91 results on '"Protease-activated receptor 4"'
Search Results
2. MicroRNA‐26a alleviates tubulointerstitial fibrosis in diabetic kidney disease by targeting PAR4.
- Author
-
Qu, Gaoting, Li, Xingyue, Jin, Ran, Guan, Dian, Ji, Jialing, Li, Shanwen, Shi, Huimin, Tong, Pingfan, Gan, Weihua, and Zhang, Aiqing
- Subjects
DIABETIC nephropathies ,RENAL fibrosis ,PROTEASE-activated receptors ,MICRORNA ,ADENO-associated virus - Abstract
Our previous study found that miR‐26a alleviates aldosterone‐induced tubulointerstitial fibrosis (TIF). However, the effect of miR‐26a on TIF in diabetic kidney disease (DKD) remains unclear. This study clarifies the role and possible mechanism of exogenous miR‐26a in controlling the progression of TIF in DKD models. Firstly, we showed that miR‐26a was markedly decreased in type 2 diabetic db/db mice and mouse tubular epithelial cells (mTECs) treated with high glucose (HG, 30 mM) using RT‐qPCR. We then used adeno‐associated virus carrying miR‐26a and adenovirus miR‐26a to enhance the expression of miR‐26a in vivo and in vitro. Overexpressing miR‐26a alleviated the TIF in db/db mice and the extracellular matrix (ECM) deposition in HG‐stimulated mTECs. These protective effects were caused by reducing expression of protease‐activated receptor 4 (PAR4), which involved in multiple pro‐fibrotic pathways. The rescue of PAR4 expression reversed the anti‐fibrosis activity of miR‐26a. We conclude that miR‐26a alleviates TIF in DKD models by directly targeting PAR4, which may provide a novel molecular strategy for DKD therapy. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
3. An optimized agonist peptide of protease-activated receptor 4 and its use in a validated platelet-aggregation assay
- Author
-
Jing Yang, Claudio Mapelli, Zhaoqing Wang, Chi Shing Sum, Ji Hua, R. Michael Lawrence, Yan Ni, and Dietmar A. Seiffert
- Subjects
antiplatelet assay ,platelet ,protease-activated receptor 4 ,thrombin ,thrombosis ,Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Protease-activated receptor 4 (PAR4) is a promising drug target to improve the efficacy/safety window of antiplatelet agents. The native peptide GYPGQV, and the more-potent peptide AYPGKF, are PAR4-specific activators. However, these PAR4 agonist peptides (APs) elicit an agonist response, for example, platelet aggregation, at concentrations of 50 to 1000 µM in platelet-function assays, thereby limiting their utility to monitor the pharmacodynamic effects of PAR4 antagonists over a wide concentration range. Improved pharmacodynamic assays are needed for clinical development of PAR4 antagonists. We attempted to identify potent PAR4 APs to aid development of robust assays for optimization of PAR4 antagonists. Using an AYPG-based biased phage-display peptide library approach followed by chemical peptide optimization, A-Phe(4-F)-PGWLVKNG was identified. This peptide demonstrated an EC50 value of 3.4 µM in a platelet-aggregation assay, which is 16-fold more potent than AYPGKF. Using this new PAR4 AP, a platelet-rich plasma-aggregation assay using light-transmission aggregometry was developed and validated in a series of precision and reproducibility tests. PAR4 antagonist responses to PAR4 AP A-Phe(4-F)-PGWLVKNG (12.5 µM to 100 µM) were subsequently evaluated in this assay in vitro and ex vivo in a human study using BMS-986120, a PAR4 antagonist that entered clinical studies.
- Published
- 2022
- Full Text
- View/download PDF
4. Discovery of quinazoline-benzothiazole derivatives as novel potent protease-activated receptor 4 antagonists with improved pharmacokinetics and low bleeding liability.
- Author
-
Li, Shanshan, Liu, Shangde, Yuan, Duo, Liu, Renjie, Hu, Lifang, and Zhu, Xiong
- Subjects
- *
PROTEASE-activated receptors , *STRUCTURE-activity relationships , *LIVER microsomes , *GEOGRAPHICAL discoveries , *BENZOTHIAZOLE derivatives - Abstract
Protease-activated receptor 4 (PAR4) plays a critical role in the development of pathological thrombosis, and targeting PAR4 is considered a promising strategy for improving antiplatelet therapies. Here, we reported the design of a series of quinazoline-benzothiazole-based PAR4 antagonists using a scaffold-hopping strategy. Systematic structure-activity relationship exploration leads to the discovery of compounds 20f and 2 0g , which displayed optimal activity (h. PAR4-AP PRP IC 50 = 6.39 nM and 3.45 nM, respectively) on human platelets and high selectivity for PAR4. Both of them also showed excellent metabolic stability in human liver microsomes (compound 20f , T 1/2 = 249.83 min, compound 20g , T 1/2 = 282.60 min) and favourable PK profiles in rats (compound 20f , T 1/2 = 5.16 h, F = 50.5 %, compound 20g , T 1/2 = 7.05 h, F = 27.3 %). More importantly, neither compound prolonged the bleeding time in the mouse tail-cutting model (10 mg/kg, p.o.). These results suggest that these compounds have great potential for use in antiplatelet therapies. [Display omitted] • Starting from compound XZ-13, a series of quinazoline benzothiazole derivatives were designed and synthesized. • Compounds 20f and 20g were highly potent and selective PAR4 antagonists. • Compounds 20f and 20g exhibited excellent i n vitro metabolic properties. • Compounds 20f and 20g have improved pharmacokinetics and do not prolong bleeding time in a mouse haemostasis model. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
5. An optimized agonist peptide of protease-activated receptor 4 and its use in a validated platelet-aggregation assay.
- Author
-
Yang, Jing, Mapelli, Claudio, Wang, Zhaoqing, Sum, Chi Shing, Hua, Ji, Lawrence, R. Michael, Ni, Yan, and Seiffert, Dietmar A.
- Subjects
PROTEASE-activated receptors ,PEPTIDE receptors ,PEPTIDES ,BLOOD platelet aggregation ,THROMBIN receptors - Abstract
Protease-activated receptor 4 (PAR4) is a promising drug target to improve the efficacy/safety window of antiplatelet agents. The native peptide GYPGQV, and the more-potent peptide AYPGKF, are PAR4-specific activators. However, these PAR4 agonist peptides (APs) elicit an agonist response, for example, platelet aggregation, at concentrations of 50 to 1000 µM in platelet-function assays, thereby limiting their utility to monitor the pharmacodynamic effects of PAR4 antagonists over a wide concentration range. Improved pharmacodynamic assays are needed for clinical development of PAR4 antagonists. We attempted to identify potent PAR4 APs to aid development of robust assays for optimization of PAR4 antagonists. Using an AYPG-based biased phage-display peptide library approach followed by chemical peptide optimization, A-Phe(4-F)-PGWLVKNG was identified. This peptide demonstrated an EC
50 value of 3.4 µM in a platelet-aggregation assay, which is 16-fold more potent than AYPGKF. Using this new PAR4 AP, a platelet-rich plasma-aggregation assay using light-transmission aggregometry was developed and validated in a series of precision and reproducibility tests. PAR4 antagonist responses to PAR4 AP A-Phe(4-F)-PGWLVKNG (12.5 µM to 100 µM) were subsequently evaluated in this assay in vitro and ex vivo in a human study using BMS-986120, a PAR4 antagonist that entered clinical studies. [ABSTRACT FROM AUTHOR]- Published
- 2022
- Full Text
- View/download PDF
6. Platelets differentially modulate CD4+ Treg activation via GPIIa/IIIb-, fibrinogen-, and PAR4-dependent pathways.
- Author
-
Bock, Matthias, Bergmann, Christian B., Jung, Sonja, Biberthaler, Peter, Heimann, Laura, and Hanschen, Marc
- Abstract
CD4
+ FoxP3+ regulatory T cells (CD4+ Tregs) are known to dampen inflammation following severe trauma. Platelets were shown to augment their posttraumatic activation in burn injury, but the exact mechanisms remain unclear. We hypothesized that platelet activation mechanisms via GPIIb/IIIa, fibrinogen, and PAR4 have an immunological effect and modulate CD4+ Treg activation early after trauma. Therefore, C57Bl/6 N mice were injected with tirofiban (GPIIb/IIIa inhibition), ancrod (fibrinogen splitting enzyme), or tcY-NH2 (selective PAR4 antagonist peptide) before inducing a third-degree burn injury of 25% of the total body surface area. Changes in coagulation, and local and systemic CD4+ Treg activity were assessed via rotational thromboelastometry (ROTEM®) and phospho-flow cytometry 1 h post intervention. The inhibition of GPIIb/IIIa and fibrinogen locally led to a higher basic activity of CD4+ Tregs compared to non-inhibited animals. In contrast, PAR4 disruption on platelets locally led to an increased posttraumatic activation of CD4+ Tregs. Fibrinogen led to complete elimination of coagulation, whereas GPIIb/IIIa or PAR4 inhibition did not. GPIIb/IIIa receptor and fibrinogen inhibition increase CD4+ Tregs activity independently of trauma. Both are crucial for thrombus formation. We suggest platelets trapped in thrombi are unable to interact with CD4+ Tregs but augment their activity when circulating freely. In contrast, PAR4 seems to reduce CD4+ Treg activation following trauma. In summary, GPIIb/IIIa-, PAR4-, and fibrinogen-dependent pathways in platelets modulate CD4+ Treg baseline activity, independently from their hemostatic functionality. PAR4-dependent pathways modulate the posttraumatic interplay of platelets and CD4+ Tregs. [ABSTRACT FROM AUTHOR]- Published
- 2022
- Full Text
- View/download PDF
7. Blocking protease-activated receptor 4 alleviates liver injury induced by brain death.
- Author
-
Fang, Hongbo, Yuan, Zibo, Zhu, Yaohua, Tang, Hongwei, Pang, Chun, Li, Jie, Shi, Jihua, Guo, Wenzhi, and Zhang, Shuijun
- Subjects
- *
BRAIN death , *PROTEASE-activated receptors , *LIVER injuries , *BLOOD platelet activation , *THROMBIN receptors , *BRAIN injuries - Abstract
Brain death (BD) induces a systemic inflammatory response that influences donor liver quality. Protease-activated receptor 4 (PAR4) is a thrombin receptor that mediates platelet activation and is involved in inflammatory and apoptotic processes. Therefore, we investigated the role of PAR4 blockade in liver injury induced by BD and its associated mechanisms. In this study, we constructed a BD rat model and treated rats with TcY-NH2, a selective PAR4 antagonist, to block PAR4 signaling at the onset of BD induction. Our results revealed that PAR4 protein expression increased in the livers of rats with BD. PAR4 blockade alleviated liver injury induced by BD, as indicated by lower serum ALT/AST levels and an improvement in histomorphology. Blood platelet activation and hepatic platelet accumulation in BD rats were reduced by PAR4 blockade. Additionally, PAR4 blockade attenuated the inflammatory response and apoptosis in the livers of BD rats. Moreover, the activation of NF-κB and MAPK pathways induced by BD was inhibited by PAR4 blockade. Thus, our results suggest that PAR4 contributes to liver injury induced by BD by regulating inflammation and apoptosis through the NF-κB and MAPK pathways. Thus, PAR4 blockade may provide a feasible approach to improve the quality of organs from BD donors. • PAR4 protein expression increases in livers after brain death. • PAR4 blockade alleviates liver injury induced by brain death. • PAR4 blockade reduces platelet activation and accumulation induced by brain death. • PAR4 blockade reduces inflammatory response and apoptosis induced by brain death. • PAR4 blockade inhibits NF-κB and MAPK pathway activation induced by brain death. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
