow chambers replicate arterial or venous shear rates, allowing researchers to observe platelet to collagen-coated surfaces and subsequent growth under flow, which has revealed the critical role of Ib-IX-V in initial and GPIbα-von Willebrand factor interactions in stabilizing early plugs. Microfluidic devices offer higher resolution imaging of plug dynamics, enabling of platelet recruitment, formation, and occlusion in microchannels coated with thrombogenic substrates, thus providing insights into spatial heterogeneity and shear-dependent plug stability. Aggregometers, particularly light transmission variants, measure platelet aggregation in stirred by quantifying changes in light transmittance as platelets clump in response to agonists like or , serving as a foundational to assess plug consolidation kinetics and receptor signaling pathways. Animal models complement in vitro approaches by enabling in vivo observation of platelet plug formation in intact vascular systems. Intravital microscopy of the mouse cremaster muscle arterioles allows non-invasive, real-time imaging of platelet accumulation and plug development following vascular injury, highlighting the temporal sequence of adhesion, activation, and reinforcement under physiological blood flow. Laser-induced injury models in mice generate localized endothelial damage via focused laser ablation, facilitating the study of rapid plug initiation and growth while minimizing systemic effects, and have demonstrated the essential contributions of P-selectin and PSGL-1 to platelet-leukocyte interactions within forming plugs. Zebrafish larvae serve as a translucent model for genetic manipulation, where morpholino knockdown or CRISPR/Cas9 editing of platelet-related genes permits high-throughput screening of hemostatic defects, such as impaired thrombocyte aggregation, offering a cost-effective platform to identify novel regulators of plug assembly. Studies of hemostasis in mice underscore both similarities and divergences from human physiology that inform model interpretation. Knockout of the GPIIb/IIIa integrin (β3 integrin-deficient mice) results in severe defects in platelet aggregation and prolonged tail bleeding times, mimicking Glanzmann thrombasthenia and confirming the integrin's indispensable role in irreversible plug stabilization through fibrinogen bridging. Notably, mice lack the FcγRIIA receptor on platelets, which in humans mediates immune complex-induced activation and aggregation, necessitating transgenic humanization for studies of antibody-driven thrombotic plugs. These genetic models have established that while murine platelets effectively form hemostatic plugs in response to injury, subtle differences in receptor expression can alter plug robustness compared to human systems. Despite their utility, experimental models face inherent limitations that temper their translational value. Species variations, such as differences in multimer composition—where murine VWF exhibits less pronounced ultra-large multimers under shear—can lead to discrepancies in platelet capture efficiency and plug initiation rates relative to s. Ethical constraints further restrict the use of larger animals like pigs or nonhuman , which more closely mimic coagulation cascades but require extensive regulatory oversight, higher costs, and justification for invasive procedures, thereby favoring smaller models despite their physiological gaps.

Recent advances and future directions

Recent computational modeling has advanced the understanding of platelet plug formation under hemodynamic conditions. A 2025 study introduced a two-dimensional () model that simulates shear-induced platelet , activation, and aggregation by integrating chemical triggers like () release with mechanical factors such as shear gradients. This approach predicts plug growth and stability in vascular environments, revealing how in high-shear flows enhances aggregation beyond traditional models. Emerging research has uncovered novel roles for red blood cells (RBCs) in modulating platelet plugs. In 2025 findings, RBCs were shown to drive clot shrinkage independently of platelets through aggregation within networks, primarily via osmotic depletion forces that compact the structure by over 20% in platelet-depleted conditions. This mechanism, previously overlooked, suggests RBCs actively contribute to plug stabilization post-formation, potentially influencing hemostatic efficiency . Therapeutic targeting of platelet signaling has progressed with selective inhibitors. The PI3Kβ blocker MIPS-9922 effectively suppresses ADP-induced platelet aggregation and integrin αIIbβ3 activation, offering a pathway-specific approach to mitigate thrombotic risks without broad bleeding complications. Complementing this, structure-based drug design has yielded hemostatic modulators, such as engineered peptides that fine-tune plug consolidation by altering fibrin-platelet interactions. Beyond , platelets show promise in neurological repair. Platelet-derived growth factors, including , promote blood-brain barrier () maturation and in 2024 studies, enhancing endothelial integrity and neurovascular repair in ischemic models. In care, lyophilized platelet products like Thrombosomes have advanced with extended shelf lives up to three years, enabling rapid reconstitution for hemorrhage control while reducing contamination risks. Looking ahead, multi-omics integration promises deeper insights into platelet signaling. Combining , transcriptomics, and will map dynamic pathways in plug formation, identifying novel targets for personalized antithrombotics. Platelet extracellular vesicles, derived from platelet exosomes, emerge as therapeutic vehicles; these nanoscale carriers show potential for delivering therapies targeting mitochondrial dysfunction in cardiovascular and neurodegenerative diseases.

