Fact-checked by Grok 2 weeks ago

P2Y12

The P2Y<sub>12</sub> receptor is a (GPCR) that binds (ADP), playing a central role in platelet activation, aggregation, and formation, as well as in immune cell functions such as and inflammation modulation. Primarily expressed on platelets, it couples to G<sub>i</sub> proteins to inhibit , reducing cyclic AMP levels and amplifying platelet responses including and fibrinogen binding. Beyond platelets, P2Y<sub>12</sub> is found on , monocytes, macrophages, dendritic cells, and T lymphocytes, where it regulates processes like microglial , T-cell toward Th17 phenotypes, and inflammatory signaling via pathways such as PI3K and calcium . Structurally, the P2Y<sub>12</sub> gene is located on 3q21-q25 and encodes a 342-amino-acid protein with seven transmembrane domains typical of GPCRs, four extracellular residues involved in bonds, and two N-linked sites at the . It forms homo-oligomers within rafts on the and primarily signals through Gα<sub>i2</sub> subunits, though it can interact with other G<sub>i</sub> family members. In platelets, P2Y<sub>12</sub> requires co-stimulation with the P2Y<sub>1</sub> receptor for full ADP-induced aggregation, leading to downstream effects like Rap1b , Akt , and stabilization of platelet aggregates under . In non-hematopoietic cells like , it promotes ADP-dependent motility and contributes to neuroinflammatory responses. Clinically, P2Y<sub>12</sub> is a major therapeutic target for preventing thrombotic events in cardiovascular diseases, with antagonists such as clopidogrel, , and inhibiting its function to reduce platelet reactivity and lower risks of and . Congenital deficiencies in P2Y<sub>12</sub>, often due to mutations like Arg256Gln, result in mild to moderate disorders characterized by impaired platelet aggregation and mucocutaneous hemorrhage. Emerging research highlights its role in immune-related pathologies, including , , , and , suggesting potential for P2Y<sub>12</sub> inhibitors in modulating these conditions beyond antiplatelet therapy, though increased risk remains a key concern.

Molecular biology

Gene and expression

The P2RY12 gene, which encodes the P2Y12 receptor, is located on the long arm of human at position 3q25.1, with genomic coordinates spanning approximately 48 kb from 151,336,843 to 151,384,753 (GRCh38). The gene consists of three exons, two of which are non-coding, and was first cloned and characterized in 2001 through screening of platelet cDNA libraries. It encodes a 342-amino acid protein belonging to the G-protein-coupled receptor family, with a predicted molecular weight of about 39 kDa. Expression of P2RY12 is predominantly observed in platelets and their precursors, megakaryocytes, where it serves as a key regulator of platelet function. Studies using (RT-PCR) and Western blotting have quantified high mRNA and protein levels in these cells, with platelets showing robust surface expression essential for ADP-mediated responses. Lower expression levels are detected in non-hematopoietic cells, including in the , where P2RY12 acts as a homeostatic marker, as confirmed by immunohistochemical and analyses showing selective cytoplasmic and nuclear localization. Additionally, modest expression has been reported in cells and subsets of neurons, with RT-PCR detecting mRNA in isolated cells and Western blots revealing protein in certain neuronal populations, though at levels significantly below those in platelets. The P2RY12 gene exhibits strong evolutionary conservation across mammals, reflecting its critical physiological roles. reveals approximately 86% identity between and orthologs, and 83% identity with the rat ortholog, enabling functional studies in models. This high , particularly in the transmembrane domains, underscores the receptor's preserved structure and signaling mechanisms from to s.

Protein structure

The P2Y12 receptor is a class A G-protein-coupled receptor (GPCR) characterized by a canonical seven-transmembrane (7TM) domain topology, consisting of seven α-helical segments that span the plasma membrane. The N-terminus is located extracellularly and features two potential N-linked glycosylation sites, while the C-terminus is intracellular and includes a conserved helix VIII that runs parallel to the membrane. This architecture positions the ligand-binding pocket within the transmembrane bundle, accessible from the extracellular space. Key structural motifs in P2Y12 include the highly conserved sequence (Asp-Arg-Tyr) at the cytoplasmic end of transmembrane helix 3 (TM3), specifically at residues D120-R121-Y122, which is involved in stabilizing the inactive receptor conformation. A conserved bond between Cys97 in TM3 and Cys175 in extracellular 2 (ECL2) helps maintain the integrity of the ligand-binding , although this bond exhibits flexibility in some structures. Additionally, a bridge connects Cys17 in the to Cys270 in ECL3, further stabilizing the extracellular domains. Insights into the P2Y12 structure have been derived from studies in the 2010s, including high-resolution structures (2.5–3.5 ) of the human receptor bound to antagonists like AZD1283 and agonists such as 2-methylthio- (2MeSADP), a stable analog of the endogenous . These structures reveal a distinct straight conformation of TM5 without the typical proline-induced kink seen in many class A GPCRs, and an extended TM6 that positions key residues toward the orthosteric in the upper transmembrane region. based on earlier GPCR templates, such as or β2-adrenergic receptor, preceded these crystal structures and predicted the overall 7TM bundle, but the resolved P2Y12 models highlighted unique features like the polar pocket for recognition. Specific residues critical for ligand binding include Arg256^{6.55} and Tyr259^{6.58} in TM6, which interact with the moiety of and analogs through hydrogen bonding and π-stacking, respectively. Other key contacts involve Lys280^{7.35} in TM7, which forms ionic interactions with the groups, and residues like His187^{5.47} in TM5 that contribute to both and selectivity in the orthosteric site. These interactions define a binding pocket that accommodates the and diphosphate regions of in a manner distinct from other P2Y receptors.