8. Mice with Reduced PAR4 Reactivity show Decreased Venous Thrombosis and Platelet Procoagulant Activity.
- Author
-
Knauss EA, Guci J, Luc N, Disharoon D, Huang GH, Gupta AS, and Nieman MT
- Abstract
Background: Hypercoagulation and thrombin generation are major risk factors for venous thrombosis. Sustained thrombin signaling through PAR4 promotes platelet activation, phosphatidylserine exposure, and subsequent thrombin generation. A single-nucleotide polymorphism in PAR4 (rs2227376) changes proline to leucine extracellular loop 3 (P310L), which decreases PAR4 reactivity and is associated with a lower risk for venous thromboembolism (VTE) in a GWAS meta-analysis., Objective: The goal of this study is to determine the mechanism for the association of rs2227376 with reduced risk for VTE in using mice with a homologous mutation (PAR4-P322L)., Methods: Venous thrombosis was examined using our recently generated PAR4-P322L mice in the inferior vena cava stasis and stenosis models. Coagulation and clot stability was measured using rotational thromboelastometry (ROTEM). Thrombin generating potential was measured in platelet-rich plasma. Phosphatidylserine surface expression and platelet-neutrophil aggregates were analyzed using flow cytometry., Results: PAR4
P/L and PAR4L/L had reduced incidence and size of venous clots at 48 hours. PAR4P/L and PAR4L/L platelets had progressively decreased phosphatidylserine in response to thrombin and convulxin, which led to decreased thrombin generation and decreased PAR4-mediated platelet-neutrophil aggregation., Conclusions: The leucine allele in extracellular loop 3, PAR4-322L leads to fewer procoagulant platelets and decreased endogenous thrombin potential. This decreased ability to generate thrombin offers a mechanism for PAR4's role in VTE highlighting a key role for PAR4 signaling., Competing Interests: Authors conflict of interest statement: Elizabeth A. Knauss, Johana Guci, Norman Luc, Dante Disharoon, Grace H. Huang, Anirban Sen Gupta, and Marvin T. Nieman all declare no conflicts of interest.- Published
- 2024
- Full Text
- View/download PDF
9. Platelet Activation in Venous Thrombosis is Driven by Protease Activated Receptor 4 (PAR4)
- Author
-
Knauss, Elizabeth Ann
- Subjects
- Pharmacology, Biomedical Research, Blood platelets, protease-activated receptors, protease-activated receptor 4, venous thrombosis, thrombosis, hemostasis
- Abstract
Platelets are traditionally understood for their role in responding to vascular damage and forming platelet plugs to block blood loss. Platelets adhere to the vasculature, activate, and provide a surface for coagulation factors to generate thrombin. Hypercoagulation and thrombin generation are major risk factors for pathological conditions like venous thrombosis. Sustained thrombin signaling through protease-activated receptor 4 (PAR4) promotes platelet activation, phosphatidylserine (PS) exposure, and procoagulant activity. A single-nucleotide polymorphism (SNP, rs2227376) leads to an amino acid change in extracellular loop 3 (ECL3) of PAR4, which decreases PAR4 activation (PAR4-P310L). We aim to determine the mechanism by which PAR4-P310L (rs2227376) impacts platelet function and reduces the risk of thrombosis using mice with a homologous mutation (PAR4-P322L). Here, we use these mice to model reduced PAR4 function, and characterize how this translates to platelet biology and thrombosis. Platelets from PAR4-322P/L and PAR4-322L/L mice had a significantly reduced response to PAR4 activation, measured by granule release and integrin activation. PAR4-P322L mice also had an extended time to arterial thrombosis. In humans this SNP was associated with a 15% risk reduction for venous thromboembolism (VTE) in a GWAS meta-analysis. We extended our study to investigate how PAR4-P322L specifically impairs platelet activity venous thrombosis. Indeed, PAR4-322P/L mice had significantly smaller venous clots induced by complete ligation of the inferior vena cava (IVC) after 48 hours. PAR4-/-mice were unable to develop venous thrombi by 24 hours. Both PAR4-322P/L and 322L/L mice had significantly smaller thrombi 48 hours after partial IVC stenosis. The PAR4-P322L mutation also negatively impacted platelet-mediated thrombin generation and PS exposure, as well as platelet-neutrophil aggregation. Importantly, whole blood from these mice did not show impaired clot stability in ROTEM. In conclusion, I have shown here that the PAR4-P322L mutation negatively impacts PAR4-mediated platelet activation in arterial and venous thrombosis without significantly impairing normal hemostasis. This adds to growing evidence that PAR4 antagonists would be effective antithrombotic therapy in the clinic. Our mouse model also provides a foundation to further evaluate the impact of reducing PAR4 function in other pathophysiological contexts.
- Published
- 2024
10. Protease‐activated receptor‐mediated platelet aggregation in acute coronary syndrome patients on potent P2Y12 inhibitors
- Author
-
Patricia P. Wadowski, Joseph Pultar, Constantin Weikert, Beate Eichelberger, Benjamin Panzer, Kurt Huber, Irene M. Lang, Renate Koppensteiner, Simon Panzer, and Thomas Gremmel
- Subjects
antiplatelet therapy ,protease‐activated receptor 1 ,protease‐activated receptor 4 ,prasugrel ,ticagrelor ,platelet aggregation ,Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Abstract Background Despite the increasing use of potent P2Y12 inhibitors, further atherothrombotic events still impair the prognosis of many acute coronary syndrome (ACS) patients. This may in part be attributable to intact platelet aggregation via the human thrombin receptors protease‐activated receptor (PAR)‐1 and PAR‐4. Objective We studied PAR mediated platelet aggregation in ACS patients following percutaneous coronary intervention (PCI) with stent implantation in a cross‐sectional study. Methods Platelet aggregation to ADP as well as to the PAR‐1 agonist SFLLRN and the PAR‐4 agonist AYPGKF was assessed by multiple electrode aggregometry in 194 ACS patients on dual antiplatelet therapy with aspirin and either prasugrel (n = 114) or ticagrelor (n = 80) 3 days after PCI. Results Based on the consensus cutoff value, high on‐treatment residual platelet reactivity to ADP (HRPR ADP) was observed in only 2 prasugrel‐treated patients. Both patients with HRPR ADP had also a normal response to SFLLRN and AYPGKF. Among the 112 prasugrel‐treated patients with adequate P2Y12 inhibition, 50 patients (45%) still had a normal response to SFLLRN, and 70 patients (63%) still had a normal response to AYPGKF. Among the 80 ticagrelor‐treated patients with adequate P2Y12 inhibition, 25 patients (31%) still had a normal response to SFLLRN, and 50 (63%) still had a normal response to AYPGKF. Conclusion Normal platelet aggregation via PAR‐1 and PAR‐4 is preserved in many patients with adequate P2Y12 inhibition by prasugrel and ticagrelor. The present findings may at least in part explain adverse ischemic events despite potent P2Y12 inhibition.
- Published
- 2019
- Full Text
- View/download PDF
11. A mouse model of the protease-activated receptor 4 Pro310Leu variant has reduced platelet reactivity.
- Author
-
Han X, Knauss EA, Fuente M, Li W, Conlon RA, LePage DF, Jiang W, Renna SA, McKenzie SE, and Nieman MT
- Subjects
- Animals, Disease Models, Animal, Crotalid Venoms pharmacology, Crotalid Venoms toxicity, Adenosine Diphosphate metabolism, Adenosine Diphosphate pharmacology, P-Selectin metabolism, P-Selectin genetics, Point Mutation, Gene Knock-In Techniques, Signal Transduction, Thrombosis genetics, Thrombosis blood, Male, Chlorides, Mice, Platelet Activation, CRISPR-Cas Systems, Humans, Phenotype, Ferric Compounds, Oligopeptides, Lectins, C-Type, Receptors, Proteinase-Activated, Blood Platelets metabolism, Receptors, Thrombin genetics, Receptors, Thrombin metabolism, Thrombin metabolism, Platelet Aggregation, Platelet Glycoprotein GPIIb-IIIa Complex metabolism, Platelet Glycoprotein GPIIb-IIIa Complex genetics, Mice, Inbred C57BL
- Abstract
Background: Protease-activated receptor 4 (PAR4) mediates thrombin signaling on platelets and other cells. Our recent structural studies demonstrated that a single nucleotide polymorphism in extracellular loop 3 and PAR4-P310L (rs2227376) leads to a hyporeactive receptor., Objectives: The goal of this study was to determine how the hyporeactive PAR4 variant in extracellular loop 3 impacts platelet function in vivo using a novel knock-in mouse model (PAR4-322L)., Methods: A point mutation was introduced into the PAR4 gene F2rl3 via CRISPR/Cas9 to create PAR4-P322L, the mouse homolog to human PAR4-P310L. Platelet response to PAR4 activation peptide (AYPGKF), thrombin, ADP, and convulxin was monitored by αIIbβ3 integrin activation and P-selectin translocation using flow cytometry or platelet aggregation. In vivo responses were determined by the tail bleeding assay and the ferric chloride-induced carotid artery injury model., Results: PAR4-P/L and PAR4-L/L platelets had a reduced response to AYPGKF and thrombin measured by P-selectin translocation or αIIbβ3 activation. The response to ADP and convulxin was unchanged among genotypes. In addition, both PAR4-P/L and PAR4-L/L platelets showed a reduced response to thrombin in aggregation studies. There was an increase in the tail bleeding time for PAR4-L/L mice. The PAR4-P/L and PAR4-L/L mice both showed an extended time to arterial thrombosis., Conclusion: PAR4-322L significantly reduced platelet responsiveness to AYPGKF and thrombin, which is in agreement with our previous structural and cell signaling studies. In addition, PAR4-322L had prolonged arterial thrombosis time. Our mouse model provides a foundation to further evaluate the role of PAR4 in other pathophysiological contexts., Competing Interests: Declaration of competing interests There are no competing interests to disclose, (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
12. Protease-activated receptor 4 activity promotes platelet granule release and platelet-leukocyte interactions
- Author
-
Rachel A. Rigg, Laura D. Healy, Tiffany T. Chu, Anh T. P. Ngo, Annachiara Mitrugno, Jevgenia Zilberman-Rudenko, Joseph E. Aslan, Monica T. Hinds, Lisa Dirling Vecchiarelli, Terry K. Morgan, András Gruber, Kayla J. Temple, Craig W. Lindsley, Matthew T. Duvernay, Heidi E. Hamm, and Owen J. T. McCarty
- Subjects
par4 ,platelet-leukocyte interactions ,platelet dense granule release ,platelet activation ,protease-activated receptor 4 ,granulocytes ,Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Human platelets express two protease-activated receptors (PARs), PAR1 (F2R) and PAR4 (F2RL3), which are activated by a number of serine proteases that are generated during pathological events and cause platelet activation. Recent interest has focused on PAR4 as a therapeutic target, given PAR4 seems to promote experimental thrombosis and procoagulant microparticle formation, without a broadly apparent role in hemostasis. However, it is not yet known whether PAR4 activity plays a role in platelet-leukocyte interactions, which are thought to contribute to both thrombosis and acute or chronic thrombo-inflammatory processes. We sought to determine whether PAR4 activity contributes to granule secretion from activated platelets and platelet-leukocyte interactions. We performed in vitro and ex vivo studies of platelet granule release and platelet-leukocyte interactions in the presence of PAR4 agonists including PAR4 activating peptide, thrombin, cathepsin G, and plasmin in combination with small-molecule PAR4 antagonists. Activation of human platelets with thrombin, cathepsin G, or plasmin potentiated platelet dense granule secretion that was specifically impaired by PAR4 inhibitors. Platelet-leukocyte interactions and platelet P-selectin exposure the following stimulation with PAR4 agonists were also impaired by activated PAR4 inhibition in either a purified system or in whole blood. These results indicate PAR4-specific promotion of platelet granule release and platelet-leukocyte aggregate formation and suggest that pharmacological control of PAR4 activity could potentially attenuate platelet granule release or platelet-leukocyte interaction-mediated pathological processes.