References

  1. [1]
    Physiology, Hemostasis - StatPearls - NCBI Bookshelf - NIH
    The mechanism of hemostasis can divide into four stages. 1) Constriction of the blood vessel. 2) Formation of a temporary “platelet plug." 3) Activation of the ...Introduction · Issues of Concern · Function · Mechanism
  2. [2]
    Hemostasis: What It Is & Stages - Cleveland Clinic
    Platelets circulating in your blood stick to the damaged tissue to form a temporary plug (primary hemostasis). That clot works like a bottle cork, keeping ...
  3. [3]
    Mechanism Action of Platelets and Crucial Blood Coagulation ...
    Oct 1, 2017 · Released secretary granules will recruit additional platelets to form the platelet plug, which is referred to as primary hemostasis. Following ...
  4. [4]
    Current Understanding of Hemostasis - PMC - PubMed Central - NIH
    There are two main components of hemostasis. Primary hemostasis refers to platelet aggregation and platelet plug formation. Platelets are activated in a ...
  5. [5]
    Physiological Haemostasis - Mechanisms of Vascular Disease - NCBI
    ... platelets, adhesive proteins located in the subendothelial matrix (including collagen and von Willebrand factor), and circulating fibrinogen. The end ...
  6. [6]
    The role of von Willebrand factor in thrombus formation - PMC - NIH
    VWF mediates adhesion and aggregation of platelets at sites of vascular injury, processes that are critical for both haemostasis and thrombosis.
  7. [7]
    Platelets and Their Role in Hemostasis and Thrombosis ... - MDPI
    In this review, we aim to provide a comprehensive view of the complex role that platelets play in hemostasis and thrombosis, and to discuss the major clinical ...
  8. [8]
    Histology, Platelets - StatPearls - NCBI Bookshelf
    Platelets are the smallest blood cells, typically around 2 μm in diameter and anucleated, with an average lifespan of 7 to 10 days in humans.Bookshelf · Structure · Clinical Significance
  9. [9]
    Platelet Shape Changes and Cytoskeleton Dynamics as Novel ...
    PROCESS OF THE SHAPE CHANGES AND CYTOSKELETON DYNAMICS IN PLATELETS. Resting platelets are small discoid shapes around 1.5–3 μm in size. The cytoskeleton of ...
  10. [10]
    New Fundamentals in Hemostasis | Physiological Reviews
    Jan 1, 2013 · Hemostasis encompasses the tightly regulated processes of blood clotting, platelet activation, and vascular repair.
  11. [11]
    Hemostasis without clot formation: how platelets guard the ...
    Oct 26, 2023 · Platelets are key vascular effectors in hemostasis, with activation signals leading to fast recruitment, aggregation, and clot formation.
  12. [12]
    Hemostasis – Anatomy & Physiology - UH Pressbooks
    Hemostasis involves three basic steps: vascular spasm, the formation of a platelet plug, and coagulation, in which clotting factors promote the formation of a ...Missing: sequence timings
  13. [13]
    6
    Jan 5, 2009 · Part 2 of Hemostasis) Platelet plug formation (platelet response ... seconds in severe trauma and within 1-4 minutes with minor trauma.
  14. [14]
    Of vascular defense, hemostasis, cancer, and platelet biology
    We explore the evolutionary connections that place platelets at the center of hemostasis, immunity, and adaptive phylogeny.