Physiological function

Role in platelet aggregation

The P2Y12 receptor, a on platelets, is activated by () released from damaged cells or platelet-dense granules, initiating a signaling cascade that sustains platelet aggregation during and . This activation couples to Gi proteins, leading to inhibition of and amplification of () pathways, which in turn enhance the conformational change and ligand-binding affinity of the () . The resulting firm adhesion between platelets via fibrinogen bridges promotes stable aggregate formation. Additionally, P2Y12 signaling triggers the release of alpha and dense granules, releasing further and other pro-aggregatory mediators that provide to reinforce aggregation. P2Y12 integrates with other ADP receptors, notably P2Y1, to orchestrate a coordinated platelet response. P2Y1, coupled to , mediates the initial transient shape change and weak reversible aggregation through intracellular calcium mobilization and transient GPIIb/IIIa activation. In contrast, P2Y12 drives the irreversible, sustained phase of aggregation by prolonging GPIIb/IIIa engagement and granule secretion, ensuring consolidation. Full ADP-induced aggregation requires simultaneous activation of both receptors, as isolated P2Y12 signaling alone is insufficient for initiation but critical for amplification and stability. Studies using P2Y12 mice provide direct evidence of the receptor's essential role . These mice display markedly prolonged tail bleeding times compared to wild-type controls, reflecting impaired hemostatic plug formation. Under flow conditions or vascular injury models, such as ferric chloride-induced mesenteric artery damage, in knockout mice grow slowly, remain small (typically 25–50 μm), and frequently embolize rather than occlude, demonstrating reduced thrombus stability and highlighting P2Y12's contribution to platelet accumulation and retention at injury sites. analyses confirm defective platelet activation, including diminished P-selectin exposure, fibrinogen binding, and aggregation responses to low concentrations (e.g., 0.5 μM). In vitro assessments quantify P2Y12's dominance in ADP responses, where the receptor mediates the majority (~50–70%) of maximal aggregation induced by physiological ADP levels (1–10 μM), with deficiencies resulting in only partial, reversible aggregation even at high concentrations. This underscores P2Y12's role in amplifying weak stimuli into robust, thrombus-stabilizing events essential for preventing excessive while minimizing pathologic clotting.

Functions in other cell types

Beyond platelets, the P2Y12 receptor is expressed in various non-hematopoietic and hematopoietic cells, including , monocytes, macrophages, mast cells, dendritic cells, (VSMCs), and T lymphocytes, where it modulates immune responses, migration, and inflammatory processes. Tissue-specific expression studies, such as RT-PCR and on and tissues, confirm P2Y12 mRNA and protein presence in these cell types, with functional validation through knockout models and antagonist treatments demonstrating roles in cellular and signaling. In , the resident immune cells of the , P2Y12 serves as a key sensor for extracellular released from damaged neurons or during , promoting rapid extension and toward injury sites. This G_i-coupled receptor activates downstream pathways involving PI3K, Akt, and Rac, facilitating microglial migration to maintain tissue and blood- barrier integrity, as evidenced in models of where P2Y12-deficient microglia show impaired and reduced . Additionally, P2Y12 signaling couples with apoptotic progression, enabling efficient engulfment of dying cells to prevent secondary ; inhibition of P2Y12 signaling delays clearance of apoptotic cells in retinal models. In stroke, P2Y12 activation drives microglial and of necrotic debris in ischemic regions, but excessive signaling exacerbates , with genetic knockout or pharmacological blockade (e.g., clopidogrel) reducing neuronal and proinflammatory release in rodent models of . In monocytes and macrophages, P2Y12 promotes and , enhancing migration to sites of and clearance of apoptotic cells, which contributes to resolution of but can also drive chronic states in diseases like . In dendritic cells, it modulates and production, influencing adaptive immune responses. In T lymphocytes, P2Y12 signaling supports differentiation toward pro-inflammatory Th17 phenotypes via PI3K pathways, amplifying autoimmune and inflammatory conditions. In mast cells, P2Y12 expression on perivascular and mucosal subsets enhances responsiveness to cysteinyl leukotrienes like LTE4, a key mediator of allergic . Activation of P2Y12 on these cells triggers , releasing and proteases, alongside production (e.g., TNF-α, IL-6), which amplifies recruitment and airway hyperreactivity in models of and . Evidence from P2Y12-deficient mice shows abolished LTE4-induced pulmonary , including reduced vascular leakage and influx, highlighting its contribution to reactions without direct stimulation. In VSMCs, P2Y12 modulates vascular tone and remodeling by coupling ADP signaling to G_i-mediated inhibition of /PKA pathways, leading to cofilin dephosphorylation and actin cytoskeleton reorganization that promotes cell contraction and . This is particularly relevant in , where P2Y12 upregulation by oxidized LDL drives VSMC proliferation and intimal invasion, increasing plaque formation; in ApoE^{-/-} mice, chronic clopidogrel treatment significantly reduces atherosclerotic lesions via diminished VSMC , as shown in transwell assays and plaque . P2Y12 also impairs efflux in VSMCs, fostering [foam cell](/page/foam cell) accumulation and lipid-laden plaque progression. Selective P2Y12 inhibitors, such as clopidogrel and , attenuate immune cell across these types by blocking ADP-induced ; for instance, in microglial cultures, antagonists reduce by over 60% in response to injury signals, while in VSMCs and leukocytes, they suppress - or inflammation-driven in transplant models, underscoring P2Y12's broader role in non-hemostatic immunity.