- Published
- 2019
- Full Text
- View/download PDF
13. Loss of Protease-Activated Receptor 4 Prevents Inflammation Resolution and Predisposes the Heart to Cardiac Rupture After Myocardial Infarction.
- Author
-
Kolpakov, Mikhail A., Guo, Xinji, Rafiq, Khadija, Vlasenko, Liudmila, Hooshdaran, Bahman, Seqqat, Rachid, Wang, Tao, Fan, Xiaoxuan, Tilley, Douglas G., Kostyak, John C., Kunapuli, Satya P., Houser, Steven R., and Sabri, Abdelkarim
- Subjects
- *
PROTEASE-activated receptors , *MYOCARDIAL infarction , *HEART diseases , *INFLAMMATION , *APOPTOSIS , *THROMBOLYTIC therapy , *INFLAMMATION prevention , *HEART metabolism , *RESEARCH , *ANIMAL experimentation , *RESEARCH methodology , *HEART rupture , *CELL receptors , *MEDICAL cooperation , *EVALUATION research , *COMPARATIVE studies , *GENES , *RESEARCH funding , *MICE - Abstract
Background: Cardiac rupture is a major lethal complication of acute myocardial infarction (MI). Despite significant advances in reperfusion strategies, mortality from cardiac rupture remains high. Studies suggest that cardiac rupture can be accelerated by thrombolytic therapy, but the relevance of this risk factor remains controversial.Methods: We analyzed protease-activated receptor 4 (Par4) expression in mouse hearts with MI and investigated the effects of Par4 deletion on cardiac remodeling and function after MI by echocardiography, quantitative immunohistochemistry, and flow cytometry.Results: Par4 mRNA and protein levels were increased in mouse hearts after MI and in isolated cardiomyocytes in response to hypertrophic and inflammatory stimuli. Par4-deficient mice showed less myocyte apoptosis, reduced infarct size, and improved functional recovery after acute MI relative to wild-type (WT). Conversely, Par4-/- mice showed impaired cardiac function, greater rates of myocardial rupture, and increased mortality after chronic MI relative to WT. Pathological evaluation of hearts from Par4-/- mice demonstrated a greater infarct expansion, increased cardiac hemorrhage, and delayed neutrophil accumulation, which resulted in impaired post-MI healing compared with WT. Par4 deficiency also attenuated neutrophil apoptosis in vitro and after MI in vivo and impaired inflammation resolution in infarcted myocardium. Transfer of Par4-/- neutrophils, but not of Par4-/- platelets, in WT recipient mice delayed inflammation resolution, increased cardiac hemorrhage, and enhanced cardiac dysfunction. In parallel, adoptive transfer of WT neutrophils into Par4-/- mice restored inflammation resolution, reduced cardiac rupture incidence, and improved cardiac function after MI.Conclusions: These findings reveal essential roles of Par4 in neutrophil apoptosis and inflammation resolution during myocardial healing and point to Par4 inhibition as a potential therapy that should be limited to the acute phases of ischemic insult and avoided for long-term treatment after MI. [ABSTRACT FROM AUTHOR]- Published
- 2020
- Full Text
- View/download PDF
14. Platelets differentially modulate CD4+ Treg activation via GPIIa/IIIb-, fibrinogen-, and PAR4-dependent pathways
- Author
-
Bock, Matthias, Bergmann, Christian B., Jung, Sonja, Biberthaler, Peter, Heimann, Laura, and Hanschen, Marc
- Published
- 2022
- Full Text
- View/download PDF
15. Differential Effects of Platelets Selectively Activated by Protease-Activated Receptors on Meniscal Cells.
- Author
-
Xu, Hongyao, Zou, Xiangjie, Xia, Pengcheng, Aboudi, Mohammad Ahmad Kamal, Chen, Ran, and Huang, He
- Subjects
- *
CELL proliferation , *ANIMAL experimentation , *BLOOD platelets , *BLOOD platelet activation , *CELL receptors , *COLLAGEN , *IMMUNOHISTOCHEMISTRY , *MENISCUS (Anatomy) , *MENISCUS injuries , *MICE , *POLYMERASE chain reaction , *RESEARCH funding , *T-test (Statistics) , *WOUND healing , *PROTEASE inhibitors , *VASCULAR endothelial growth factors , *IN vitro studies - Abstract
Background: Meniscal injury is very common, and injured meniscal tissue has a limited healing ability because of poor vascularity. Platelets contain both pro- and anti-angiogenic factors, which can be released by platelet selective activation. Hypothesis: Platelets release a high level of vascular endothelial growth factor (VEGF) when they are activated by protease-activated receptor 1 (PAR1), whereas the platelets release endostatin when they are activated by protease-activated receptor 4 (PAR4). The PAR1-treated platelets enhance the proliferation of meniscal cells in vitro and promote in vivo healing of wounded meniscal tissue. Study Design: Controlled laboratory study. Method: Platelets were isolated from human blood and activated with different reagents. The released growth factors from the activated platelets were determined by immunostaining and enzyme-linked immunosorbent assay. The effects of the platelets with different treatments on meniscal cells were tested by an in vitro model of cell culture and an in vivo model of wounded meniscal healing. Results: The results indicated that platelets contained both pro- and antiangiogenic factors including VEGF and endostatin. In unactivated platelets, VEGF and endostatin were contained inside of the platelets. Both VEGF and endostatin were released from the platelets when they were activated by thrombin. However, only VEGF was released from the platelets when they were activated by PAR1, and only endostatin was released from the platelets when they were activated by PAR4. The rat meniscal cells grew much faster in the medium that contained PAR1-activated platelets than in the medium that contained either PAR4-activated platelets or unactivated platelets. The wounds treated with PAR1-activated platelets healed faster than those treated with either PAR4-activated platelets or unactivated platelets. Many blood vessel–like structures were found in the wounded menisci treated with PAR1-activated platelets. Conclusion: The PAR1-activated platelets released high levels of VEGF, which increased the proliferation of rat meniscal cells in vitro, enhanced the vascularization of menisci in vivo, and promoted healing of wounded menisci. Clinical Relevance: Our results suggested that selective activated platelets can be used clinically to enhance healing of wounded meniscal tissue. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
16. Antimicrobial Peptide Cathelicidin-BF Inhibits Platelet Aggregation by Blocking Protease-Activated Receptor 4.
- Author
-
Shu, Guofang, Chen, Yahui, Liu, Tongdan, Ren, Shenhong, and Kong, Yi
- Subjects
- *
ANTI-infective agents , *CATHELICIDINS , *BLOOD platelets , *PROTEOLYTIC enzymes , *HEMORRHAGE - Abstract
Cathelicidin-BF (BF-30), a peptide isolated from the snake venom of Bungarus fasciatus, exhibits multiple biological functions, including antimicrobial, anticancer and anti-inflammatory. However, the effect of BF-30 on platelet and thrombus formation was reported rarely. In this study, we investigated the antiplatelet and antithrombotic effects of BF-30 and its underlying mechanism. Our results showed that BF-30 potently inhibited thrombin-induced platelet aggregation, and further specifically blocked protease-activated receptor 4 (PAR4). It also reduced P-selectin expression, AktSer473 phosphorylation and platelet spreading on fibrinogen. Furthermore, BF-30 exhibited potent inhibitory activity on thrombus formation in vivo: it decreased death of mice with acute pulmonary thrombosis and attenuated thrombosis weight in arterio-venous shunt model. Additionally, a tail cutting bleeding time assay revealed that BF-30 did not prolong bleeding time in mice at efficient dosage. Taken together, BF-30 is a PAR4 antagonist, and inhibits thrombus formation without obvious bleeding risk in vivo. We believe that this study may provide a source for the development of PAR4 antagonist for the treatment of thrombotic disorders. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
17. Protease-activated receptor 4 activity promotes platelet granule release and platelet-leukocyte interactions.
- Author
-
Rigg, Rachel A., Chu, Tiffany T., Ngo, Anh T. P., Mitrugno, Annachiara, Zilberman-Rudenko, Jevgenia, Hinds, Monica T., McCarty, Owen J. T., Gruber, András, Healy, Laura D., Aslan, Joseph E., Vecchiarelli, Lisa Dirling, Morgan, Terry K., Duvernay, Matthew T., Hamm, Heidi E., Temple, Kayla J., and Lindsley, Craig W.
- Subjects
THROMBIN receptors ,PROTEASE-activated receptors ,BLOOD platelet activation - Abstract
Human platelets express two protease-activated receptors (PARs), PAR1 (F2R) and PAR4 (F2RL3), which are activated by a number of serine proteases that are generated during pathological events and cause platelet activation. Recent interest has focused on PAR4 as a therapeutic target, given PAR4 seems to promote experimental thrombosis and procoagulant microparticle formation, without a broadly apparent role in hemostasis. However, it is not yet known whether PAR4 activity plays a role in platelet-leukocyte interactions, which are thought to contribute to both thrombosis and acute or chronic thrombo-inflammatory processes. We sought to determine whether PAR4 activity contributes to granule secretion from activated platelets and platelet-leukocyte interactions. We performed in vitro and ex vivo studies of platelet granule release and platelet-leukocyte interactions in the presence of PAR4 agonists including PAR4 activating peptide, thrombin, cathepsin G, and plasmin in combination with small-molecule PAR4 antagonists. Activation of human platelets with thrombin, cathepsin G, or plasmin potentiated platelet dense granule secretion that was specifically impaired by PAR4 inhibitors. Platelet-leukocyte interactions and platelet P-selectin exposure the following stimulation with PAR4 agonists were also impaired by activated PAR4 inhibition in either a purified system or in whole blood. These results indicate PAR4-specific promotion of platelet granule release and platelet-leukocyte aggregate formation and suggest that pharmacological control of PAR4 activity could potentially attenuate platelet granule release or platelet-leukocyte interaction-mediated pathological processes. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
18. PAR4 overexpression promotes colorectal cancer cell proliferation and migration.
- Author
-
Zhang, Hongshan, Jiang, Ping, Zhang, Chuanrao, Lee, Siman, Wang, Wei, and Zou, Hao
- Subjects
- *
G proteins , *CANCER invasiveness , *ANCHORAGE , *LYMPH nodes , *CANCER cell culture - Abstract
Protease-activated receptor 4 (PAR4), a member of the G-protein-coupled receptor family, was previously identified to be involved in the progression of cancer. Previous study revealed that the expression of PAR4 was increased in colorectal cancer tissues compared with the associated normal tissues, particularly in positive lymph node and poorly differentiated types of cancer. We hypothesized that PAR4 serves a function in the progression of colorectal cancer. In the present study, overexpression of PAR4 in colorectal cancer LoVo cells promoted proliferation, anchorage-independent growth and migration. In vivo, PAR4 increased LoVo cell tumorgenicity. In contrast, knockdown of PAR4 in HT-29 cells decreased proliferation, anchorage-independent growth and migration. Mechanistic studies revealed that PAR4 increased the phosphorylation of extracellular-signal-regulated kinase 1/2 in colorectal cancer cells, which is the potential molecular mechanism that promotes cellular proliferation and migration. Taken together, the results of the present study indicated that overexpression of PAR4 promoted colorectal cancer cell proliferation, survival and metastasis, indicating that PAR4 is a promising therapeutic target for preventing colon cancer progression. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