  15. [15]
    The Discovery of Blood Cells - Annals of Clinical & Laboratory Science
    In 1842, he discovered a third element, the platelets, in blood [5]. Despite indifference and often even open hostility by his colleagues, Donné gave ...
  16. [16]
    Max Schultze (1865), G. Bizzozero (1882) and the ... - PubMed
    Max Schultze published the first accurate and convincing description of platelets as part of a study devoted mainly to the white blood cells in 1865.
  17. [17]
    Hayem in 1877 provided a firm histologic basis for platelets
    of Schultze, who in 1865 noted abundant, irregular masses of colorless globules in normal blood that were almost certainly platelets. The impact of Osler's ...
  18. [18]
    Are Platelets Cells? And if Yes, are They Immune Cells? - PMC - NIH
    However, at this time, platelets were often considered to be of leukocyte origin and to be degenerate or degraded. Some observers favored an erythrocyte origin ...
  19. [19]
    Giulio Bizzozero and the discovery of platelets - PubMed
    Platelets were discovered by the Italian pathologist Giulio Bizzozero in 1882. He observed them microscopically in the circulating blood of living animals.Missing: Donné 1842 Hayem 1878 Schultze 1865
  20. [20]
    The Mechanism of Hemostasis - PubMed
    The Mechanism of Hemostasis. Ann Surg. 1947 Mar;125(3):292-310. doi: 10.1097/00000658-194703000-00003. Author. L M Tocantins.Missing: primary secondary 1930s
  21. [21]
    Historical overview of the role of platelets in hemostasis ... - PubMed
    ... (aggregation). The discovery of the role of ADP as the principle of platelet aggregation stimuli was rapidly followed by other important discoveries such as ...
  22. [22]
    Aggregation of blood platelets by adenosine diphosphate and its ...
    Aggregation of blood platelets by adenosine diphosphate and its reversal. Nature. 1962 Jun 9:194:927-9. doi: 10.1038/194927b0. Author. G V BORN. PMID: 13871375 ...Missing: aggregometer | Show results with:aggregometer
  23. [23]
    Human platelet membrane receptor for bovine von Willebrand factor ...
    Its relationship with platelet adhesion to subendothelium and with the factor VIII von Willebrand protein. J Lab Clin Med. 1976 Apr;87(4):586–596. [PubMed] ...
  24. [24]
    The Organizing Principle of Platelet Glycoprotein Ib-IX-V Complex
    Apr 1, 2014 · The glycoprotein (GP)Ib-IX-V complex is expressed exclusively on platelets and mega karyocytes. It is the second most abundant receptor complex ...
  25. [25]
    The GPIIb/IIIa (integrin αIIbβ3) odyssey: a technology-driven saga of ...
    Oct 15, 2008 · It was soon discovered that platelets contain 4 other integrins. In contrast to αIIbβ3, however, these receptors were expressed at low levels, ...Abstract · Introduction · Glanzmann thrombasthenia... · Conclusions
  26. [26]
    Integrin Signaling: The Platelet Paradigm | Blood - ASH Publications
    Apr 15, 1998 · The purpose of this review is to describe the platelet paradigm of integrin signaling and to emphasize the advances and gaps in our understanding of this ...
  27. [27]
    New Concepts and Mechanisms of Platelet Activation Signaling - PMC
    In this review, we will provide an overview and update to the concepts and mechanisms of platelet activation signaling. With the recent advances and ...
  28. [28]
    Platelet Activation: The Mechanisms and Potential Biomarkers - PMC
    May 26, 2016 · In this review, we discuss recent advances in the pathways of platelet activation and potential application of platelet activation biomarkers ...
  29. [29]
    Mechanisms of Platelet Activation and Integrin αIIβ3 - PubMed Central
    May 24, 2012 · This review will focus on the recent progress of knowledge about platelet activation mechanism. Mediators of Platelet Activation. Adenosine ...
  30. [30]
    Signaling during platelet adhesion and activation - PMC
    VWF forms a so-called “catch-bond” or “flex-bond” with the ligand binding domain of GPIb-IX,, allowing transient platelet adhesion under high shear stress. VWF/ ...
  31. [31]
    Platelet adhesion: a game of catch and release - PMC
    This behavior of individual GPIbα-vWF bonds under shear correlated with adhesion of platelets or beads to vWF under flow, where increasing shear forces ...
