Fact-checked by Grok 2 weeks ago

P-selectin

P-selectin, also known as CD62P or granule membrane protein-140 (GMP-140), is a 140 kDa transmembrane glycoprotein belonging to the selectin family of cell adhesion molecules, which mediate calcium-dependent interactions between leukocytes and vascular endothelium during inflammation and hemostasis. It is constitutively stored in the α-granules of platelets and the Weibel-Palade bodies of endothelial cells, enabling rapid surface expression—within minutes—upon activation by agonists such as thrombin, histamine, or inflammatory cytokines like TNF-α and IL-1β. Its structure consists of an N-terminal C-type lectin domain for carbohydrate recognition, followed by an epidermal growth factor (EGF)-like domain, nine complement regulatory protein-like repeats, a transmembrane domain, and a short cytoplasmic tail that facilitates trafficking and signaling. The primary function of P-selectin is to initiate leukocyte recruitment by mediating the and rolling of neutrophils, monocytes, and other leukocytes on activated or platelets through high-affinity binding to (PSGL-1) on leukocytes, which requires post-translational modifications including (sLe^x) glycans and tyrosine sulfation for optimal interaction. This rolling step slows leukocytes from blood flow, allowing subsequent firm adhesion via and into tissues, a process essential for acute inflammatory responses, , and immune surveillance. In , P-selectin promotes platelet-leukocyte aggregates, enhances formation by inducing expression on monocytes, and supports vascular integrity, but its dysregulation contributes to pathological conditions such as , , and vaso-occlusion. Soluble forms of P-selectin, shed from the cell surface by proteolytic cleavage, circulate in plasma at baseline levels of approximately 100 ng/mL and elevate in inflammatory states like (ARDS) up to 1 μg/mL, serving as a for endothelial activation and cardiovascular risk. Genetic polymorphisms, such as the N562D variant, have been associated with increased risk in conditions like (APS), highlighting P-selectin's dual role in protective and deleterious processes. Therapeutic strategies targeting P-selectin, including monoclonal antibodies like , have shown promise in reducing inflammation-related complications in by blocking PSGL-1 interactions.

Genetics and Regulation

Gene Organization

The SELP gene, which encodes P-selectin, is located on the long arm of human chromosome 1 at the cytogenetic band 1q24.2. It spans approximately 41 kb of genomic DNA and consists of 17 exons interrupted by 16 introns, with the exons ranging in size from about 50 to over 1,000 base pairs. The SELP gene was first identified and cloned in 1989 through sequencing of platelet-derived cDNA, initially recognizing the protein as GMP-140, a granule membrane protein involved in cellular adhesion. SELP exhibits strong evolutionary across mammalian , reflecting its role in vascular and immune processes; for instance, the ortholog Selp, located on , shares high sequence similarity with SELP and has been instrumental in generating models to study functions. Several polymorphisms in the SELP influence its expression and , notably the Thr715Pro variant (rs6131), which alters the protein's terminal and reduces surface expression on cells, thereby impairing leukocyte adhesion to and platelets.

Expression and Regulation

P-selectin exhibits constitutive low-level expression in endothelial cells and megakaryocytes, where it is primarily stored in specialized secretory granules rather than being constitutively displayed on the cell surface. In endothelial cells, P-selectin is packaged into Weibel-Palade bodies, while in platelets derived from megakaryocytes, it resides in α-granules. This intracellular storage allows for rapid mobilization to the plasma membrane upon cellular activation, occurring within seconds to minutes without requiring new protein synthesis. Stimuli such as , , and (ROS) trigger this translocation by promoting of the granules, thereby enabling swift endothelial and platelet responses to vascular injury or . At the transcriptional level, P-selectin expression undergoes dynamic upregulation in response to inflammatory signals. Cytokines like tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) induce increased P-selectin mRNA levels in endothelial cells within hours, leading to de novo synthesis and sustained surface expression. This process is mediated by the pathway, which binds to promoter elements in the SELP gene to drive transcription. For longer-term regulation, interleukin-4 (IL-4) promotes prolonged P-selectin expression through activation of the transcription factor STAT6, enhancing endothelial adhesiveness in chronic inflammatory contexts. Post-translational mechanisms further fine-tune P-selectin surface levels. modifies P-selectin, influencing its trafficking, stability, and retention on the cell surface in endothelial cells and platelets. Downregulation occurs primarily through clathrin-mediated , which internalizes surface P-selectin and directs it to late endosomes and lysosomes for degradation, thereby limiting prolonged leukocyte interactions. Additionally, ectodomain shedding by the metalloprotease ADAM17 (also known as TACE) cleaves the extracellular portion of P-selectin from activated platelets and endothelial cells, releasing soluble forms into the plasma and reducing membrane-bound expression. These processes collectively ensure tight spatiotemporal control of P-selectin availability during inflammatory and hemostatic events.