19. Endogenous H2S sensitizes PAR4-induced bladder pain.
- Author
-
Wenfu Wang, Qiyu Bo, Jian Du, Xin Yu, Kejia Zhu, Jianfeng Cui, Hongda Zhao, Yong Wang, Benkang Shi, and Yaofeng Zhu
- Subjects
- *
INTERSTITIAL cystitis , *HYDROGEN sulfide , *CYSTATHIONINE beta-synthase , *CYSTATHIONINE beta-lyase , *ACETIC acid analysis , *THERAPEUTICS - Abstract
Bladder pain is a prominent symptom of interstitial cystitis/painful bladder syndrome. Hydrogen sulfide (H2S) generated by cystathionine γ-synthase (CBS) or cystathionine β-lyase (CSE) facilitates bladder hypersensitivity. We assessed involvement of the H2S pathway in protease-activated receptor 4 (PAR4)-induced bladder pain. A bladder pain model was induced by intravesical instillation of PAR4-activating peptide in mice. The role of H2S in this model was evaluated by intraperitoneal preadministration of D,L-propargylglycine (PAG), aminooxyacetic acid (AOAA), or S-adenosylmethionine or the preintravesical administration of NaHS. SV-HUC-1 cells were treated in similar manners. Assessments of CBS, CSE, and macrophage migration inhibitory factor (MIF) expression, bladder voiding function, bladder inflammation, H2S production, and referred bladder pain were performed. The CSE and CBS pathways existed in both mouse bladders and SV-HUC-1 cells. H2S signaling was upregulated in PAR4-induced bladder pain models, and H2S-generating enzyme activity was upregulated in human bladders, mouse bladders, and SV-HUC-1 cells. Pretreatment with AOAA or NaHS inhibited or promoted PAR4-induced mechanical hyperalgesia, respectively; however, PAG only partially inhibited PAR4-induced bladder pain. Treatment with PAG or AOAA decreased H2S production in both mouse bladders and SV-HUC-1 cells. Pretreatment with AOAA increased MIF protein levels in bladder tissues and cells, whereas pretreatment with NaHS lowered MIF protein levels. Bladder pain triggered by the H2S pathway was not accompanied by inflammation or altered micturition behavior. Thus endogenous H2S generated by CBS or CSE caused referred hyperalgesia mediated through MIF in mice with PAR4-induced bladder pain, without causing bladder injury or altering micturition behavior. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
20. Protease-activated receptor 4 causes Akt phosphorylation independently of PI3 kinase pathways
- Author
-
Satya P. Kunapuli and Carol Dangelmaier
- Subjects
0301 basic medicine ,Platelet aggregation ,030204 cardiovascular system & hematology ,Article ,Mice ,Phosphatidylinositol 3-Kinases ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Animals ,Humans ,LY294002 ,Platelet ,Platelet activation ,Phosphorylation ,Pi 3 kinase ,Kinase ,Hematology ,General Medicine ,Cell biology ,Disease Models, Animal ,030104 developmental biology ,chemistry ,PROTEASE-ACTIVATED RECEPTOR 4 ,Receptors, Thrombin ,Akt phosphorylation ,Proto-Oncogene Proteins c-akt - Abstract
PI-3 Kinase plays an important role in platelet activation mainly through regulation of RASA3. Akt phosphorylation is an indicator for the activity of PI3 kinase. The aim of this study is to characterize the pathways leading to Akt phosphorylation in platelets. We performed concentration response curves of LY294002, a pan-PI3 kinase inhibitor, on platelet aggregation and Akt phosphorylation, in washed human and mouse platelets. At concentrations as low as 3.12 μM, LY294002 abolished Akt phosphorylation induced by 2MeSADP and SFLLRN, but not by AYPGKF. It required much higher concentrations of LY294002 (12.5-25 μM) to abolish AYPGKF-induced Akt phosphorylation, both in wild type and P2Y12 null mouse platelets. We propose that 3.12 μM LY294002 is sufficient to inhibit PI3 kinase isoforms in platelets and higher concentrations might inhibit other pathways regulating Akt phosphorylation by AYPGKF. We conclude that Protease-activated receptor 4 (PAR4) might cause Akt phosphorylation through pathways distinctly different from those of Protease-activated receptor 1 (PAR1).
- Published
- 2020
- Full Text
- View/download PDF
21. Protease-Activated Receptor 4: A Critical Participator in Inflammatory Response.
- Author
-
Fu, Qiang, Cheng, Jing, Gao, Yebo, Zhang, Yonglei, Chen, Xiaobing, and Xie, Jianguo
- Subjects
- *
PROTEASE-activated receptors , *INFLAMMATION , *IMMUNE response , *PROTEOLYTIC enzymes , *LIGANDS (Biochemistry) , *HEMOSTASIS , *THROMBOSIS - Abstract
Protease-activated receptors (PARs) are G protein-coupled receptors of which four members PAR1, PAR2, PAR3, and PAR4 have been identified, characterized by a typical mechanism of activation involving various related proteases. The amino-terminal sequence of PARs is cleaved by a broad array of proteases, leading to specific proteolytic cleavage which forms endogenous tethered ligands to induce agonist-biased PAR activation. The biological effect of PARs activated by coagulation proteases to regulate hemostasis and thrombosis plays an enormous role in the cardiovascular system, while PAR4 can also be activated by trypsin, cathepsin G, the activated factor X of the coagulation cascade, and trypsin IV. Irrespective of its role in thrombin-induced platelet aggregation, PAR4 activation is believed to be involved in inflammatory lesions, as show by investigations that have unmasked the effects of PAR4 on neutrophil recruitment, the regulation of edema, and plasma extravasation. This review summarizes the roles of PAR4 in coagulation and other extracellular protease pathways, which activate PAR4 to participate in normal regulation and disease. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
22. Platelets differentially modulate CD4+ Treg activation via GPIIa/IIIb-, fibrinogen-, and PAR4-dependent pathways
- Author
-
Matthias Bock, Christian B. Bergmann, Sonja Jung, Peter Biberthaler, Laura Heimann, and Marc Hanschen
- Subjects
Immunology ,hemic and immune systems ,Original Article ,Thrombocytes ,CD4 ,Trauma ,Glycoprotein IIb/IIIa ,α ,Fibrinogen ,Protease-activated receptor 4 ,ddc - Abstract
CD4+FoxP3+ regulatory T cells (CD4+ Tregs) are known to dampen inflammation following severe trauma. Platelets were shown to augment their posttraumatic activation in burn injury, but the exact mechanisms remain unclear. We hypothesized that platelet activation mechanisms via GPIIb/IIIa, fibrinogen, and PAR4 have an immunological effect and modulate CD4+ Treg activation early after trauma. Therefore, C57Bl/6 N mice were injected with tirofiban (GPIIb/IIIa inhibition), ancrod (fibrinogen splitting enzyme), or tcY-NH2 (selective PAR4 antagonist peptide) before inducing a third-degree burn injury of 25% of the total body surface area. Changes in coagulation, and local and systemic CD4+ Treg activity were assessed via rotational thromboelastometry (ROTEM®) and phospho-flow cytometry 1 h post intervention. The inhibition of GPIIb/IIIa and fibrinogen locally led to a higher basic activity of CD4+ Tregs compared to non-inhibited animals. In contrast, PAR4 disruption on platelets locally led to an increased posttraumatic activation of CD4+ Tregs. Fibrinogen led to complete elimination of coagulation, whereas GPIIb/IIIa or PAR4 inhibition did not. GPIIb/IIIa receptor and fibrinogen inhibition increase CD4+ Tregs activity independently of trauma. Both are crucial for thrombus formation. We suggest platelets trapped in thrombi are unable to interact with CD4+ Tregs but augment their activity when circulating freely. In contrast, PAR4 seems to reduce CD4+ Treg activation following trauma. In summary, GPIIb/IIIa-, PAR4-, and fibrinogen-dependent pathways in platelets modulate CD4+ Treg baseline activity, independently from their hemostatic functionality. PAR4-dependent pathways modulate the posttraumatic interplay of platelets and CD4+ Tregs.
- Published
- 2020
23. Loss of Protease-Activated Receptor 4 Prevents Inflammation Resolution and Predisposes the Heart to Cardiac Rupture after Myocardial Infarction
- Author
-
Satya P. Kunapuli, Douglas G. Tilley, Rachid Seqqat, Liudmila Vlasenko, Mikhail A. Kolpakov, Bahman Hooshdaran, Xinji Guo, Tao Wang, Xiaoxuan Fan, Abdelkarim Sabri, John C. Kostyak, Khadija Rafiq, and Steven R. Houser
- Subjects
Male ,medicine.medical_specialty ,Heart Rupture ,Myocardial Infarction ,Inflammation ,030204 cardiovascular system & hematology ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Inflammation resolution ,Physiology (medical) ,Internal medicine ,medicine ,Animals ,Myocardial infarction ,030304 developmental biology ,Mice, Knockout ,0303 health sciences ,business.industry ,Myocardium ,Cardiac Rupture ,medicine.disease ,Gene Expression Regulation ,Cardiology ,PROTEASE-ACTIVATED RECEPTOR 4 ,Female ,Receptors, Thrombin ,medicine.symptom ,Cardiology and Cardiovascular Medicine ,business ,Complication - Abstract
Background: Cardiac rupture is a major lethal complication of acute myocardial infarction (MI). Despite significant advances in reperfusion strategies, mortality from cardiac rupture remains high. Studies suggest that cardiac rupture can be accelerated by thrombolytic therapy, but the relevance of this risk factor remains controversial. Methods: We analyzed protease-activated receptor 4 (Par4) expression in mouse hearts with MI and investigated the effects of Par4 deletion on cardiac remodeling and function after MI by echocardiography, quantitative immunohistochemistry, and flow cytometry. Results: Par4 mRNA and protein levels were increased in mouse hearts after MI and in isolated cardiomyocytes in response to hypertrophic and inflammatory stimuli. Par4-deficient mice showed less myocyte apoptosis, reduced infarct size, and improved functional recovery after acute MI relative to wild-type (WT). Conversely, Par4 −/− mice showed impaired cardiac function, greater rates of myocardial rupture, and increased mortality after chronic MI relative to WT. Pathological evaluation of hearts from Par4 −/− mice demonstrated a greater infarct expansion, increased cardiac hemorrhage, and delayed neutrophil accumulation, which resulted in impaired post-MI healing compared with WT. Par4 deficiency also attenuated neutrophil apoptosis in vitro and after MI in vivo and impaired inflammation resolution in infarcted myocardium. Transfer of Par4 −/− neutrophils, but not of Par4 −/− platelets, in WT recipient mice delayed inflammation resolution, increased cardiac hemorrhage, and enhanced cardiac dysfunction. In parallel, adoptive transfer of WT neutrophils into Par4 −/− mice restored inflammation resolution, reduced cardiac rupture incidence, and improved cardiac function after MI. Conclusions: These findings reveal essential roles of Par4 in neutrophil apoptosis and inflammation resolution during myocardial healing and point to Par4 inhibition as a potential therapy that should be limited to the acute phases of ischemic insult and avoided for long-term treatment after MI.
- Published
- 2020
24. Discovery of 7, 4′-dimethoxy-3-hydroxyflavone as a protease-activated receptor 4 antagonist with antithrombotic activity and less bleeding tendency in mice.