Pharmacology

Endogenous ligands and signaling

The P2Y12 receptor, a , is primarily activated by the endogenous ligand (ADP), which serves as its main with a binding affinity (Kd) of approximately 1-10 μM (pKi = 5.9). (ATP) acts as a weaker compared to ADP, exhibiting lower potency in stimulating the receptor. Upon , ADP induces conformational changes that facilitate receptor coupling to heterotrimeric G proteins of the Gi/Go family, predominantly Gαi2. This coupling leads to the inhibition of activity, resulting in decreased intracellular (cAMP) levels from a basal state through Gi-mediated suppression, thereby modulating platelet responsiveness and other cellular functions. Downstream signaling is mediated by the released Gβγ subunits, which activate (PI3K), promoting phosphoinositide hydrolysis and subsequent activation of the Akt pathway. PI3K further facilitates the loading of (GTP) onto Rap1b, a , by inhibiting the GTPase-activating protein RASA3, which ultimately supports activation essential for cellular adhesion processes. To prevent prolonged activation, P2Y12 undergoes rapid desensitization following stimulation, primarily through of its C-terminal tail by kinases (GRKs), including GRK2 and GRK6. This recruits β-arrestins, which sterically hinder further interactions, leading to receptor uncoupling and .

Synthetic agonists and antagonists

Synthetic agonists of the P2Y12 receptor are primarily utilized in research settings to study receptor activation and signaling pathways, as they mimic the effects of endogenous but offer greater stability and selectivity. A prominent example is 2-methylthio- (2-MeSADP), a potent that binds orthosterically to the receptor's extracellular domain, inducing G_i-mediated inhibition of with a pEC50 of 8.3 in platelet assays. This compound has been instrumental in structural studies, including the 2.5 Å of the human P2Y12 receptor bound to 2-MeSADP, which reveals key interactions in the transmembrane bundle for recognition. Unlike endogenous ligands, synthetic like 2-MeSADP lack clinical applications due to their short and potential off-target effects on other P2Y subtypes, limiting them to experimental contexts such as probing receptor desensitization. In contrast, synthetic antagonists dominate pharmacological interest in P2Y12, developed since the through platelet aggregation assays that identified inhibitors of ADP-induced responses prior to receptor . Thienopyridines like were developed as antiplatelet agents through screening for inhibition of ADP-induced platelet aggregation, with first introduced in 1978, marking the initial identification of P2Y12-specific antagonists from functional screens. Subsequent advancements led to clopidogrel, approved in 1997, which acts as a metabolized in the liver to an active that irreversibly inhibits the receptor by forming a covalent bond with Cys105 in the first extracellular loop, preventing ADP binding with high specificity. This irreversible mechanism ensures prolonged inhibition but requires daily dosing due to the absence of reversibility. Reversible antagonists represent a newer class, offering faster onset and offset compared to thienopyridines. , developed from ATP analogs in the early 2000s, binds allosterically to an extracellular pocket distinct from the orthosteric site, exerting with a pIC50 of 7.9 ( ≈ 126 ) in platelet aggregation assays. Structural insights from cryo-EM studies confirm ticagrelor's interaction stabilizes an inactive receptor conformation without covalent modification, allowing reversibility within hours. Other reversible agents, such as the intravenous , similarly target the allosteric site but are limited to acute settings due to their short duration. These synthetic antagonists' development was guided by seminal works on receptor structure, including the 2014 of antagonist-bound P2Y12, which elucidated binding pockets for rational .