  32. [32]
    Signaling mechanisms of the platelet glycoprotein Ib-IX complex
    The GPIb-IX-VWF interaction is particularly crucial in platelet adhesion and thrombus formation under high shear blood flow (such as in arteries and arterioles) ...
  33. [33]
    [PDF] Shear-Dependent Platelet Aggregation: Mechanisms and ... - Monash
    Sep 20, 2019 · Other transmembrane platelet receptors mediating platelet adhesion include GPVI (8) and integrin α2β1 (9), that bind directly to collagen (10).
  34. [34]
    Imaging Platelet Processes and Function—Current and Emerging ...
    Arterial thrombosis usually is triggered by rupture of a collagen- and tissue factor-rich stenotic plaque at relatively high (1,000–5,000 s−1) wall shear rates, ...
  35. [35]
    Platelets roll on stimulated endothelium in vivo: an interaction ... - NIH
    Our results demonstrate that (i) platelets roll on endothelium in vivo, (ii) this interaction requires endothelial but not platelet P-selectin, and (iii) ...
  36. [36]
    Proteolytic Processing of Von Willebrand Factor by Adamts13 and ...
    VWF is an abundant plasma glycoprotein synthesized in all vascular endothelial cells and megakaryocytes as a precursor containing a signal peptide and large ...
  37. [37]
    Inflammation, von Willebrand factor, and ADAMTS13 - PMC - NIH
    Jul 12, 2018 · VWF is synthesized only in megakaryocytes and endothelial cells. Although a portion of VWF is constitutively secreted, a major portion is ...
  38. [38]
    ADP and platelets: the end of the beginning - PMC - NIH
    Platelet responses to ADP require the coordinate activation of two G protein–coupled receptors, P2Y1 and P2Y12, whose actions are described in the text.
  39. [39]
    Adenosine diphosphate (ADP)–induced thromboxane A2generation ...
    Adenosine diphosphate (ADP) is a platelet agonist that causes platelet shape change and aggregation as well as generation of thromboxane A 2.
  40. [40]
    Adenosine Diphosphate–Induced Platelet Aggregation Is ...
    In healthy subjects, ADP-induced platelet aggregation is associated with a haplotype of the P2Y 12 receptor gene.
  41. [41]
    Platelet Aggregometry Testing: Molecular Mechanisms, Techniques ...
    Irreversible platelet response includes the release reaction and secondary irreversible aggregation. Increased free calcium concentration within the platelet ...
  42. [42]
    Exposure of platelet membrane phosphatidylserine regulates blood ...
    Platelet membrane phosphatidylserine (PS) exposure regulates thrombin production, a key molecule in blood coagulation, and acts as a second messenger.
  43. [43]
    Platelets and Thrombin Generation | Arteriosclerosis, Thrombosis ...
    This review examines the evidence that platelets play a major role in localizing and controlling the burst of thrombin generation leading to fibrin clot ...
  44. [44]
    Factor XIII: driving (cross-)links in hemostasis, thrombosis, and disease
    FXIIIa cross-linking (X) of fibrin mechanically stabilizes fibrin fibers and increases red blood cell retention during platelet-mediated clot contraction.
  45. [45]
    Functional factor XIII-A is exposed on the stimulated platelet surface
    Dec 18, 2014 · Factor XIII (FXIII) stabilizes thrombi against fibrinolysis by cross-linking α2-antiplasmin (α2AP) to fibrin. Cellular FXIII (FXIII-A) is ...Fxiii Activity Assay · Results · Platelet Activation Is...
  46. [46]
    Platelet receptors and signaling in the dynamics of thrombus formation
    Binding of αIIbβ3 to its ligands, mainly fibrinogen, supports processes such as clot retraction and platelet aggregation. Stabilization of thrombi is supported ...
  47. [47]
    Mechanisms Underlying Dichotomous Procoagulant COAT Platelet ...
    Feb 25, 2022 · Procoagulant platelets are a subtype of activated platelets that sustains thrombin generation in order to consolidate the clot and stop bleeding.