Structure and Ligands

Protein Structure

P-selectin is a type-1 transmembrane composed of 830 in humans, with a calculated of 90,820 that increases to approximately 140 due to extensive post-translational . This heavy , including both N-linked and O-linked modifications at multiple sites in the extracellular region, is essential for the protein's structural stability and functional conformation in ligand interactions. The extracellular domain of P-selectin is organized into distinct modules: an N-terminal C-type lectin-like domain that mediates calcium-dependent binding to carbohydrate ligands, an adjacent (EGF)-like domain that stabilizes the overall architecture, and nine tandem short repeats (SCRs) resembling complement regulatory protein domains, which contribute to the extended rod-like . These SCRs, each approximately 60 long, form beta-sheet-rich folds that space the N-terminal domains from the membrane. Anchoring P-selectin to the is a single transmembrane spanning residues 772-794, followed by a short cytoplasmic tail of 36 that includes a C-terminal dileucine (Leu-Leu at positions 798-799) crucial for intracellular trafficking and endocytic retrieval. This facilitates rapid from the plasma to intracellular storage compartments in endothelial cells and platelets. Structural insights into P-selectin derive from crystallographic studies, such as the 3.40 Å structure of its and EGF domains (PDB: 1G1R), which reveals a bent conformation with the lectin domain coordinating Ca²⁺ via a conserved pocket involving , Asn, and Glu residues for ligand engagement. This exhibits high to the corresponding domains in and , underscoring the conserved mechanism of selectin-mediated adhesion across the family.

Binding Ligands

P-selectin primarily binds to (PSGL-1), a mucin-like glycoprotein encoded by the SELPLG gene and expressed on the surface of leukocytes. High-affinity binding to PSGL-1 requires specific post-translational modifications on its N-terminal region, including sulfation of tyrosine residues, as well as sialylation, fucosylation, and core-2 branching of O-glycans bearing the (sLe<sup>X</sup>) motif. These modifications enable PSGL-1 to interact with the calcium-dependent domain of P-selectin, resulting in a (K<sub>d</sub>) of approximately 320 nM for the soluble monomeric . In addition to PSGL-1, P-selectin recognizes secondary ligands such as proteoglycans on the endothelial surface, which facilitate interactions independent of glycoprotein counter-receptors. Other ligands include the glycosylphosphatidylinositol-anchored protein on certain leukocytes and , a sulfated fucose-containing that mimics sLe<sup>X</sup>-like structures. The binding mechanism is calcium-dependent, with the lectin domain coordinating Ca<sup>2+</sup> ions to form a shallow binding pocket that accommodates the and residues of sLe<sup>X</sup>. Structural studies, including NMR and crystallographic analyses of the P-selectin domain complexed with sLe<sup>X</sup>, reveal that the ligand's ring stacks against a conserved aromatic residue (Trp140) in the binding site, while additional contacts from the and groups on PSGL-1 enhance specificity and beyond simple sLe<sup>X</sup> recognition. Under physiological flow conditions, P-selectin-PSGL-1 interactions are enhanced, promoting leukocyte rolling on endothelial surfaces by increasing lifetime and frequency at higher shear rates. Binding assays, such as and parallel-plate flow chamber experiments, confirm that monomeric P-selectin dissociates rapidly from PSGL-1 (k<sub>off</sub> ≈ 1.4 s<sup>-1</sup>), but multivalent on surfaces stabilizes . Species-specific differences influence binding efficiency; for instance, P-selectin interacts with mouse PSGL-1 less effectively due to variations in the sulfotyrosine and motifs, limiting cross-species in experimental models.