Clinical significance

Antiplatelet therapy mechanisms

P2Y12 receptor blockade by antiplatelet agents inhibits ()-induced platelet activation, a critical step in the amplification of platelet aggregation and formation. By preventing ADP binding to the P2Y12 receptor on platelet surfaces, these inhibitors disrupt downstream signaling pathways, including the inhibition of and subsequent reduction in cyclic AMP levels, which normally promote platelet shape change, granule release, and fibrinogen binding to the αIIbβ3 . This blockade thereby limits the stabilization and growth of thrombi, reducing the risk of and ischemic events such as or . For instance, clopidogrel, an irreversible P2Y12 inhibitor, typically achieves 50-60% inhibition of ADP-induced platelet aggregation in responsive patients following standard dosing. Dual antiplatelet therapy combining a P2Y12 inhibitor with aspirin provides synergistic inhibition by targeting complementary pathways: aspirin irreversibly acetylates cyclooxygenase-1 to block production, while P2Y12 inhibitors prevent ADP-mediated amplification. This combination yields additive suppression of platelet function, often resulting in 70-80% overall reduction in platelet aggregation compared to monotherapy, enhancing efficacy without complete overlap in mechanisms. Clinical studies, such as trial, have demonstrated this in reducing composite cardiovascular outcomes by approximately 20% relative to aspirin alone. P2Y12 inhibitors differ in their binding kinetics, with irreversible agents like clopidogrel forming a that persists for the platelet's lifespan (approximately 7-10 days), leading to a prolonged offset time of 5-7 days after discontinuation as new platelets are produced. In contrast, reversible inhibitors such as bind non-covalently with a of about 12 hours, allowing for a faster of platelet function within 24-48 hours post-cessation, which may facilitate urgent or management. The primary on-target adverse effect of P2Y12 inhibition is a mild increase in bleeding risk, attributable to impaired primary through diminished platelet aggregation at sites of vascular injury. This manifests as prolonged and higher rates of minor and major hemorrhagic events, particularly with dual , though the absolute risk remains low (e.g., 1-3% for major bleeding in large trials). Potent inhibitors like may elevate non-procedural bleeding compared to clopidogrel, but overall profiles are comparable when adjusted for ischemic benefits.

Indications and guidelines

P2Y12 inhibitors, used in combination with aspirin as dual antiplatelet therapy (DAPT), are indicated for the prevention of atherothrombotic events in patients with (ACS), following (PCI), and in select cases of stable (CAD). In ACS and post-PCI settings, these agents reduce the risk of cardiovascular death, (MI), and by inhibiting platelet aggregation. For stable CAD patients undergoing PCI, DAPT is recommended for a minimum of 6 months to mitigate stent thrombosis and ischemic events. The Clopidogrel for the Reduction of Events During Observation () trial demonstrated the benefit of clopidogrel plus aspirin in patients following , with up to 12 months of therapy reducing the composite endpoint of death, , or by 26.9% (95% CI, 3.9% to 44.5%; P=0.03) compared to aspirin alone after an initial of clopidogrel. According to the 2025 ACC/AHA guidelines, DAPT with aspirin and a P2Y12 (preferably or over clopidogrel) is recommended for at least 12 months in ACS patients without high risk, with durations shortened to 1-6 months in those at elevated risk followed by P2Y12 monotherapy. Prasugrel is specifically indicated for high-risk in ACS patients, as evidenced by the TRITON-TIMI 38 , where it reduced the primary efficacy endpoint (composite of cardiovascular death, , or ) from 12.1% with clopidogrel to 9.9% ( 0.81, 95% CI 0.73-0.90). In patients with ST-elevation (STEMI) receiving , clopidogrel as an adjunct reduces the odds of an occluded infarct-related artery, death, or recurrent before by 36%, per the CLARITY-TIMI 28 . For secondary prevention in minor ischemic or high-risk (TIA), short-term DAPT with aspirin and clopidogrel is recommended for 21 days to reduce recurrent risk, based on /ASA guidelines supported by trials like and POINT. Comparative efficacy among P2Y12 inhibitors favors over clopidogrel in ACS, as shown in the trial, where reduced the primary endpoint (vascular death, , or ) from 11.7% to 9.8% ( 0.84, 95% CI 0.77-0.92) and vascular mortality from 5.1% to 4.0%. The 2025 / guidelines endorse or as preferred P2Y12 inhibitors in ACS with due to this superior efficacy, reserving clopidogrel for high-bleeding-risk patients or contraindications to the others. Congenital P2Y<sub>12</sub> deficiency, resulting from biallelic mutations in the P2Y<sub>12</sub> gene, manifests as a mild bleeding disorder with prolonged , impaired ADP-induced platelet aggregation, and episodes of mucocutaneous hemorrhage, , and menorrhagia. typically involves platelet function testing, and management focuses on avoiding antiplatelet agents and using or platelet transfusions for episodes.

