1,557 results on '"thrombin receptor"'
Search Results
2. The GPIb-IX complex on platelets: insight into its novel physiological functions affecting immune surveillance, hepatic thrombopoietin generation, platelet clearance and its relevance for cancer development and metastasis
- Author
-
Gerd Bendas and Martin Schlesinger
- Subjects
GPIb-IX complex ,Platelets ,Bernard-Soulier syndrome ,Thrombin receptor ,Mechanosensitive domains ,Platelet biogenesis ,Diseases of the blood and blood-forming organs ,RC633-647.5 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract The glycoprotein (GP) Ib-IX complex is a platelet receptor that mediates the initial interaction with subendothelial von Willebrand factor (VWF) causing platelet arrest at sites of vascular injury even under conditions of high shear. GPIb-IX dysfunction or deficiency is the reason for the rare but severe Bernard-Soulier syndrome (BSS), a congenital bleeding disorder. Although knowledge on GPIb-IX structure, its basic functions, ligands, and intracellular signaling cascades have been well established, several advances in GPIb-IX biology have been made in the recent years. Thus, two mechanosensitive domains and a trigger sequence in GPIb were characterized and its role as a thrombin receptor was deciphered. Furthermore, it became clear that GPIb-IX is involved in the regulation of platelet production, clearance and thrombopoietin secretion. GPIb is deemed to contribute to liver cancer development and metastasis. This review recapitulates these novel findings highlighting GPIb-IX in its multiple functions as a key for immune regulation, host defense, and liver cancer development.
- Published
- 2022
- Full Text
- View/download PDF
3. The GPIb-IX complex on platelets: insight into its novel physiological functions affecting immune surveillance, hepatic thrombopoietin generation, platelet clearance and its relevance for cancer development and metastasis.
- Author
-
Bendas, Gerd and Schlesinger, Martin
- Subjects
CARCINOGENESIS ,THROMBOPOIETIN ,METASTASIS ,BLOOD platelets ,THROMBIN receptors - Abstract
The glycoprotein (GP) Ib-IX complex is a platelet receptor that mediates the initial interaction with subendothelial von Willebrand factor (VWF) causing platelet arrest at sites of vascular injury even under conditions of high shear. GPIb-IX dysfunction or deficiency is the reason for the rare but severe Bernard-Soulier syndrome (BSS), a congenital bleeding disorder. Although knowledge on GPIb-IX structure, its basic functions, ligands, and intracellular signaling cascades have been well established, several advances in GPIb-IX biology have been made in the recent years. Thus, two mechanosensitive domains and a trigger sequence in GPIb were characterized and its role as a thrombin receptor was deciphered. Furthermore, it became clear that GPIb-IX is involved in the regulation of platelet production, clearance and thrombopoietin secretion. GPIb is deemed to contribute to liver cancer development and metastasis. This review recapitulates these novel findings highlighting GPIb-IX in its multiple functions as a key for immune regulation, host defense, and liver cancer development. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
4. 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
5. 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
6. The Platelet PARs
- Author
-
Arachiche, Amal, Nieman, Marvin T., Gresele, Paolo, editor, Kleiman, Neal S., editor, Lopez, José A., editor, and Page, Clive P., editor
- Published
- 2017
- Full Text
- View/download PDF
7. Thrombin Receptor
- Author
-
Offermanns, Stefan, editor and Rosenthal, Walter, editor
- Published
- 2021
- Full Text
- View/download PDF
8. Discovery of (E)‐5,5‐Difluoro‐1‐[2‐[5‐(3‐fluorophenyl)pyridin‐2‐yl]vinyl]octahydrospiro(indene‐2,5′‐oxazolidin)‐2′‐one as a PAR1 Antagonist.