- Author
-
Lin, Ying-Ting, Li, Yu, Hsu, Hui-Ching, Tsai, Ju-Ying, Lee, Jia-Hau, Tai, Chi-Jung, Wu, Ming-Jung, and Wu, Chin-Chung
- Subjects
- *
PROTEASE-activated receptors , *BLOOD platelet aggregation , *MITOGEN-activated protein kinases , *CHEMICAL inhibitors , *STRUCTURE-activity relationships , *ARTERIAL occlusions , *THROMBIN receptors - Abstract
[Display omitted] There is growing evidence of the importance of protease-activated receptor 4 (PAR4), one of thrombin receptors, as a therapeutic target in thrombotic cardiovascular diseases. In the present study, we utilized ligand-based virtual screening, bioassay, and structure–activity relationship study to discover PAR4 antagonists with new chemical scaffolds from natural origin, and examined their application as antiplatelet agents. By using these approaches, we have identified a flavonoid, 7, 4′-dimethoxy-3-hydroxyflavone, that exhibits anti-PAR4 activity. 7, 4′-Dimethoxy-3-hydroxyflavone inhibited PAR4-mediated human platelet aggregation, GPIIb/IIIa activation, and P-selectin secretion. Also, it inhibited PAR4 downstream signaling pathways, including Ca2+/protein kinase C, Akt, and MAP kinases ERK and p38, in human platelets, and suppressed PAR4-mediated β-arrestin recruitment in CHO-K1 cells exogenously expressed human PAR4. In a microfluidic system, 7, 4′-dimethoxy-3-hydroxyflavone reduced thrombus formation on collagen-coated chambers at an arterial shear rate in recalcified whole blood. Furthermore, mice treated with 7, 4′-dimethoxy-3-hydroxyflavone were significantly protected from FeCl 3 -induced carotid arterial occlusions, without significantly affecting tail bleeding time. In conclusion, 7, 4′-dimethoxy-3-hydroxyflavone represents a new class of nature-based PAR4 antagonist, it shows effective in vivo antithrombotic properties with less bleeding tendency, and could be a potential candidate for developing new antiplatelet agents. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
25. The Expression of Protease-Activated Receptor 2 and 4 in the Colon of Irritable Bowel Syndrome Patients.
- Author
-
Zhao, Ju-hui, Dong, Lei, Shi, Hai-tao, Wang, Zong-yan, Shi, Hong-yang, and Ding, Hui
- Subjects
- *
PROTEASE-activated receptors , *INTESTINAL diseases , *PATHOLOGICAL physiology , *POLYMERASE chain reaction , *PROTEIN genetics , *ACTIN , *IMMUNOHISTOCHEMISTRY , *IRRITABLE colon , *PATIENTS - Abstract
Background: Irritable bowel syndrome (IBS) pathophysiology has not been fully understood. Abnormalities of serine proteases have been identified in IBS patients. In addition, protease-activated receptors (PAR) activation interferes with several components of the pathogenesis of IBS, so, evaluating the PAR expression in IBS patients may contribute to understanding the pathogenesis of the disease. Aims: This study aimed to investigate whether the expression of PAR4 and PAR2 in the colon was changed in IBS patients and was associated with IBS. Methods: Colon mucosal biopsies of 34 IBS patients (16 constipation- and 18 diarrhea-predominant) and 18 control subjects were collected. Gene transcripts of PAR2, PAR4, tryptase, and trypsin were quantified using real-time polymerase chain reaction (PCR) and the expression of PAR2 and PAR4 receptors was also measured by immunohistology and image analysis. Results: In IBS patients, the mRNA expression of tryptase and trypsin normalized against β-actin gene was higher compared to control subjects ( P < 0.001). No difference was observed in the PAR2 mRNA level or protein level between control subjects on the one hand and IBS patients or subgroups on the other. In IBS or IBS subgroups patients, the expression of PAR4 in the mRNA level or protein level was lower than the control subjects. Conclusions: This study, for the first time, indicated the PAR4 expression in IBS patients. Decreased PAR4 expression may help us to understand the pathogenesis of IBS. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
26. Deficiency of PAR4 attenuates cerebral ischemia/reperfusion injury in mice.
- Author
-
Yingying Mao, Ming Zhang, Tuma, Ronald F., and Kunapuli, Satya P.
- Subjects
- *
ISCHEMIA , *REPERFUSION , *BRAIN diseases , *LABORATORY mice ,THERAPEUTIC use of fibrinolytic agents - Abstract
Stroke is the third leading cause of death in the USA. Antithrombotic therapy targeting platelet activation is one of the treatments for ischemic stroke. Here we investigate the role of one of the thrombin receptors, protease-activated receptor 4 (PAR4), in a mouse transient middle cerebral artery occlusion (MCAO) model. After a 60 min MCAO and 23 h reperfusion, leukocyte and platelet rolling and adhesion on cerebral venules, blood–brain barrier (BBB) permeability, and cerebral edema were compared in PAR4-deficient mice and wild-type mice. Cerebral infarction volume and neuronal death were also measured. PAR4−/− mice had more than an 80% reduction of infarct volume and significantly improved neurologic and motor function compared with wild-type mice after MCAO. Furthermore, deficiency of PAR4 significantly inhibits the rolling and adhesion of both platelets and leukocytes after MCAO. BBB disruption and cerebral edema were also attenuated in PAR4−/− mice compared with wild-type animals. The results of this investigation indicate that deficiency of PAR4 protects mice from cerebral ischemia/reperfusion (I/R) injury, partially through inhibition of platelet activation and attenuation of microvascular inflammation. [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
27. Platelet PAR1 receptor density—Correlation to platelet activation response and changes in exposure after platelet activation
- Author
-
Ramström, Sofia, Öberg, Karin Vretenbrant, Åkerström, Finn, Enström, Camilla, and Lindahl, Tomas L.
- Subjects
- *
BLOOD platelet activation , *PLATELET activating factor , *PROTEIN analysis , *AMINO acid sequence - Abstract
Abstract: Introduction: A polymorphism (-14 A/T) affecting PAR1 expression on the platelet surface has recently been identified. A two-fold variation in receptor density, which correlated with the platelet response to PAR1-activating peptide (PAR1-AP), has been reported. Materials and methods: We used flow cytometry to measure the correlation between the number of PAR1 receptors and platelet activation. We also measured the changes in receptor exposure after platelet activation with PAR1-AP, ADP, PAR4-AP or a collagen-related peptide (CRP). Results: In our study, the PAR1 receptor number varied almost four-fold, from 547 to 2063 copies/platelet (mean±S.D. 1276±320, n =70). The number of PAR1 receptors on resting platelets correlated to platelet fibrinogen binding and P-selectin expression following platelet activation with PAR1-AP (r 2 =0.30, p <0.01 and r 2 =0.15, p <0.05, respectively, n =36). The correlation was not improved by exclusion of the ADP-component from the PAR1-AP-induced response. We found a trend, but no statistically significant differences in PAR1 receptor number and platelet reactivity between A/A individuals and T/A or T/T individuals. Ex vivo activation with PAR1-AP decreased PAR1 surface exposure to 71±19% of the exposure on resting platelets (mean±S.D., p <0.01, n =19), while activation by ADP, PAR4-AP or CRP significantly increased the exposure, to 151±27%, 120±21% and 138±25%, respectively (n =11, 11 and 10). Conclusions: This study shows a large variation in PAR1 receptor number in healthy individuals, a variation correlated to the platelet activation response. We found a significant reduction in PAR1 surface exposure after adding PAR1-AP, while activation with ADP, PAR4-AP or CRP increased the exposure. [Copyright &y& Elsevier]
- Published
- 2008
- Full Text
- View/download PDF
28. Protease-activated receptor 4-mediated Ca2+ signaling in mouse lung alveolar epithelial cells
- Author
-
Ando, Seijitsu, Otani, Hitomi, Yagi, Yasuhiro, Kawai, Kenzo, Araki, Hiromasa, Nakamura, Tomoyuki, Fukuhara, Shirou, and Inagaki, Chiyoko
- Subjects
- *
EPITHELIAL cells , *PROTEASE inhibitors , *DNA polymerases , *ADENOSINE triphosphatase - Abstract
Abstract: Protease-activated receptor (PAR)-4 is a recently identified low-affinity thrombin receptor that plays a pathophysiological role in many types of tissues including the lung. Here, we showed for the first time that PAR4 mRNA and protein are expressed on primary cultured mouse lung alveolar epithelial cells by reverse transcriptase–polymerase chain reaction (RT–PCR) and immunocytochemical analyses. In a fura 2-AM-loaded single epithelial cell, stimulation with thrombin (1 U/ml) and a PAR4 agonist peptide (AYPGKF-NH2, 1–100 μM) increased intracellular Ca2+ concentration ([Ca2+]i), which consisted of an initial peak phase followed by a slowly decaying delayed phase, while a PAR1 agonist peptide, TFLLR-NH2 (1–100 μM), induced a transient increase in [Ca2+]i. AYPGKF-NH2 (10 μM)-induced [Ca2+]i response was attenuated by a PAR4 antagonist peptide (tcY-NH2), a phospholipase C inhibitor, U-73122 (1–10 μM) or a Ca2+-ATPase inhibitor, thapsigargin (1 μM). Removal of extracellular Ca2+ or an inhibitor of store-operated Ca2+ entry, trans-resveratrol (1 μM) shortened the time to shut off the Ca2+ response without any significant effects on the magnitude of the peak [Ca2+]i. Thus, stimulation of PAR4 appeared to mobilize Ca2+ from intracellular stores in the initial peak response and to enhance Ca2+ entry through the store depletion-operated pathway in the delayed phase. The latter mechanism probably contributed to the longer responsiveness of PAR4 stimulation. [Copyright &y& Elsevier]
- Published
- 2007
- Full Text
- View/download PDF
29. Development of a Series of (1-Benzyl-3-(6-methoxypyrimidin-3-yl)-5-(trifluoromethoxy)-1H-indol-2-yl)methanols as Selective Protease Activated Receptor 4 (PAR4) Antagonists with in Vivo Utility and Activity Against γ-Thrombin
- Author
-
Summer E. Young, Shaun R. Stauffer, Kayla J. Temple, Craig W. Lindsley, Wandong Wen, Matthew T. Duvernay, Jae G. Maeng, Anna L. Blobaum, Wenjun Wu, and Heidi E. Hamm
- Subjects
0301 basic medicine ,Indoles ,Stereochemistry ,030204 cardiovascular system & hematology ,Article ,Structure-Activity Relationship ,03 medical and health sciences ,0302 clinical medicine ,Thrombin ,In vivo ,Drug Discovery ,medicine ,Humans ,Potency ,Enzyme Inhibitors ,IC50 ,Indole test ,Dose-Response Relationship, Drug ,Molecular Structure ,Chemistry ,In vitro ,Pyrimidines ,030104 developmental biology ,cardiovascular system ,Molecular Medicine ,PROTEASE-ACTIVATED RECEPTOR 4 ,Receptors, Thrombin ,Selectivity ,medicine.drug - Abstract
Here, we describe the development of a series of highly selective PAR4 antagonists with nanomolar potency and selectivity versus PAR1, derived from the indole-based 3. Of these, 9j (PAR4 IC50 = 445 nM, PAR1 response IC50 > 30 µM) and 10h (PAR4 IC50 = 179 nM, PAR1 response IC50 > 30 µM) maintained an overall favorable in vitro DMPK profile, encouraging rat/mouse in vivo pharmacokinetics (PK) and activity against γ-thrombin.