  48. [48]
    Feeling the Force: Measurements of Platelet Contraction and Their ...
    The interaction of actin and myosin for retraction is regulated by phosphorylation of myosin light chain (MLC) through calcium-dependent activation of MLC ...
  49. [49]
    Clot contraction: compression of erythrocytes into tightly packed ...
    The function of clot contraction is not fully known, but it appears to reinforce hemostasis by forming a seal, promote wound healing by approximating the edges, ...
  50. [50]
    Platelet heterogeneity enhances blood clot volumetric contraction
    During blood clot contraction, microscopic platelets actively pull fibers to shrink the macroscale clot to less than 10% of its initial volume. We discovered ...Missing: edges | Show results with:edges<|control11|><|separator|>
  51. [51]
    Platelets and vascular integrity: how platelets prevent bleeding in ...
    Jan 18, 2018 · Platelets play a central role in primary hemostasis by forming aggregates that plug holes in injured vessels.
  52. [52]
    Platelet secretion: From haemostasis to wound healing and beyond
    Platelet dense granule components, such as ADP and polyphosphates, contribute to haemostasis and coagulation, but also play a role in cancer metastasis.
  53. [53]
    Platelets in Thrombosis and Atherosclerosis: A Double-Edged Sword
    Platelets are known to play a crucial role in atherosclerosis by mediating thrombosis, inflammation, and angiogenesis.Platelets And Interactions · Platelets: Hemostatic And... · Endothelium And Platelet...<|control11|><|separator|>
  54. [54]
    REVIEW Platelets at work in primary hemostasis - ScienceDirect.com
    Nitric oxide (NO) and prostacyclin (PGI2) and PGD2 are strong negative ... platelet plug by forming a mesh of cross-linked fibrin. Platelets and ...
  55. [55]
    Platelets and platelet adhesion molecules: novel mechanisms of ...
    Oct 4, 2016 · Inappropriate platelet adhesion, activation and aggregation promote excessive platelet plug formation. Activated platelets can also provide ...Platelets And Platelet... · Arterial Thrombosis And... · Platelet Adhesion Molecules...<|separator|>
  56. [56]
    von Willebrand factor–mediated platelet adhesion is critical for deep ...
    Our results demonstrate a pathogenetic role for VWF-platelet interaction in flow disturbance-induced venous thrombosis.Vwf Mice Are Protected From... · Vwf-Platelet Gpibα... · Vwf Is Expressed On The...
  57. [57]
    Arterial Thrombus Stability: Does It Matter and Can We Detect It?
    Oct 9, 2017 · In comparison, unstable thrombi, comprising sparse platelet–platelet contacts and less dense fibrin mesh composed of thicker, more porous fibers ...
  58. [58]
    Platelet-Rich Emboli in Cerebral Large Vessel Occlusion Are ...
    May 29, 2019 · High platelet content of emboli is associated with a large artery atherosclerosis etiology of large vessel occlusion.Missing: embolized fragments
  59. [59]
  60. [60]
    Thrombocytopenia - StatPearls - NCBI Bookshelf - NIH
    Patients with platelets greater than 50000/mL, rarely have symptoms. Patients with platelets under 20000/mL most likely have spontaneous bleeding. Ask the ...Missing: formation | Show results with:formation
  61. [61]
    Glanzmann Thrombasthenia - StatPearls - NCBI Bookshelf - NIH
    Glanzmann thrombasthenia is a congenital bleeding disorder caused by a deficiency of the platelet integrin alpha IIb beta3.Bookshelf · Treatment / Management · Enhancing Healthcare Team...
  62. [62]
    Glanzmann's thrombasthenia: pathogenesis, diagnosis, and current ...
    Jul 8, 2015 · The increased amount of thrombin enhances platelet adhesion and promotes platelet aggregation, including those lacking GPIIb/IIIa. This has ...Pathogenesis Of Gt · Role Of Transfusions In The... · Use Of Rfviia In GtMissing: poor | Show results with:poor
  63. [63]
    Bernard-Soulier Syndrome - StatPearls - NCBI Bookshelf
    Jan 11, 2024 · Bernard-Soulier syndrome is a rare inherited blood clotting disorder characterized by unusually large, giant platelet cells, thrombocytopenia, and prolonged ...Bernard-Soulier Syndrome · Pathophysiology · Treatment / Management
  64. [64]
    Patients with Bernard-Soulier syndrome and different severity of the ...