- Author
-
Park, Chul Min, Lee, Sunkyung, Song, Jong‐Hwan, and Lee, Joo‐Youn
- Subjects
- *
KRA , *BLOOD platelet aggregation , *THROMBIN receptors , *STEREOISOMERS - Abstract
In a previous study, we showed that several octahydroindene derivatives are potent protease activated receptor 1 (PAR1) antagonists. In the current study, we prepared a series of trans‐fused 5,5‐difulorooctahydroindenes which do not have a C5 stereogenic center, and evaluated their biological activities and metabolic stabilities. Compound 19 in this series, containing a spirooxazolidinone moiety at C2, showed excellent efficacy in both PAR1 binding (IC50 = 70 nM) and human platelet rich plasma (PRP) aggregation (IC50 = 0.19 μM), along with good metabolic stability (R50 = 345.8, 337.2, and 43.4 min in human, rat, and monkey liver microsomes, each), which is comparable to that of vorapaxar. Four stereoisomers of 19 were prepared and evaluated. (1S,2R,3aR,7aR)‐5,5‐Difluoro‐1‐[(E)‐2‐[5‐(3‐fluorophenyl)pyridin‐2‐yl]vinyl]octahydrospiro(indene‐2,5′‐oxazolidin)‐2′‐one (31) was found to be the most active (IC50 = 21 nM) stereoisomer as PAR1 antagonist. Compound 31 exhibited good in vivo oral PK profiles and significant ex vivo antithrombotic efficacy in cynomolgus monkeys upon oral administration. When the plasma concentration of 31 was maintained above 200 ng/mL, platelet aggregation induced by haTRAP was completely inhibited. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
9. Proteinase-activated receptor 1 antagonism ameliorates experimental pulmonary hypertension.
- Author
-
Kuwabara, Yukimitsu, Tanaka-Ishikawa, Mariko, Abe, Kohtaro, Hirano, Mayumi, Hirooka, Yoshitaka, Tsutsui, Hiroyuki, Sunagawa, Kenji, and Hirano, Katsuya
- Subjects
- *
PULMONARY hypertension , *THROMBIN receptors , *VASCULAR remodeling , *CARDIOVASCULAR system , *VASCULAR smooth muscle , *MEDICAL sciences - Published
- 2019
- Full Text
- View/download PDF
10. A Novel Compound Targeting Protease Receptor 1 Activators for the Treatment of Glioblastoma
- Author
-
Efrat Shavit-Stein, Ehud Sheinberg, Valery Golderman, Shirley Sharabi, Anton Wohl, Shany Guly Gofrit, Zion Zivli, Natalia Shelestovich, David Last, David Guez, Dianne Daniels, Orna Gera, Kate Feingold, Zeev Itsekson-Hayosh, Nurit Rosenberg, Ilia Tamarin, Amir Dori, Nicola Maggio, Yael Mardor, Joab Chapman, and Sagi Harnof
- Subjects
glioblastoma ,thrombin receptor ,PAR1 ,tumor size ,edema ,survival ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Data from human biopsies, in-vitro and in-vivo models, strongly supports the role of thrombin, and its protease-activated receptor (PAR1) in the pathology and progression of glioblastoma (GBM), a high-grade glial tumor. Activation of PAR1 by thrombin stimulates vasogenic edema, tumor adhesion and tumor growth. We here present a novel six amino acid chloromethyl-ketone compound (SIXAC) which specifically inhibits PAR1 proteolytic activation and counteracts the over-activation of PAR1 by tumor generated thrombin. SIXAC effects were demonstrated in-vitro utilizing 3 cell-lines, including the highly malignant CNS-1 cell-line which was also used as a model for GBM in-vivo. The in-vitro effects of SIXAC on proliferation rate, invasion and thrombin activity were measured by XTT, wound healing, colony formation and fluorescent assays, respectively. The effect of SIXAC on GBM in-vivo was assessed by measuring tumor and edema size as quantified by MRI imaging, by survival follow-up and brain histopathology. SIXAC was found in-vitro to inhibit thrombin-activity generated by CNS-1 cells (IC50 = 5 × 10−11M) and significantly decrease proliferation rate (p < 0.03) invasion (p = 0.02) and colony formation (p = 0.03) of these cells. In the CNS-1 GBM rat animal model SIXAC was found to reduce edema volume ratio (8.8 ± 1.9 vs. 4.9 ± 1, p < 0.04) and increase median survival (16 vs. 18.5 days, p < 0.02 by Log rank Mental-Cox test). These results strengthen the important role of thrombin/PAR1 pathway in glioblastoma progression and suggest SIXAC as a novel therapeutic tool for this fatal disease.