- Published
- 2016
- Full Text
- View/download PDF
30. Protease-activated receptor 4: from structure to function and back again
- Author
-
Justin Raymond Hamilton and Shauna L. French
- Subjects
0301 basic medicine ,Pharmacology ,Antagonist ,Disease ,Molecular Pharmacology ,Biology ,Bioinformatics ,03 medical and health sciences ,030104 developmental biology ,medicine ,PROTEASE-ACTIVATED RECEPTOR 4 ,Receptor ,Function (biology) ,Vorapaxar ,medicine.drug ,G protein-coupled receptor - Abstract
Protease-activated receptors are a family of four GPCRs (PAR1-PAR4) with a number of unique attributes. Nearly two and a half decades after the discovery of the first PAR, an antagonist targeting this receptor has been approved for human use. The first-in-class PAR1 antagonist, vorapaxar, was approved for use in the USA in 2014 for the prevention of thrombotic cardiovascular events in patients with a history of myocardial infarction or with peripheral arterial disease. These recent developments indicate the clinical potential of manipulating PAR function. While much work has been aimed at uncovering the function of PAR1 and, to a lesser extent, PAR2, comparatively little is known regarding the pharmacology and physiology of PAR3 and PAR4. Recent studies have begun to develop the pharmacological and genetic tools required to study PAR4 function in detail, and there is now emerging evidence for the function of PAR4 in disease settings. In this review, we detail the discovery, structure, pharmacology, physiological significance and therapeutic potential of PAR4. Linked Articles This article is part of a themed section on Molecular Pharmacology of G Protein-Coupled Receptors. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v173.20/issuetoc.
- Published
- 2016
- Full Text
- View/download PDF
31. Protease activated receptor 4 (PAR4) antagonists: Research progress on small molecules in the field of antiplatelet agents
- Author
-
Roujie Xie, Enmao Wang, Yi Kong, Zhang Weiqi, Duo Yuan, Xiong Zhu, Shanshan Li, and Liu Shangde
- Subjects
Blood Platelets ,Indazoles ,Indoles ,Pharmacology ,01 natural sciences ,Small Molecule Libraries ,03 medical and health sciences ,Thiadiazoles ,Drug Discovery ,Clinical information ,medicine ,Animals ,Humans ,cardiovascular diseases ,Platelet activation ,Thrombus ,030304 developmental biology ,0303 health sciences ,010405 organic chemistry ,Chemistry ,Organic Chemistry ,General Medicine ,Platelet Activation ,medicine.disease ,Small molecule ,0104 chemical sciences ,PROTEASE-ACTIVATED RECEPTOR 4 ,Receptors, Thrombin ,Platelet Aggregation Inhibitors ,Thrombotic complication ,circulatory and respiratory physiology - Abstract
Protease activated receptor 4 (PAR4) is a key target in antiplatelet medication to reduce the risk of heart attack and thrombotic complications in stroke. PAR4 antagonists can prevent harmful and stable thrombus growth while retaining initial thrombus formation by acting on the late diffusion stage of platelet activation, which may provide a safer alternative than other antiplatelet agents. Currently, research on PAR4 antagonists is of increasing interest in the field of antiplatelet agents. This article provides an overview of the discovery and development of small-molecule antagonists of PAR4 as novel antiplatelet agents, including structure-activity relationship (SAR) analysis, progress of structure and bioassay optimization, and the latest structural and/or clinical information of representative small-molecule antagonists of PAR4.
- Published
- 2021
- Full Text
- View/download PDF
32. The Role of Protease‐Activated Receptor ‐4 in Vascular Smooth Muscle Cells
- Author
-
Jamill Jackson, Carla Gardner-Jones, LaKeshia Wright, Evangeline D. Motley, and Kyle Simtion
- Subjects
Vascular smooth muscle ,Chemistry ,Genetics ,PROTEASE-ACTIVATED RECEPTOR 4 ,Molecular Biology ,Biochemistry ,Biotechnology ,Cell biology - Published
- 2020
- Full Text
- View/download PDF
33. Protease-activated receptor-mediated platelet aggregation in acute coronary syndrome patients on potent P2Y
- Author
-
Patricia P, Wadowski, Joseph, Pultar, Constantin, Weikert, Beate, Eichelberger, Benjamin, Panzer, Kurt, Huber, Irene M, Lang, Renate, Koppensteiner, Simon, Panzer, and Thomas, Gremmel
- Subjects
platelet aggregation ,protease‐activated receptor 1 ,Original Article ,protease‐activated receptor 4 ,Original Articles: Thrombosis ,antiplatelet therapy ,prasugrel ,ticagrelor - Abstract
Background Despite the increasing use of potent P2Y12 inhibitors, further atherothrombotic events still impair the prognosis of many acute coronary syndrome (ACS) patients. This may in part be attributable to intact platelet aggregation via the human thrombin receptors protease‐activated receptor (PAR)‐1 and PAR‐4. Objective We studied PAR mediated platelet aggregation in ACS patients following percutaneous coronary intervention (PCI) with stent implantation in a cross‐sectional study. Methods Platelet aggregation to ADP as well as to the PAR‐1 agonist SFLLRN and the PAR‐4 agonist AYPGKF was assessed by multiple electrode aggregometry in 194 ACS patients on dual antiplatelet therapy with aspirin and either prasugrel (n = 114) or ticagrelor (n = 80) 3 days after PCI. Results Based on the consensus cutoff value, high on‐treatment residual platelet reactivity to ADP (HRPR ADP) was observed in only 2 prasugrel‐treated patients. Both patients with HRPR ADP had also a normal response to SFLLRN and AYPGKF. Among the 112 prasugrel‐treated patients with adequate P2Y12 inhibition, 50 patients (45%) still had a normal response to SFLLRN, and 70 patients (63%) still had a normal response to AYPGKF. Among the 80 ticagrelor‐treated patients with adequate P2Y12 inhibition, 25 patients (31%) still had a normal response to SFLLRN, and 50 (63%) still had a normal response to AYPGKF. Conclusion Normal platelet aggregation via PAR‐1 and PAR‐4 is preserved in many patients with adequate P2Y12 inhibition by prasugrel and ticagrelor. The present findings may at least in part explain adverse ischemic events despite potent P2Y12 inhibition.
- Published
- 2018
34. P2502In diabetic rats, long-term thrombin inhibition leads to upregulation of vascular protease-activated receptor 4, increased platelet aggregation, and coronary lipid deposits
- Author
-
D Gheban, A Mărginean, Alina Scridon, D Ghertescu, A Vantu, C Fisca, A Hutanu, Marcel Perian, Razvan Constantin Serban, and L Chinezu
- Subjects
Thrombin ,Downregulation and upregulation ,business.industry ,Medicine ,PROTEASE-ACTIVATED RECEPTOR 4 ,Pharmacology ,Cardiology and Cardiovascular Medicine ,business ,Increased Platelet Aggregation ,medicine.drug - Published
- 2018
- Full Text
- View/download PDF
35. Protease activated receptor 4: a backup receptor or a dark horse as a target in antiplatelet therapy?
- Author
-
Marvin T. Nieman and Xu Han
- Subjects
0301 basic medicine ,Acute coronary syndrome ,business.industry ,Horse ,General Medicine ,Pharmacology ,medicine.disease ,03 medical and health sciences ,Therapeutic approach ,030104 developmental biology ,medicine ,Primary component ,PROTEASE-ACTIVATED RECEPTOR 4 ,Platelet ,cardiovascular diseases ,Thrombus ,Receptor ,business ,circulatory and respiratory physiology - Abstract
Platelets are the primary component of pathological arterial thrombi. As such, antiplatelet therapy is a key strategy for treatment of acute coronary syndrome (ACS). The recent publication by Wong and colleagues earlier this year reported a compound, BMS-986120, that targets protease activated receptor 4 (PAR4) in a reversible manner (1). BMS-986120 effectively inhibits thrombus formation with minimal bleeding complications in their animal model (1). Their paper demonstrates the potential of targeting PAR4 as a promising antiplatelet therapeutic approach (1).
- Published
- 2018
36. Inhibitors of protease-activated receptor 4 (PAR4): a review of recent patents (2013-2021).
- Author
-
Yu X, Li S, Zhu X, and Kong Y
- Subjects
- Animals, Clinical Trials as Topic, Humans, Mice, Platelet Aggregation, Receptors, Thrombin, Thrombin chemistry, Thrombin pharmacology, Patents as Topic, Platelet Aggregation Inhibitors chemistry, Platelet Aggregation Inhibitors pharmacology, Platelet Aggregation Inhibitors therapeutic use
- Abstract
Introduction: Protease-activated receptor 4 (PAR4), belonging to a subfamily of G-protein-coupled receptors (GPCR), is expressed on the surface of Human platelets, and the activation of it can lead to platelets aggregation. Studies demonstrated that PAR4 inhibition protect mice from arterial/arteriolar thrombosis, pulmonary embolism and cerebral infarct, while do not affect the hemostatic responses integrity. Therefore, PAR4 has been a promising target for the development of anti-thrombotic agents., Areas Covered: This review covers recent patents and literature on PAR4 and their application published between 2013 and 2021., Expert Opinion: PAR4 is a promising anti-thrombotic target and PAR4 inhibitors are important biologically active compounds for the treatment of thrombosis. Most the recent patents and literature focus on PAR4 selective inhibitors, and BMS-986120 and BMS-986141, which were developed by BMS, have entered clinical trials. With the deep understanding of the crystal structures and biological functions of PAR4, we believe that many other novel types of molecules targeting PAR4 would enter the clinical studies or the market.