    Bernard-Soulier syndrome is a rare (1:1million), hereditary bleeding disorder caused by defects of the platelet GPIb-IX-V complex.Missing: failure | Show results with:failure
  65. [65]
    Von Willebrand Disease - StatPearls - NCBI Bookshelf - NIH
    Von Willebrand disease is estimated to affect approximately 1% of the unselected population, but clinically significant disease prevalence is estimated to be ...Missing: plug | Show results with:plug
  66. [66]
    [PDF] von Willebrand Disease - NHLBI
    time; RIPA, ristocetin-induced platelet aggregation; VWF, von Willebrand ... *Note: persons who have platelet-type VWD (PLT-VWD) have a defect in their platelet ...<|separator|>
  67. [67]
    Essential Thrombocytosis - StatPearls - NCBI Bookshelf - NIH
    Aug 9, 2025 · Thrombocytosis increases the risks of vascular events such as thrombosis, hemorrhage, and sometimes the conversion to a blast phase of ...
  68. [68]
    Thrombocytosis - Symptoms and causes - Mayo Clinic
    Dec 30, 2022 · Headache. Confusion or changes in speech. Chest pain. Shortness of breath and nausea. Weakness. Burning pain in the hands or feet. Less commonly ...
  69. [69]
    Heparin-Induced Thrombocytopenia - StatPearls - NCBI Bookshelf
    A fall in platelet counts and a hypercoagulable state characterize HIT. Patients who experience HIT may also develop thromboembolic complications that are ...Missing: autoantibodies | Show results with:autoantibodies
  70. [70]
    Heparin Induced Thrombocytopenia: A Focus on Thrombosis - PMC
    Heparin induced thrombocytopenia (HIT) is an immune mediated disorder caused by antibodies that recognize complexes of platelet factor 4 and heparin.
  71. [71]
    Demystifying Platelet Function Testing | ASH Clinical News
    Aug 29, 2023 · The platelet function analyzer (PFA)-100 or PFA-200 evaluates platelet function by measuring the time required for whole blood under high shear ...
  72. [72]
    Bleeding Time | myadlm.org
    Dec 12, 2023 · Bleeding Time: Optimal Testing Recommendations. Nonspecific and insensitive test of platelet function; This test should no longer be used.Missing: outdated | Show results with:outdated
  73. [73]
    Platelet function testing - UpToDate
    Apr 25, 2024 · This topic summarizes available platelet function tests and provides information about how to interpret the results of platelet function testing ...
  74. [74]
    Platelet function tests: a comparative review - PMC - PubMed Central
    In this review, well-tried and innovative platelet function tests and their methodological features and clinical applications are considered.
  75. [75]
    [PDF] Platelet Function Testing (PDF) - Vitalant Health
    It is a point of care test requiring a hematology analyzer and measures the change in platelet count due to aggregation of functional platelets. Flow cytometry, ...
  76. [76]
    Functional Platelet Disorders | Choose the Right Test - ARUP Consult
    Apr 29, 2025 · Initial laboratory testing for a functional platelet disorder includes a CBC with platelet count and a peripheral smear. These tests are useful ...Functional Platelet... · Laboratory Testing · Platelet Flow Cytometry
  77. [77]
    Antiplatelet Medications - StatPearls - NCBI Bookshelf - NIH
    Nov 7, 2022 · Antiplatelet medications divide into oral and parenteral agents, and oral agents subdivide further based on the mechanism of action. Aspirin was ...
  78. [78]
    Antiplatelet medications: Video, Causes, & Meaning - Osmosis
    Antiplatelet medications inhibit the steps of primary hemostasis to prevent the platelet plug from forming. ... Aspirin, as an antiplatelet medication ...
  79. [79]
    The platelet P2Y receptors as targets for new antithrombotic drugs
    The problem of drug resistance is certainly more relevant for thienopyridines. Ticlopidine and clopidogrel are prodrugs, which need to be metabolized by the ...