Research and future directions

Genetic variants and pharmacogenomics

The P2RY12 , encoding the P2Y12 receptor, harbors several common genetic variants that influence receptor expression and platelet function. The H1/H2 , defined by single nucleotide polymorphisms (SNPs) such as rs6809699 (G52T), rs10935838 (i-C139T), rs2046934 (i-T744C), and rs5853517 (i-ins801A), affects promoter and regulatory regions, with the H2 associated with increased transcriptional activity and higher P2Y12 receptor density on platelets. This leads to enhanced ADP-induced maximal platelet aggregation in H2 carriers compared to H1 homozygotes. Additionally, the CYP2C19*2 allele (rs4244285), a loss-of-function variant in the 2C19 enzyme required for clopidogrel bioactivation, reduces formation of the by approximately 30-50% in heterozygotes. These variants contribute to interindividual variability in response, particularly for clopidogrel, a targeting the P2Y12 receptor. Carriers of the H2 haplotype exhibit higher residual platelet reactivity on clopidogrel treatment, with increased odds of high on-treatment platelet reactivity (OR 1.45 for G52T; 95% CI 1.14-1.85). Similarly, CYP2C19 poor metabolizers (homozygous for *2 or other loss-of-function alleles, prevalence 2% in Caucasians) show markedly diminished clopidogrel efficacy, with 1.5- to 2-fold higher risk of , such as , compared to normal metabolizers. The U.S. issued a in 2010 highlighting reduced clopidogrel effectiveness in poor metabolizers. Pharmacogenomic testing for variants is recommended by the Clinical Pharmacogenetics Implementation Consortium (CPIC) for high-risk patients, such as those undergoing , to guide therapy selection. In intermediate or poor metabolizers, alternatives like or —direct-acting P2Y12 inhibitors not dependent on —are preferred at standard doses to achieve adequate platelet inhibition without increased bleeding risk. P2RY12 genotyping, while less routinely implemented, may identify H2 carriers at risk for suboptimal response, though evidence for routine use remains limited. Genome-wide association studies (GWAS) in the have further elucidated P2RY12's role in platelet reactivity variability. A meta-analysis of over 66,000 individuals identified a locus near P2RY12 (3q21.1) significantly associated with ADP-induced platelet aggregation (P = 2.5 × 10^{-11}), confirming its influence beyond candidate gene approaches. Subsequent studies linked specific P2RY12 SNPs, such as rs2046934, to altered clopidogrel in diverse populations, supporting personalized antiplatelet strategies.

Emerging therapeutic targets

Research into P2Y12 receptors has expanded beyond traditional antiplatelet applications, identifying potential therapeutic roles in modulating . In preclinical models of ischemic , P2Y12 inhibitors such as have demonstrated neuroprotective effects by reducing microglial activation and migration toward injury sites, thereby limiting the progression of neuroinflammatory responses. For instance, in rat models of permanent focal cerebral ischemia, administration significantly decreased infarct volume and improved neurological outcomes, an effect attributed to the inhibition of P2Y12-mediated microglial and proinflammatory signaling via Gαi and PI3K pathways. Similarly, the reversible inhibitor , with its rapid onset and offset, has shown promise in preclinical studies by attenuating microglial activation and , potentially reducing infarct size in models, though clinical translation remains under investigation. In , P2Y12 signaling has emerged as a contributor to tumor-associated and metastatic processes, prompting exploration of inhibitors for anticancer applications. Studies in models indicate that P2Y12 antagonists, such as clopidogrel, reduce cancer-associated and inhibit tumor by disrupting platelet-tumor cell interactions that promote emboli formation and dissemination. More specifically, in , P2Y12 receptor antagonism suppresses cell proliferation, migration, and invasion while inducing , as evidenced by in vitro experiments where ticagrelor treatment decreased glioma cell motility through blockade of ADP-induced signaling. Recent research, including TCGA database analyses, has confirmed elevated P2Y12 expression in tissues compared to other tumors, highlighting its role in tumor progression and suggesting P2Y12 as a target for therapies in high-grade gliomas. Development of novel P2Y12 inhibitors focuses on allosteric modulators and antibody-based approaches to achieve reversible, short-acting inhibition suitable for periprocedural or non-chronic use. , an allosteric antagonist that binds a site distinct from the orthosteric pocket, exemplifies this class and has informed structural studies revealing opportunities for designing subtype-selective modulators with enhanced specificity. , a reversible small-molecule inhibitor, advanced to phase trials (INNOVATE-PCI) for acute coronary syndromes, demonstrating rapid platelet inhibition and recovery, which could extend to short-term applications in non-cardiac settings. therapies, such as anti-P2Y12 monoclonal antibodies, have shown functional blockade of platelet aggregation and , offering potential for targeted, parenteral inhibition with minimized off-target effects, though further clinical development is needed. A key challenge in repurposing P2Y12 inhibitors for non-cardiac indications like or is balancing efficacy against heightened risks, particularly in patients with comorbidities. Potent inhibitors increase major events compared to clopidogrel, complicating their use in contexts without routine cardiovascular , such as or . Strategies like to less potent agents or short-duration therapy aim to mitigate this, but evidence from high--risk cohorts underscores the need for personalized approaches to optimize benefits in diverse applications.