Physiological Functions

Role in

P-selectin, rapidly mobilized to the surface of activated endothelial cells and platelets upon stimulation by inflammatory mediators such as or , mediates the initial tethering and rolling of circulating leukocytes along the vascular under hydrodynamic shear forces in postcapillary venules. This transient, low-affinity interaction slows leukocytes from free-flowing velocities to a rolling motion at speeds of approximately 10-100 μm/s, enabling closer contact with activating like IL-8 or MCP-1 presented on the , which in turn trigger inside-out signaling for high-affinity activation (e.g., LFA-1 binding to ) and firm . Through this mechanism, P-selectin facilitates the recruitment of , , and to sites of , such as those arising from tissue injury or , where it supports the early influx of immune cells essential for pathogen clearance and . In acute inflammatory models, P-selectin expression peaks within minutes, promoting neutrophil predominance in the initial hours, while also contributing to and over longer periods. Its primary ligand on leukocytes is (PSGL-1), which enables these adhesive interactions. P-selectin exhibits synergy with to enhance leukocyte capture efficiency, particularly in acute responses; while P-selectin handles immediate rolling, sustains it during prolonged , with combined deficiency abolishing nearly all rolling . In thioglycollate-induced models, P-selectin deficiency reduces accumulation, and combined P- and deficiency leads to up to 90% reduction at early time points (2-4 hours), underscoring its dominant role in initiating inflammatory cell recruitment. Additionally, P-selectin engagement with PSGL-1 on leukocytes triggers bidirectional signaling pathways, including and MAPK activation, which amplify the inflammatory response by promoting production (e.g., TNF-α, IL-1β) and survival signals in recruited cells, thereby sustaining leukocyte activation and effector functions at inflammatory sites. Recent studies (as of 2025) have also revealed that P-selectin regulates proliferation and differentiation under inflammatory stress, contributing to hematopoiesis maintenance.

Role in Hemostasis and Thrombosis

P-selectin, stored in the α-granules of resting platelets, is rapidly mobilized to the platelet surface upon activation at sites of vascular injury, where it contributes to primary by promoting procoagulant activity and facilitating intercellular bridging. This surface exposure enables P-selectin to bind (PSGL-1) on adjacent platelets or leukocytes, stabilizing initial platelet aggregates through glycoprotein IIb/IIIa-fibrinogen interactions and enhancing overall stability under flow conditions. In high-shear environments, such as arterial circulation, P-selectin also interacts with (VWF) released from endothelial Weibel-Palade bodies, supporting VWF multimer adhesion to the vessel wall and thereby aiding GPIbα-mediated platelet recruitment to the injury site. In the context of thrombosis, particularly venous thrombosis, platelet-derived P-selectin drives the formation of platelet-leukocyte aggregates by engaging PSGL-1 on leukocytes, which recruits tissue factor-bearing microparticles to the growing and accelerates generation. These aggregates amplify through P-selectin-dependent pathways that enhance fibrinogen binding and polymerization, independent of traditional platelet-leukocyte inflammatory signaling. Additionally, P-selectin promotes propagation by stabilizing networks, as evidenced by reduced deposition in models where P-selectin function is disrupted. Studies in P-selectin-deficient mice underscore its essential role in both and pathological . These mice exhibit prolonged tail times, approximately 40% longer than wild-type controls, indicating impaired primary hemostatic plug formation at sites. In models, such as ferric chloride (FeCl3)-induced arterial , P-selectin deficiency or inhibition results in significantly reduced size and stability, with decreased platelet accumulation and formation observed via intravital microscopy. Similarly, in models, P-selectin knockout leads to diminished burden without excessive , highlighting its balanced contribution to clot formation and resolution.