- Published
- 2018
- Full Text
- View/download PDF
11. Proteinase-Activated Receptors (PARs) and Calcium Signaling in Cancer
- Author
-
Kaufmann, Roland, Hollenberg, Morley D., and Islam, Md. Shahidul, editor
- Published
- 2012
- Full Text
- View/download PDF
12. Thrombin in Ischemic Stroke Targeting
- Author
-
Chen, Bo, Lapchak, Paul A., editor, and Zhang, John H., editor
- Published
- 2012
- Full Text
- View/download PDF
13. Thrombin receptor PAR1 silencing in endothelial colony-forming cells modifies stemness and vasculogenic properties.
- Author
-
Smadja DM, Rossi E, Haviari S, Bieche I, Cras A, and Gaussem P
- Subjects
- Humans, Cells, Cultured, Neovascularization, Physiologic, Receptors, Thrombin metabolism, RNA, Small Interfering genetics, RNA, Small Interfering metabolism, Endothelial Cells metabolism, Receptor, PAR-1 genetics, Receptor, PAR-1 metabolism
- Abstract
Background: The involvement of thrombin receptor PAR1 in blood vessel development has been largely demonstrated in knockout mice; however, its implication in adult mouse angiogenesis seems very moderate., Objectives: We aimed to explore the potential relationships between PAR1, stemness, and angiogenic properties of human endothelial colony-forming cells (ECFCs)., Methods and Results: PAR1 activation on ECFCs using the selective PAR1-activating peptide induced a significant decrease in CD133 expression (RTQ-PCR analysis). In line, silencing of PAR1 gene expression with siRNA increased CD133 mRNA as well as intracellular CD133 protein expression. To confirm the link between CD133 and PAR1, we explored the association between PAR1 and CD133 levels in fast and slow fibroblasts prone to reprogramming. An imbalance between PAR1 and CD133 levels was evidenced, with a decreased expression of PAR1 in fast reprogramming fibroblasts expressing a high CD133 level. Regarding in vitro ECFC angiogenic properties, PAR1 silencing with specific siRNA induced cell proliferation evidenced by the overexpression of Ki67. However, it did not impact migration properties nor ECFC adhesion on smooth muscle cells or human arterial endothelial cells. In a mouse model of hind-limb ischemia, PAR1 silencing in ECFCs significantly increased postischemic revascularization compared to siCtrl-ECFCs along with a significant increase in cutaneous blood flows (P < .0001), microvessel density (P = .02), myofiber regeneration (P < .0001), and human endothelial cell incorporation in muscle (P < .0001)., Conclusion: In conclusion, our work describes for the first time a link between PAR1, stemness, and vasculogenesis in human ECFCs., Competing Interests: Declaration of competing interests There are no competing interests to disclose., (Copyright © 2023 International Society on Thrombosis and Haemostasis. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
14. A Mouse Model of the Protease Activated Receptor 4 (PAR4) Pro310Leu Variant has Reduced Platelet Reactivity.
- Author
-
Han X, Knauss EA, de la Fuente M, Li W, Conlon RA, LePage DF, Jiang W, Renna SA, McKenzie SE, and Nieman MT
- Abstract
Background: Protease activated receptor 4 (PAR4) mediates thrombin signaling on platelets and other cells. Our recent structural studies demonstrated a single nucleotide polymorphism in extracellular loop 3 (ECL3), PAR4-P310L (rs2227376) leads to a hypo-reactive receptor., Objectives: The goal of this study was to determine how the hypo-reactive PAR4 variant in ECL3 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., Conclusions: 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: Authors conflict of interest statement The authors declare no conflicts of interest.