References

  1. [1]
    Central role of the P2Y12 receptor in platelet activation - PMC
    The P2Y 12 receptor, activated by ADP, plays a central role in platelet activation and is the target of P2Y 12 receptor antagonists that have proven ...
  2. [2]
    The platelet P2Y12 receptor for adenosine diphosphate: congenital ...
    P2Y12, the Gi-coupled platelet receptor for adenosine diphosphate (ADP), plays a central role in platelet function. Patients with congenital P2Y12 defects ...
  3. [3]
    The Signaling Pathway of the ADP Receptor P2Y 12 in the Immune ...
    P2Y 12 is a G-protein-coupled receptor (GPCR) found first on platelets and microglia and is activated by ADP but also by ATP binding.
  4. [4]
    P2RY12 purinergic receptor P2Y12 [ (human)] - NCBI
    Sep 5, 2025 · The results suggest that P2Y12 is a useful marker for the identification of human microglia throughout the lifespan. Moreover, P2Y12 expression ...Missing: chromosome exons
  5. [5]
    600515 - PURINERGIC RECEPTOR P2Y, G PROTEIN-COUPLED ...
    HGNC Approved Gene Symbol: P2RY12. Cytogenetic location: 3q25.1 Genomic coordinates (GRCh38) : 3:151,336,843-151,384,753 (from NCBI). Gene-Phenotype ...
  6. [6]
    Identification of the platelet ADP receptor targeted by antithrombotic ...
    Jan 11, 2001 · Here we describe the cloning of this receptor, designated P2Y 12 , and provide evidence that a patient with a bleeding disorder 10 has a defect in this gene.
  7. [7]
    P2RY12 - P2Y purinoceptor 12 - Homo sapiens (Human) | UniProtKB
    Mar 1, 2001 · Receptor for ADP and ATP coupled to G-proteins that inhibit the adenylyl cyclase second messenger system. Not activated by UDP and UTP.Missing: span | Show results with:span
  8. [8]
    P2RY12 protein expression summary - The Human Protein Atlas
    Selective cytoplasmic and nuclear expression in microglia and subsets of cells in bone marrow and lymphoid tissues. Main subcellular location based on data ...Missing: megakaryocytes mast
  9. [9]
    Adenine nucleotides inhibit cytokine generation by human mast ...
    Dec 15, 2004 · We determined the expression and function of adenine nucleotide-specific P2Y receptors on cord blood-derived human mast cells (hMCs).
  10. [10]
  11. [11]
  12. [12]
    Structure of the human P2Y12 receptor in complex with an ... - NIH
    The structure reveals a distinct straight conformation of helix V, which sets P2Y12R apart from all other known class A GPCR structures. With AZD1283 bound, the ...
  13. [13]
    Agonist-bound structure of the human P2Y12 receptor - PMC - NIH
    Here, we report the structures of the human P2Y12R in complex with a full agonist 2-methylthio-adenosine-5′-diphosphate (2MeSADP, a close analogue of endogenous ...
  14. [14]
    Role of ADP Receptor P2Y12 in Platelet Adhesion and Thrombus ...
    Recently, Hollopeter et al described the cloning of the ADP receptor P2Y12 ... 2001). The reduced platelet consumption by the growing thrombus ...
  15. [15]
    P2Y12 regulates platelet adhesion/activation, thrombus growth ... - NIH
    P2Y 12 is involved in several key steps of thrombosis: platelet adhesion/activation, thrombus growth, and stability.<|control11|><|separator|>
  16. [16]
    Adenosine Diphosphate–Induced Platelet Aggregation Is ...
    The adenosine diphosphate (ADP) receptor P2Y12 plays a pivotal role in platelet aggregation, as demonstrated by the benefit conferred by its blockade in ...Platelet Aggregation Studies · P2y Receptor · Results<|separator|>
  17. [17]
    P2Y12 receptors in platelets and other hematopoietic and non ...
    Beyond the platelet physiology and pharmacology, recent studies have revealed the expression of the P2Y12 receptor in other hematopoietic cells including ...
  18. [18]
  19. [19]
  20. [20]
    Microglial P2Y 12 Deficiency/Inhibition Protects against Brain Ischemia
    P2Y12 participates in ischemia related inflammation by mediating microglial migration and potentiation of neurotoxicity. These data also suggest an additional ...<|control11|><|separator|>
  21. [21]
    Leukotriene E4-induced pulmonary inflammation is mediated by the ...
    Oct 26, 2009 · P2Y12 receptor expression permits LTE4-induced activation of extracellular signal-regulated kinase in Chinese hamster ovary cells and permits ...