Pathological Roles

Role in Cancer

P-selectin, expressed on the surface of activated endothelial cells in tumor vessels and on platelets, facilitates the adhesion and of circulating tumor cells (CTCs) by binding to sialylated carbohydrate ligands such as (sLeX) and PSGL-1-like structures on cancer cells. This interaction initiates the rolling of tumor cells along the , promoting their transmigration into metastatic sites. In various carcinomas, elevated sLeX expression on tumor cells correlates with enhanced metastatic potential and poorer patient prognosis. Platelets activated in the release P-selectin, which mediates the formation of platelet-tumor aggregates that shield CTCs from hydrodynamic shear forces in circulation and from by natural killer () cells. These complexes enhance the of CTCs and facilitate their and in distant organs, particularly the lungs and liver. By cloaking tumor cells, P-selectin-dependent platelet interactions create an immunosuppressive barrier, allowing CTCs to evade immune surveillance during hematogenous dissemination. Experimental models demonstrate P-selectin's critical role in ; for instance, in P-selectin mice, experimental of B16 cells is significantly reduced compared to wild-type controls, with fewer metastatic foci observed in the s and liver. Reinfusion of wild-type platelets into mice restores metastatic burden, confirming that P-selectin-mediated platelet drives this process. P-selectin contributes to tumor angiogenesis by promoting platelet and leukocyte infiltration into the , where these cells release (VEGF) to stimulate new vessel formation. In mouse models of and , P-selectin deficiency leads to lower intratumoral VEGF levels and reduced microvascular density. Aberrant in cancers, including expression of the sialyl-Tn , further enhances P-selectin availability on tumor cells, linking these modifications to pro-angiogenic signaling. Recent post-2020 research highlights P-selectin's involvement in progression, where it drives activation toward a pro-tumorigenic , enhancing tumor and invasion. In co-culture models and patient-derived xenografts, P-selectin inhibition reduces -mediated invasion and tumor growth by 67% in volume, underscoring its role in reshaping the microenvironment.

Role in Cardiovascular and Other Diseases

P-selectin plays a critical role in by facilitating the recruitment of monocytes to endothelial surfaces within atherosclerotic plaques, thereby promoting lesion growth and plaque instability. Expressed on activated endothelial cells and platelets, P-selectin mediates the initial tethering and rolling of monocytes, enabling their infiltration into the vessel wall and subsequent differentiation into foam cells that exacerbate inflammation and lipid accumulation. Elevated levels of soluble P-selectin (sP-selectin), released from activated cells, serve as a for subclinical and clinical , with higher concentrations independently associated with increased risk of (CAD) in population studies. These sP-selectin levels also correlate with , reflecting ongoing vascular inflammation and providing diagnostic utility in assessing cardiovascular risk. In , P-selectin contributes to vaso-occlusive crises by enhancing leukocyte-endothelial adhesion, which promotes the heterotypic aggregation of sickled red cells, leukocytes, and platelets, leading to microvascular . Upregulation of endothelial P-selectin under hypoxic conditions facilitates this adhesive cascade, amplifying and ischemia during painful crises. Genetic variants in the SELP gene encoding P-selectin have been linked to altered expression levels, increasing susceptibility to frequent vaso-occlusive events in affected individuals. P-selectin amplifies extracellular trap () formation in (DVT) and ischemia-reperfusion , exacerbating thrombotic and inflammatory responses. In DVT models, platelet-derived P-selectin binds to PSGL-1 on neutrophils, triggering NET release that scaffolds formation and propagates venous . Similarly, during ischemia-reperfusion, such as in myocardial or cerebral , P-selectin-mediated neutrophil activation promotes excessive NETosis, contributing to tissue damage and vascular dysfunction. Beyond cardiovascular pathologies, P-selectin drives synovial inflammation in rheumatoid arthritis by supporting leukocyte recruitment to inflamed joints, where its expression on synovial endothelium enhances monocyte and neutrophil infiltration, perpetuating chronic synovitis. In HIV infection, research highlights the role of PSGL-1, a key ligand for P-selectin, in facilitating viral entry; PSGL-1 incorporated into HIV virions interacts with P-selectin on target cells, promoting trans-infection and dissemination. Emerging evidence from stroke models implicates P-selectin in neuroinflammation, where its upregulation post-ischemia recruits leukocytes to the brain, worsening blood-brain barrier disruption and infarct expansion. In , P-selectin contributes to endothelial activation, platelet-leukocyte interactions, and , with elevated soluble P-selectin levels associated with disease severity and risk. Studies as of 2024 indicate that P-selectin facilitates spike protein binding to platelets and endothelial cells, promoting vascular complications.