- Published
- 2022
- Full Text
- View/download PDF
37. Genetic deletion of platelet PAR4 results in reduced thrombosis and impaired hemostatic plug stability.
- Author
-
Lee RH, Kawano T, Grover SP, Bharathi V, Martinez D, Cowley DO, Mackman N, Bergmeier W, and Antoniak S
- Subjects
- Animals, Blood Platelets, Hemostasis, Mice, Platelet Activation physiology, Platelet Aggregation, Receptors, Thrombin genetics, Thrombin, Hemostatics, Thrombosis genetics
- Abstract
Background: Protease-activated receptor 4 (PAR4) is expressed by a wide variety of cells, including megakaryocytes/platelets, immune cells, cardiomyocytes, and lung epithelial cells. It is the only functional thrombin receptor on murine platelets. A global deficiency of PAR4 is associated with impaired hemostasis and reduced thrombosis., Objective: We aimed to generate a mouse line with a megakaryocyte/platelet-specific deletion of PAR4 (PAR4
fl/fl ;PF4Cre+ ) and use the mouse line to investigate the role of platelet PAR4 in hemostasis and thrombosis in mice., Methods: Platelets from PAR4fl/fl ;PF4Cre+ were characterized in vitro. Arterial and venous thrombosis was analyzed. Hemostatic plug formation was analyzed using a saphenous vein laser injury model in mice with global or megakaryocyte/platelet-specific deletion of PAR4 or wild-type mice treated with thrombin or glycoprotein VI (GPVI) inhibitors., Results: PAR4fl/fl ;PF4Cre+ platelets were unresponsive to thrombin or specific PAR4 stimulation but not to other agonists. PAR4-/- and PAR4fl/fl ;PF4Cre+ mice both exhibited a similar reduction in arterial thrombosis compared to their respective controls. More importantly, we show for the first time that platelet PAR4 is critical for venous thrombosis in mice. In addition, PAR4-/- mice and PAR4fl/fl ;PF4Cre+ mice exhibited a similar impairment in hemostatic plug stability in a saphenous vein laser injury model. Inhibition of thrombin in wild-type mice gave a similar phenotype. Combined PAR4 deficiency on platelets with GPVI inhibition did not impair hemostatic plug formation but further reduced plug stability., Conclusion: We generated a novel PAR4fl/fl ;PF4Cre+ mouse line. We used this mouse line to show that PAR4 signaling in platelets is critical for arterial and venous thrombosis and hemostatic plug stability., (© 2021 International Society on Thrombosis and Haemostasis.)- Published
- 2022
- Full Text
- View/download PDF
38. PAR4 (Protease-Activated Receptor 4) Antagonism With BMS-986120 Inhibits Human Ex Vivo Thrombus Formation
- Author
-
Shea P. Connell, Michael Cerra, Xuewen Ma, Jennifer Raftis, Zhaoqing Wang, Jing Yang, Fraz A. Ismat, Simon Wilson, Samira Garonzik, Hafid Narayan, Timothy J. Gray, and David E. Newby
- Subjects
0301 basic medicine ,Male ,Time Factors ,Platelet Aggregation ,Administration, Oral ,030204 cardiovascular system & hematology ,Pharmacology ,antiplatelet ,0302 clinical medicine ,Translational Sciences ,Prospective Studies ,Receptor ,Chemistry ,Imidazoles ,Thrombosis ,Healthy Volunteers ,Clopidogrel ,Treatment Outcome ,ComputingMethodologies_DOCUMENTANDTEXTPROCESSING ,PROTEASE-ACTIVATED RECEPTOR 4 ,Female ,Cardiology and Cardiovascular Medicine ,Signal Transduction ,Adult ,Blood Platelets ,Morpholines ,Article ,03 medical and health sciences ,Young Adult ,Fibrinolytic Agents ,Journal Article ,medicine ,Humans ,human ,Thrombus ,Benzofurans ,novel ,Aspirin ,Antagonist ,medicine.disease ,stomatognathic diseases ,Thiazoles ,030104 developmental biology ,Scotland ,protease-activated receptor 4 ,Receptors, Thrombin ,Antagonism ,Ex vivo ,Platelet Aggregation Inhibitors - Abstract
Supplemental Digital Content is available in the text., Objective— BMS-986120 is a novel first-in-class oral PAR4 (protease-activated receptor 4) antagonist with potent and selective antiplatelet effects. We sought to determine for the first time, the effect of BMS-986120 on human ex vivo thrombus formation. Approach and Results— Forty healthy volunteers completed a phase 1 parallel-group PROBE trial (Prospective Randomized Open-Label Blinded End Point). Ex vivo platelet activation, platelet aggregation, and thrombus formation were measured at 0, 2, and 24 hours after (1) oral BMS-986120 (60 mg) or (2) oral aspirin (600 mg) followed at 18 hours with oral aspirin (600 mg) and oral clopidogrel (600 mg). BMS-986120 demonstrated highly selective and reversible inhibition of PAR4 agonist peptide (100 μM)-stimulated P-selectin expression, platelet-monocyte aggregates, and platelet aggregation (P
- Published
- 2017
- Full Text
- View/download PDF
39. Substituted indoles as selective protease activated receptor 4 (PAR-4) antagonists: Discovery and SAR of ML354
- Author
-
Charles W. Locuson, Summer E. Young, Julie L. Engers, Wenjun Wu, Kellie D. Nance, Wandong Wen, Bruce J. Melancon, J. Scott Daniels, Matthew T. Duvernay, Michael L. Schulte, Michael R. Wood, Shaun R. Stauffer, Craig W. Lindsley, and Heidi E. Hamm
- Subjects
Indoles ,Stereochemistry ,medicine.medical_treatment ,Clinical Biochemistry ,Pharmaceutical Science ,Biochemistry ,Article ,Structure-Activity Relationship ,Drug Discovery ,medicine ,Humans ,Molecule ,Molecular Biology ,IC50 ,Indole test ,Protease ,Dose-Response Relationship, Drug ,Molecular Structure ,Chemistry ,Organic Chemistry ,Antagonist ,Combinatorial chemistry ,Fluorescence ,Molecular Medicine ,PROTEASE-ACTIVATED RECEPTOR 4 ,Apoptosis Regulatory Proteins ,Selectivity - Abstract
Herein we report the discovery and SAR of an indole-based protease activated receptor-4 (PAR-4) antagonist scaffold derived from a similarity search of the Vanderbilt HTS collection, leading to MLPCN probe ML354 (VU0099704). Using a novel PAC-1 fluorescent αIIbβ3 activation assay this probe molecule antagonist was found to have an IC50 of 140 nM for PAR-4 with 71-fold selectivity versus PAR-1 (PAR-1IC50 = 10 μM).
- Published
- 2014
- Full Text
- View/download PDF
40. Protease-activated receptor 4 is more important than protease-activated receptor 1 for the thrombin-induced procoagulant effect on platelets
- Author
-
Sofia Ramström, Ankit S. Macwan, and Tomas L. Lindahl
- Subjects
0301 basic medicine ,Blood Platelets ,Chemistry ,Thrombin ,Hematology ,030204 cardiovascular system & hematology ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Protease-Activated Receptor 1 ,medicine ,PROTEASE-ACTIVATED RECEPTOR 4 ,Humans ,Protease-activated receptor ,Platelet ,Receptor, PAR-1 ,Receptors, Thrombin ,Receptor ,Coagulation factor II receptor ,medicine.drug - Abstract
Protease-activated receptor 4 is more important than protease-activated receptor 1 for the thrombin-induced procoagulant effect on platelets
- Published
- 2016
41. MP28-14 PROTEASE-ACTIVATED RECEPTOR 4 INDUCES BLADDER PAIN THROUGH DISULFIDE HIGH MOBILITY GROUP BOX-1 ACTING ON RECEPTORS FOR ADVANCED GLYCATION ENDPRODUCTS
- Author
-
Katherine L. Meyer-Siegler, Pedro L. Vera, Fei Ma, Dimitrios E. Kouzoukas, Karin N. Westlund, and David E. Hunt
- Subjects
medicine.medical_specialty ,business.industry ,Urology ,Disulfide bond ,Pharmacology ,Advanced Glycation Endproducts ,Endocrinology ,High-mobility group ,Internal medicine ,Medicine ,PROTEASE-ACTIVATED RECEPTOR 4 ,Bladder Pain ,business ,Receptor - Published
- 2016
- Full Text
- View/download PDF
42. Modulation of Dendritic Cell Function by Protease Activated Receptor 4
- Author
-
Anthony Dorling, Robert I. Lechler, Qi Peng, Daniel McCluskey, and Giovanna Lombardi
- Subjects
Transplantation ,Chemistry ,Modulation ,PROTEASE-ACTIVATED RECEPTOR 4 ,Dendritic cell ,Function (biology) ,Cell biology - Published
- 2018
- Full Text
- View/download PDF
43. Activation of protease-activated receptor-4 inhibits the intrinsic excitability of colonic dorsal root ganglia neurons
- Author
-
R. Karanjia, Ian Spreadbury, Melanie E. Tsang, Francisco Bautista-Cruz, and Stephen Vanner
- Subjects
Male ,Dorsum ,Patch-Clamp Techniques ,Colon ,Physiology ,Action Potentials ,Pain ,Mice ,Ganglia, Spinal ,Animals ,Protease-activated receptor ,Neurons, Afferent ,Receptor ,Endocrine and Autonomic Systems ,Chemistry ,Gastroenterology ,Cell biology ,Rheobase ,Nociception ,Mouse Colon ,Excitatory postsynaptic potential ,PROTEASE-ACTIVATED RECEPTOR 4 ,Female ,Receptors, Thrombin ,Peptides ,Neuroscience - Abstract
The antinociceptive mechanism underlying protease-activated receptor-4 (PAR(4)) activation was studied in Fast Blue-labelled dorsal root ganglia (DRG) neurons from mouse colon which expressed transcript for PAR(4). Whole cell perforated patch clamp recordings were obtained from these neurons and the effects on neuronal excitability of PAR(4) activating peptides (AP) and reverse peptides (RP) were examined. A 3-min application of PAR(4)-AP (100 micromol L(-1)) markedly suppressed the number of action potential discharged at twice rheobase for up to 60 min. PAR(4)-RP had no effect. PAR(4) application suppresses the excitatory effects of PAR(2). These findings demonstrated that activation of PAR(4) on colonic DRG neurons suppresses their excitability, suggesting these receptors could provide important targets for modifying pain in colonic GI disorders such as IBS and IBD.
- Published
- 2009
- Full Text
- View/download PDF
44. Abstract WP263: Mutagenesis Studies Revealed Minimal Impact of Human A120T Variant of Protease-activated Receptor 4 on Receptor function or Pharmacological Response to a Potent and Selective Antagonist
- Author
-
Scott Priestley, Jeffrey Colin, Xuewen Ma, Michel Bouvier, Jacques Banville, Sophie Desmeules, Anne Marinier, Roger Remillard, Fraz A. Ismat, Mario Callejo, Ke Xu, Tao Wang, John N. Feder, Zhaoqing Wang, Gabe Mintier, Chi Sum, and Jing Yang
- Subjects
Advanced and Specialized Nursing ,medicine.medical_specialty ,business.industry ,Mutagenesis ,Gene mutation ,Pharmacology ,Thrombin ,Endocrinology ,Internal medicine ,Thrombin receptor ,medicine ,PROTEASE-ACTIVATED RECEPTOR 4 ,Platelet ,Neurology (clinical) ,Cardiology and Cardiovascular Medicine ,Receptor ,business ,Function (biology) ,medicine.drug - Abstract
Introduction: Protease-activated receptor 4 (PAR4) is a platelet thrombin receptor and a novel target for ischemic stroke treatment. Recent reports suggested a subtle racial difference in platelet responses to submaximal concentrations of PAR4 agonist peptide (PAR4-AP). One of the PAR4 variants, A120T, is more common in African American than white (63% vs. 19%, Edelstein et al., Blood 2014). It was suggested that this variant contributes to the difference in response to PAR4-AP and impacts the in vitro response to YD-3, a PAR4 antagonist. In this study, site-directed mutagenesis was used to evaluate the effect of the A120T variant on PAR4 function and response to a newly discovered potent and selective PAR4 antagonist, UDM-001651. Unlike YD-3, UDM-001651 inhibited thrombin-induced platelet aggregation and prevented thrombosis in a monkey model and thus served as a relevant PAR4 pharmacology tool. Methods: Human PAR4 cDNAs expressing A120 and T120 variants were stably expressed in HEK293 cells. Cell surface expression of PAR4 was analyzed by FACS. Functional responses of cells were evaluated by monitoring calcium mobilization induced by BMS PAR4-AP, which was optimized based on AYPGKF. The potency of UDM-001651 was derived from an 11-point concentration response curve in the calcium assay using PAR4-AP at the EC80 concentration. Transient transfection studies were also performed to confirm the results. Results: Comparable levels of expression and functional responses were observed between A120 and T120 expressing cells. Results from side-by-side comparison between the two cell lines demonstrated no detectable difference in the mean EC50 values (0.42±0.054 versus 0.46±0.051 uM standard error, n=9) in calcium responses to PAR4-AP stimulation. Similarly, the potency of UDM-001651 in the same assay were similar between these two cell lines. Moreover, preliminary clinical results did not show differences in PAR4-mediated platelet response between African-American and white subjects. Conclusion: In contrast to the previous report, the results reported herein from site directed mutagenesis studies indicate that the A120T variant of PAR4 has no apparent impact on calcium signaling in response to agonist stimulation or response to a PAR4 antagonist.