  22. [22]
    P2Y 12 receptor - IUPHAR/BPS Guide to PHARMACOLOGY
    An in silico screening for endogenous ligands acting on the human P2Y12 receptor revealed cysteinylleukotrienes (CysLTs) and 5-phosphoribosyl 1-pyrophosphate ( ...
  23. [23]
    Update of P2Y receptor pharmacology: IUPHAR Review 27
    Feb 9, 2020 · Although P2Y1 and P2Y12 receptors share the same endogenous ligand ADP, the structures of these two receptors reveal (Figure 4) completely ...
  24. [24]
    Central role of the P2Y 12 receptor in platelet activation - JCI
    The P2Y 12 receptor, activated by ADP, plays a central role in platelet activation and is the target of P2Y 12 receptor antagonists that have proven ...
  25. [25]
    P2Y1 and P2Y12 receptors for ADP desensitize by distinct kinase ...
    Data are expressed as cAMP production (pmol cAMP/well) or as a percentage of inhibition of forskolin-stimulated adenylyl cyclase. ... ADP-induced aggregation ...
  26. [26]
    P2Y12 Antagonists in Cardiovascular Disease—Finding the Best ...
    Ticlopidine, originally developed in the search for an anti-inflammatory drug, was the first P2Y12 inhibitor available for clinical use in the early1990's.
  27. [27]
    The active metabolite of Clopidogrel disrupts P2Y12 receptor ...
    Jul 18, 2006 · In this study, we further deciphered the mechanism of action of clopidogrel and of its active metabolite (Act-Met) on P2Y12 receptors.
  28. [28]
    P2Y12 receptors: structure and function - ScienceDirect.com
    The platelet P2Y12 receptor (P2Y12R) for adenosine 5′diphosphate (ADP) plays a central role in platelet function, hemostasis, and thrombosis.
  29. [29]
    New P2Y12 Inhibitors | Circulation
    Jan 5, 2010 · Ticagrelor is a less potent P2Y12 antagonist than cangrelor, with a Pic50 value of 7.9 for inhibition of 30 μmol/L ADP–induced aggregation of ...Missing: IC50 | Show results with:IC50
  30. [30]
    Clopidogrel Resistance | Circulation
    Clopidogrel resistance, empirically defined as <10% reduction in aggregation in response to 5 μmol/L ADP compared with pretreatment values, was seen in 63% of ...
  31. [31]
    Aspirin and Clopidogrel in Acute Coronary Syndromes
    The meta-analysis found a 25% reduction in the combined end point of MI, stroke, and vascular death in users of antiplatelet therapy (P<.001). There was also a ...
  32. [32]
    International Expert Consensus on Switching Platelet P2Y 12 ...
    Oct 30, 2017 · This expert consensus provides an overview of the pharmacology of P2Y 12 inhibitors, different modalities and definitions of switching, and available literature
  33. [33]
  34. [34]
    2025 ACC/AHA/ACEP/NAEMSP/SCAI Guideline for the ...
    Dual antiplatelet therapy with aspirin and an oral P2Y12 inhibitor is indicated for at least 12 months as the default strategy in patients with ACS who are ...
  35. [35]
    Twelve Versus Six Months of Clopidogrel to Reduce Major Cardiac ...
    Conclusions— Twelve months of clopidogrel is superior to 6 months in reducing overall major cardiac events and revascularization rates at 15 months for patients ...
  36. [36]
    Prasugrel versus Clopidogrel in Patients with Acute Coronary ...
    Results. The primary efficacy end point occurred in 12.1% of patients receiving clopidogrel and 9.9% of patients receiving prasugrel (hazard ratio for prasugrel ...
  37. [37]
    Addition of Clopidogrel to Aspirin and Fibrinolytic Therapy for ...
    By 30 days, clopidogrel therapy reduced the odds of the composite end point of death from cardiovascular causes, recurrent myocardial infarction, or recurrent ...
  38. [38]
    Use of Dual Antiplatelet Therapy Following Ischemic Stroke
    Apr 8, 2020 · Aspirin plus clopidogrel for 21 to 30 days after TIA or minor stroke reduces recurrent stroke. •. Aspirin plus clopidogrel is more effective ...Missing: P2Y12 | Show results with:P2Y12
  39. [39]
    Ticagrelor versus Clopidogrel in Patients with Acute Coronary ...
    Aug 30, 2009 · Treatment with ticagrelor as compared with clopidogrel significantly reduced the rate of death from vascular causes, myocardial infarction, or stroke.
  40. [40]
    Influence of Genetic and Epigenetic Factors of P2Y12 Receptor on ...