Therapeutic Targeting

Approved Therapies

Crizanlizumab (Adakveo), a humanized targeting , is the primary approved directly inhibiting this . It received FDA approval on , , for reducing the frequency of vaso-occlusive crises in adult and pediatric patients aged 16 years and older with . The mechanism of crizanlizumab involves binding to the (PSGL-1) interaction site on P-selectin, thereby blocking the of leukocytes to the vascular and reducing inflammation-associated vaso-occlusion. It is administered intravenously at a dose of 5 mg/kg over 30 minutes at week 0 (loading dose), week 2, and every 4 weeks thereafter. Clinical evidence supporting crizanlizumab derives from the phase 2 SUSTAIN trial, a multicenter, randomized, -controlled study in 198 patients with , which demonstrated a 45% reduction in the median annualized rate of sickle cell-related pain crises requiring healthcare intervention (1.63 events per year versus 2.98 for ) with the 5 mg/kg dose. Post-approval pharmacokinetic and pharmacodynamic analyses have confirmed sustained P-selectin inhibition, consistent with phase 2 results. However, the phase 3 STAND trial, a multicenter randomized -controlled study reported in 2025, did not meet its primary endpoints, showing no significant improvement in the annualized rate of vaso-occlusive crises or associated healthcare visits with crizanlizumab versus , although the safety and tolerability profile was consistent with prior studies. Heparin and low-molecular-weight heparins (LMWHs), such as , serve as indirect inhibitors of P-selectin through competition with ligands for binding sites on the molecule. These agents are approved for the prophylaxis and treatment of venous thromboembolism, including deep vein thrombosis and , where their antithrombotic effects partly mitigate P-selectin-mediated leukocyte-platelet interactions, though primary activity stems from enhancement of III-mediated coagulation inhibition. Their use is associated with risks, necessitating careful monitoring in clinical settings.

Emerging Drug Targets and Research

Small-molecule inhibitors targeting P-selectin, often designed as pan-selectin blockers to address overlapping functions among selectins, represent a key area of ongoing research despite setbacks in clinical translation. Rivipansel (GMI-1070), a glycomimetic pan-selectin antagonist, failed to meet primary and secondary efficacy endpoints in a phase 3 trial for vaso-occlusive crisis in sickle cell disease patients in 2019, though it demonstrated safety and tolerability. Recent analogs of rivipansel have shown approximately five-fold improved binding affinity to both E- and P-selectin in preclinical studies, suggesting potential for refined pan-selectin inhibition in thromboinflammatory disorders. Similarly, PSI-421, an oral small-molecule P-selectin inhibitor, has promoted resolution of venous thrombosis in baboon models of deep vein thrombosis without anticoagulation, highlighting its prophylactic potential in preclinical thrombosis research. Therapies targeting (PSGL-1), the primary ligand for P-selectin, are advancing in pipelines for and , with over five candidates in development as of 2024. antibodies such as ALTB-268 modulate PSGL-1 to downregulate chronically activated T cells, offering promise for inflammatory conditions by preferentially targeting exhausted immune cells without broadly suppressing resting T cells. In , PSGL-1 inhibitors are being explored to enhance antitumor immunity, with preclinical data indicating that blocking the PSGL-1 checkpoint reinvigorates T-cell responses in combination with PD-1 blockade. Post-2020 research has illuminated the P-selectin/PSGL-1 axis in , particularly through interactions with immune checkpoints like . A review underscores PSGL-1 and VISTA as co-expressed inhibitory receptors on hematopoietic cells, including T cells, where dual targeting could overcome tumor immune evasion in solid and hematological malignancies. Additionally, elevated P-selectin levels have been implicated in COVID-19-associated , serving as an early of endothelial and risk, which informs targeted antithrombotic strategies in post-viral . Gene therapy and CRISPR-based approaches targeting P-selectin are in preclinical stages for , aiming to reduce endothelial expression and leukocyte recruitment in vascular models. In preclinical models, CRISPR/Cas9-mediated knockdown of P-selectin has attenuated plaque formation by disrupting selectin-dependent , paving the way for to prevent cardiovascular events. P-selectin also emerges as a for , particularly in , where flow cytometry-based assays of P-selectin adhesion predict response to targeted therapies like anti-P-selectin antibodies, enabling patient stratification in clinical settings. Challenges in P-selectin inhibition include functional redundancy with , which compensates for P-selectin blockade in leukocyte rolling and , necessitating pan-selectin or combinatorial strategies. Combination therapies with anti-integrin agents, such as those targeting , show synergistic effects in preclinical models by blocking both initial selectin-mediated tethering and subsequent integrin-dependent firm adhesion, potentially improving outcomes in thromboinflammatory diseases.