- Published
- 2016
- Full Text
- View/download PDF
45. Protease-activated receptor 4 protects mice from Coxsackievirus B3 and H1N1 influenza A virus infection.
- Author
-
Tatsumi, Kohei, Schmedes, Clare M., Houston, E. Reaves, Butler, Emily, Mackman, Nigel, and Antoniak, Silvio
- Subjects
- *
PROTEASE-activated receptors , *H1N1 influenza , *INFLUENZA A virus, H1N1 subtype , *VIRUS diseases , *COXSACKIEVIRUS diseases , *INFLUENZA A virus , *INFLUENZA - Abstract
• Protease-activated receptor 4 (PAR4) reduces Coxsackievirus B3 (CVB3) myocarditis and H1N1 influenza A virus (IAV) infection. • PAR4−/− mice exhibited increased virus load and reduced innate immune response in the heart after CVB3 infection. • PAR4−/− mice exhibit more severe myocarditis and heart function impairment. • PAR4−/− mice exhibited more pronounced cellular and cytokine immune responses in the lung after IAV infection. • More severe H1N1 IAV infection in PAR4−/− mice led to more lung injury and death compared to WT mice. PAR4 is expressed by a variety of cells, including platelets, cardiac, lung and immune cells. We investigated the contribution of PAR4 to viral infections of the heart and lung. Toll-like receptor (TLR) 3-dependent immune responses were analyzed after co-stimulation of PAR4 in murine bone-marrow derived macrophages, embryonic fibroblasts and embryonic cardiomyocytes. In addition, we analyzed Coxsackievirus B3 (CVB3) or H1N1 influenza A virus (H1N1 IAV) infection of PAR4−/− (ΔPAR4) and wild-type (WT) mice. Lastly, we investigated the effect of platelet inhibition on H1N1 IAV infection. In vitro experiments revealed that PAR4 stimulation enhances the expression of TLR3-dependent CXCL10 expression and decreases TLR3-dependent NFκB-mediated proinflammatory gene expression. Furthermore, CVB3-infected ΔPAR4 mice exhibited a decreased anti-viral response and increased viral genomes in the heart leading to more pronounced CVB3 myocarditis compared to WT mice. Similarly, H1N1 IAV-infected ΔPAR4 mice had increased immune cell numbers and inflammatory mediators in the lung, and increased mortality compared with infected WT controls. The study showed that PAR4 protects mice from viral infections of the heart and lung. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
46. Protease-Activated Receptor 4 Variant p.Tyr157Cys Reduces Platelet Functional Responses and Alters Receptor Trafficking
- Author
-
Margaret R. Cunningham, Richard B. Sessions, Matthew L. Jones, Mary E. Walker, Stuart J. Mundell, Jane E. Norman, Sarah K Westbury, and Andrew D Mumford
- Subjects
0301 basic medicine ,Male ,Models, Molecular ,Glycosylation ,Platelet Aggregation ,Protein Conformation ,Pyridines ,genotype ,Endoplasmic Reticulum ,Lactones ,HEK293 cells ,Platelet ,Tyrosine ,Receptor ,Thrombin ,Transfection ,Cell biology ,Protein Transport ,Phenotype ,Biochemistry ,England ,Platelet aggregation inhibitor ,Female ,Cardiology and Cardiovascular Medicine ,Blood Platelets ,Genotype ,Mutation, Missense ,platelet activation inhibitors ,Biology ,Polymorphism, Single Nucleotide ,RS ,03 medical and health sciences ,Structure-Activity Relationship ,platelet activation ,Humans ,Computer Simulation ,Receptor, PAR-1 ,Platelet activation ,Aged ,Dose-Response Relationship, Drug ,Endoplasmic reticulum ,HEK 293 cells ,Hydrogen Bonding ,Platelet Activation ,030104 developmental biology ,HEK293 Cells ,protease-activated receptor 4 ,Case-Control Studies ,Receptors, Thrombin ,Peptides ,Platelet Aggregation Inhibitors - Abstract
Objective— Protease-activated receptor 4 (PAR4) is a key regulator of platelet reactivity and is encoded by F2RL3 , which has abundant rare missense variants. We aimed to provide proof of principle that rare F2LR3 variants potentially affect platelet reactivity and responsiveness to PAR1 antagonist drugs and to explore underlying molecular mechanisms. Approach and Results— We identified 6 rare F2RL3 missense variants in 236 cardiac patients, of which the variant causing a tyrosine 157 to cysteine substitution (Y157C) was predicted computationally to have the greatest effect on PAR4 structure. Y157C platelets from 3 cases showed reduced responses to PAR4-activating peptide and to α-thrombin compared with controls, but no reduction in responses to PAR1-activating peptide. Pretreatment with the PAR1 antagonist vorapaxar caused lower residual α-thrombin responses in Y157C platelets than in controls, indicating greater platelet inhibition. HEK293 cells transfected with a PAR4 Y157C expression construct had reduced PAR4 functional responses, unchanged total PAR4 expression but reduced surface expression. PAR4 Y157C was partially retained in the endoplasmic reticulum and displayed an expression pattern consistent with defective N -glycosylation. Mutagenesis of Y322, which is the putative hydrogen bond partner of Y157, also reduced PAR4 surface expression in HEK293 cells. Conclusions— Reduced PAR4 responses associated with Y157C result from aberrant anterograde surface receptor trafficking, in part, because of disrupted intramolecular hydrogen bonding. Characterization of PAR4 Y157C establishes that rare F2RL3 variants have the potential to markedly alter platelet PAR4 reactivity particularly after exposure to therapeutic PAR1 antagonists.
- Published
- 2015
- Full Text
- View/download PDF
47. Abstract 15433: Thrombin Activation of Platelet Protease-activated Receptor 4 Augments Atherosclerosis
- Author
-
A. Phillip Owens, Deborah A. Howatt, Adrien Mann, Debra L. Rateri, Alan Daugherty, and Nigel Mackman
- Subjects
Thrombin ,business.industry ,Physiology (medical) ,medicine ,PROTEASE-ACTIVATED RECEPTOR 4 ,Platelet ,Platelet activation ,Cardiology and Cardiovascular Medicine ,Receptor ,business ,medicine.drug ,Cell biology - Abstract
Objective: Platelet activation has been shown to play a critical role in both formation and propagation of atherosclerosis. Protease-activated receptors (PARs) 1 and 4 mediate signaling by the coagulation protease thrombin. While PAR1 and 4 mediate thrombin activation on human platelets, mouse platelets contain PAR4 with PAR3 acting as a cofactor. Recent studies have demonstrated direct thrombin inhibitors (DTIs) reduced atherosclerosis in hypercholesterolemic mice. In this study, we examined the effect of PAR1 and 4 deficient on atherosclerosis using low-density lipoprotein receptor deficient (Ldlr -/- ) mice. Methods and Results: Ldlr -/- mice that were PAR1 +/+ , PAR1 -/- , PAR4 +/+ , or PAR4 -/- (n = 5-20 each group) were fed a fat and cholesterol-enriched diet for 12 weeks. PAR4 deficiency attenuated aortic root (65% decrease; P = 0.001) and aortic arch (71% decrease; P = 0.001) atherosclerosis with no effects on total plasma cholesterol concentrations or lipoprotein distributions. These reductions were attributable to hematopoietic-derived PAR4 from analysis of bone marrow experiments. To determine if PAR4 mediated all effects of thrombin signaling, Ldlr -/- /PAR4 +/+ or Ldlr -/- /PAR4 -/- mice were fed a fat and cholesterol-enriched diet, supplemented with placebo or the DTI dabigatran etexilate (30 g/kg diet) for 12 weeks. Dabigatran etexilate administration reduced atherosclerosis in PAR4 +/+ mice but not in PAR4 -/- mice. PAR1 deficiency had no effect on atherosclerotic burden. Conclusion: We demonstrated whole body and hematopoietic PAR4 deficiency resulted in decreased aortic root and sinus atherosclerosis. PAR1 deficiency had no effects on atherosclerosis. Thrombin inhibition did not alter atherosclerosis in PAR4 deficient mice. Together, these results suggest that thrombin activation of PAR4 on platelets contributes to atherosclerosis.
- Published
- 2015
- Full Text
- View/download PDF
48. New insights into protease-activated receptor 4 signaling pathways in the pathogenesis of inflammation and neuropathic pain: a literature review
- Author
-
Bao, Yanju, Gao, Yebo, Yang, Liping, Kong, Xiangying, Zheng, Honggang, Hou, Wei, and Hua, Baojin
- Subjects
Inflammation ,business.industry ,Biophysics ,Chronic pain ,Review ,medicine.disease ,Bioinformatics ,Biochemistry ,Pathogenesis ,Intracellular signaling pathways ,Anesthesia ,Neuropathic pain ,medicine ,PROTEASE-ACTIVATED RECEPTOR 4 ,Animals ,Humans ,Neuralgia ,Receptors, Thrombin ,Signal transduction ,medicine.symptom ,Receptor ,business ,Signal Transduction - Abstract
Pain is an unpleasant sensory and emotional experience that is commonly associated with actual or potential tissue damage. Despite decades of pain research, many patients continue to suffer from chronic pain that is refractory to current treatments. Accumulating evidence has indicated an important role of protease-activated receptor 4 (PAR4) in the pathogenesis of inflammation and neuropathic pain. Here we reviewed PAR4 expression and activation via intracellular signaling pathways and the role of PAR4 signaling pathways in the development and maintenance of pain. Understanding PAR4 and its corresponding signaling pathways will provide insight to further explore the molecular basis of pain, which will also help to identify new targets for pharmacological intervention for pain relief.
- Published
- 2015
49. Selective Inhibition of PAR4 (Protease-Activated Receptor 4)-Mediated Platelet Activation by a Synthetic Nonanticoagulant Heparin Analog.
- Author
-
Lin YC, Ko YC, Hung SC, Lin YT, Lee JH, Tsai JY, Kung PH, Tsai MC, Chen YF, and Wu CC
- Subjects
- Animals, Antithrombins chemical synthesis, CHO Cells, Calcium Signaling drug effects, Computer Simulation, Cricetulus, Drug Evaluation, Preclinical, Humans, In Vitro Techniques, Models, Molecular, Recombinant Proteins drug effects, Thrombin pharmacology, Thrombosis prevention & control, Antithrombins pharmacology, Heparin chemistry, Oligosaccharides pharmacology, Platelet Aggregation drug effects, Receptors, Thrombin antagonists & inhibitors
- Abstract
Objective- PAR4 (protease-activated receptor 4), one of the thrombin receptors in human platelets, has emerged as a promising target for the treatment of arterial thrombotic disease. Previous studies implied that thrombin exosite II, known as a binding site for heparin, may be involved in thrombin-induced PAR4 activation. In the present study, a heparin octasaccharide analog containing the thrombin exosite II-binding domain of heparin was chemically synthesized and investigated for anti-PAR4 effect. Approach and Results- PAR4-mediated platelet aggregation was examined using either thrombin in the presence of a PAR1 antagonist or γ-thrombin, which selectively activates PAR4. SCH-28 specifically inhibits PAR4-mediated platelet aggregation, as well as the signaling events downstream of PAR4 in response to thrombin. Moreover, SCH-28 prevents thrombin-induced β-arrestin recruitment to PAR4 but not PAR1 in Chinese Hamster Ovary-K1 cells using a commercial enzymatic complementation assay. Compared with heparin, SCH-28 is more potent in inhibiting PAR4-mediated platelet aggregation but has no significant anticoagulant activity. In an in vitro thrombosis model, SCH-28 reduces thrombus formation under whole blood arterial flow conditions. Conclusions- SCH-28, a synthetic small-molecular and nonanticoagulant heparin analog, inhibits thrombin-induced PAR4 activation by interfering with thrombin exosite II, a mechanism of action distinct from other PAR4 inhibitors that target the receptor. The characteristics of SCH-28 provide a new strategy for targeting PAR4 with the potential for the treatment of arterial thrombosis.
- Published
- 2019
- Full Text
- View/download PDF
50. Regulation of protease‐activated receptor‐4 signaling and trafficking
- Author
-
JoAnn Trejo and Thomas H. Smith
- Subjects
Chemistry ,Genetics ,PROTEASE-ACTIVATED RECEPTOR 4 ,Molecular Biology ,Biochemistry ,Biotechnology ,Cell biology - Published
- 2012
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.