    Aug 9, 2022 · Available resources regarding the influence of H1/H2 on the efficacy of other P2Y12 inhibitors are less abundant than for clopidogrel. One study ...H1/h2 Haplotype And Related... · C34t And P2y Inhibitors · Other P2y12 Haplotypes And...
  41. [41]
    P2Y12 H2 haplotype is associated with peripheral arterial disease
    Background: We recently described a gain-of-function haplotype, called H2, of the adenosine diphosphate (ADP) receptor P2Y12 gene associated with increased ...Missing: al | Show results with:al
  42. [42]
    Cytochrome P450 2C19 loss-of-function polymorphism is a major ...
    Oct 1, 2006 · The CYP2C19*2 loss-of-function allele is associated with a marked decrease in platelet responsiveness to clopidogrel in young healthy male ...Patients, Materials, And... · Laboratory Methods · Results
  43. [43]
    Effects of CYP2C19 Genotype on Outcomes of Clopidogrel Treatment
    Aug 29, 2010 · We genotyped patients from two large, randomized trials that showed that clopidogrel, as compared with placebo, reduced the rate of cardiovascular events.
  44. [44]
    Reduced effectiveness of Plavix in patients who are poor metabolizers
    Aug 3, 2017 · [03-12-2010] The U.S. Food and Drug Administration (FDA) has added a Boxed Warning to the label for Plavix, the anti-blood clotting medication.
  45. [45]
    CPIC® Guideline for Clopidogrel and CYP2C19
    Most recent guideline publication: Clinical Pharmacogenetics Implementation Consortium Guideline for CYP2C19 Genotype and Clopidogrel Therapy: 2022 updateMissing: high- risk prasugrel
  46. [46]
    Clinical Pharmacogenetics Implementation Consortium Guideline ...
    Jan 16, 2022 · The reported literature indicates that both CYP2C19 IMs and PMs exhibit reduced clopidogrel active metabolite formation, increased on-treatment ...
  47. [47]
    Common Variation in the Platelet Receptor P2RY12 Gene Is ...
    The P2RY12 gene consists of 3 exons (E1, E2, and E3, of which only E3 is coding) and 2 intervening sequences (IVS1 and IVS2), which span 47 kb of genomic DNA on ...
  48. [48]
    Genome-wide meta-analyses identifies 7 loci associated with ...
    Platelet function mediates both beneficial and harmful effects on human health, but few genes are known to contribute to variability in the process.
  49. [49]
  50. [50]
    (PDF) Microglia is a Key Player in the Reduction of Stroke Damage ...
    Mar 19, 2014 · Our results show that ticagrelor is protective against ischemia-induced cerebral injury and this effect is mediated, at least partly, by ...
  51. [51]
    P2RY12-Inhibitors Reduce Cancer-Associated Thrombosis and ...
    We previously demonstrated that anti-P2RY12 drugs inhibit cancer-associated-thrombosis and formation of tumor metastasis in pancreatic cancer models.
  52. [52]
    P2Y12 receptor antagonism inhibits proliferation, migration and ...
    Aug 8, 2022 · TCGA data analysis showed a higher expression of P2Y12R in gliomas samples when compared to the other tumors. These data demonstrate the ...Missing: 2020s | Show results with:2020s
  53. [53]
    [PDF] Newer agents in antiplatelet therapy: a review.
    Jan 1, 2012 · 25 Ticagrelor exerts its action via binding to the. P2Y12 receptor at a site distinct from the ADP binding site, thus making it an allosteric ...
  54. [54]
    the INNOVATE-PCI trial - PubMed
    A randomized, double-blind, active-controlled phase 2 trial to evaluate a novel selective and reversible intravenous and oral P2Y12 inhibitor elinogrel ...Missing: allosteric modulators antibodies development short- acting
  55. [55]
    P2Y 12 antibody inhibits platelet activity and protects ... - PubMed
    Nov 18, 2017 · The P2Y 12 Ab functionally blocks platelet activity in vitro and in vivo, and support the notion that it can be purposed as a parenteral antiplatelet agent.
  56. [56]
    De-Escalation of P2Y12-Inhibiting Therapies to Reduce the Risk of ...
    Oct 7, 2021 · However, this benefit is hampered by an increase in major bleeding events, underscoring the need for strategies to reduce bleeding risk without ...
  57. [57]
    Antiplatelet Therapy in High-Bleeding Risk Patients Undergoing PCI
    The present review discusses the evidence base for new pharmacotherapeutic strategies to decrease bleeding risk without compromising ischemic protection among ...<|control11|><|separator|>