  80. [80]
    Desmopressin antagonizes the in vitro platelet dysfunction induced ...
    Desmopressin antagonizes the in vitro platelet dysfunction induced by GPIIb/IIIa inhibitors and aspirin ... Platelet Aggregation / drug effects*; Platelet ...
  81. [81]
    The effect of desmopressin on bleeding time and platelet ...
    Jan 21, 2014 · Desmopressin has previously been shown to improve primary haemostasis and is widely used as first-line therapy for individuals with bleeding ...Missing: plug | Show results with:plug
  82. [82]
    Platelet transfusion for patients with platelet dysfunction
    Platelet transfusion therapy is highly effective in managing bleeding in patients with hypoproliferative thrombocytopenia.
  83. [83]
    Treatment of inherited thrombocytopenias - PMC - NIH
    Jun 1, 2022 · Thrombopoietin receptor agonists is an advantageous alternative to platelet transfusions in the preparation for surgery of patients with forms ...
  84. [84]
    Platelet gene therapy improves hemostatic function for integrin ...
    May 23, 2011 · These results indicate that gene therapy could become a practical approach for treating inherited platelet defects.
  85. [85]
    Gene therapy for platelet disorders: studies with Glanzmann's ...
    Dec 21, 2022 · These defects have been shown to lead to disruption of GPIIb and GPIIIa synthesis, receptor assembly, and/or function, thereby preventing GPIIb– ...
  86. [86]
    Monitoring platelet inhibition after clopidogrel with the VerifyNow ...
    VerifyNow-P2Y12 is a reliable, fast and sensitive device suitable for monitoring of platelet inhibition during clopidogrel therapy.
  87. [87]
    How I use laboratory monitoring of antiplatelet therapy | Blood
    This article reviews platelet function, molecular targets of antiplatelet agents, and clinical indications for antiplatelet therapy
  88. [88]
    A 2D computational model of chemically- and mechanically-induced ...
    Jul 7, 2025 · The goal of the present work is to develop a predictive computational model of platelet plug formation that accounts for the role of shear gradients.
  89. [89]
    Vorticity‐Facilitated Platelet Aggregation: a High Expansion‐Ratio ...
    Jun 4, 2025 · This study reveals how vorticity—a swirling flow pattern—enhances platelet aggregation and thrombus formation in high-shear conditions.
  90. [90]
    Red blood cell aggregation within a blood clot causes platelet ...
    Jul 10, 2025 · In summary, this study provides compelling evidence that RBCs play a role in blood clot shrinkage unrelated to platelets. The main mechanism ...
  91. [91]
    New study reveals unexpected role of red blood cells in clot formation
    Aug 6, 2025 · Red blood cells, long thought to be passive bystanders in the formation of blood clots, actually play an active role in helping clots ...
  92. [92]
    Platelets and diseases: signal transduction and advances in ...
    May 16, 2025 · This review explores the structure, signaling pathways, biological functions, and therapeutic potential of platelets, highlighting their roles in both ...
  93. [93]
    Structure-based design of therapeutics to control hemostasis | Blood
    Thrombosomes, a commercial preparation of lyophilized platelets, recently completed phase 2 clinical trials and showed a shelf life of 3 years, which is a ...
  94. [94]
    Platelets: A new therapeutic target for neurological diseases
    Platelets promote angiogenesis in the central nervous system by releasing factors such as PDGF-BB, driving the maturation and repair of the blood-brain barrier ...
  95. [95]
    Ultrasoft platelet-like particles stop bleeding in rodent and porcine ...
    Apr 10, 2024 · Innovations in platelet storage, including cold storage and lyophilization, have helped to address shelf life and contamination concerns ...<|separator|>
  96. [96]
    Multi-omics approaches to study platelet mechanisms - ScienceDirect
    In this review, we will present an overview of established and new multi-omics strategies, which can be used to study platelet biology in health and disease.Missing: directions | Show results with:directions
  97. [97]
    Platelet Extracellular Vesicles as Natural Delivery Vehicles for ...
    Apr 25, 2025 · Platelet extracellular vesicles (PEVs), once called “platelet dust”, have emerged as promising agents for mitigating mitochondrial dysfunction.