- Published
- 2023
- Full Text
- View/download PDF
15. The Role of Thrombin and Thrombin Receptors in the Brain
- Author
-
Luo, Weibo, Wang, Yingfei, Reiser, Georg, Maragoudakis, Michael E., editor, and Tsopanoglou, Nikos E., editor
- Published
- 2009
- Full Text
- View/download PDF
16. Thrombin and Thrombin Peptides in Wound Healing and Tissue Repair
- Author
-
Olszewska-Pazdrak, Barbara, Bergmann, John S., Fuller, Gerald M., Carney, Darrell H., Maragoudakis, Michael E., editor, and Tsopanoglou, Nikos E., editor
- Published
- 2009
- Full Text
- View/download PDF
17. Regulation of Thrombin Receptor Signaling
- Author
-
Trejo, JoAnn, Maragoudakis, Michael E., editor, and Tsopanoglou, Nikos E., editor
- Published
- 2009
- Full Text
- View/download PDF
18. Thrombin: To PAR or Not to PAR, and the Regulation of Inflammation
- Author
-
Ramachandran, Rithwik, El-Daly, Mahmoud, Saifeddine, Mahmoud, Hollenberg, Morley D., Maragoudakis, Michael E., editor, and Tsopanoglou, Nikos E., editor
- Published
- 2009
- Full Text
- View/download PDF
19. Amyloid β protein negatively regulates human platelet activation induced by thrombin receptor-activating protein
- Author
-
Shinji Ogura, Yukiko Enomoto, Rie Matsushima-Nishiwaki, Kodai Uematsu, Daiki Nakashima, Daisuke Mizutani, Kyohei Ueda, Takashi Onuma, Toru Iwama, Hiroki Iida, Haruhiko Tokuda, Osamu Kozawa, and Tomoaki Doi
- Subjects
Amyloid beta-Peptides ,Platelet-derived growth factor ,biology ,Chemistry ,p38 mitogen-activated protein kinases ,Organic Chemistry ,General Medicine ,Applied Microbiology and Biotechnology ,Biochemistry ,Analytical Chemistry ,Cell biology ,chemistry.chemical_compound ,Thrombin ,Mechanism of action ,Mitogen-activated protein kinase ,Thrombin receptor ,biology.protein ,medicine ,Phosphorylation ,Platelet ,medicine.symptom ,Molecular Biology ,Biotechnology ,medicine.drug - Abstract
Amyloid β protein deposition in cerebral vessels, a characteristic of Alzheimer's disease, is a risk factor for intracerebral hemorrhage. Amyloid β protein directly modulates human platelet function; however, the exact mechanism of action is unclear. Therefore, we investigated the effects of amyloid β protein on human platelet activation using an aggregometer with laser scattering. Amyloid β protein decreased platelet aggregation induced by thrombin receptor-activating protein, but not by collagen and ADP. Amyloid β protein also suppressed platelet aggregation induced by SCP0237 and A3227. Platelet-derived growth factor-AB secretion and phosphorylated-heat shock protein 27 release by thrombin receptor-activating protein were inhibited by amyloid β protein. Additionally, thrombin receptor-activating protein-induced phosphorylation of JNK and p38 MAP kinase was reduced by amyloid β protein. Collectively, our results strongly suggest that amyloid β protein negatively regulates protease-activated receptor-elicited human platelet activation. These findings may indicate a cause of intracerebral hemorrhage due to amyloid β protein.
- Published
- 2021
- Full Text
- View/download PDF
20. Synergistic activation of thrombin and angiotensin II receptors revealed by bioluminescence resonance energy transfer
- Author
-
Isra Al Zamel, Abdulrasheed Palakkott, and Mohammed Akli Ayoub
- Subjects
Angiotensin receptor ,Luminescence ,G protein ,Allosteric regulation ,Biophysics ,Biochemistry ,Receptor, Angiotensin, Type 1 ,Thrombin ,Structural Biology ,Thrombin receptor ,Genetics ,medicine ,Humans ,Receptor ,Molecular Biology ,G protein-coupled receptor ,Chemistry ,Cell Biology ,Angiotensin II ,HEK293 Cells ,Energy Transfer ,cardiovascular system ,circulatory and respiratory physiology ,medicine.drug - Abstract
We recently reported a physical interaction between the angiotensin II (AngII) receptor (AT1R) and thrombin receptor (PAR1) in HEK293 cells using bioluminescence resonance energy transfer (BRET) technology. This was characterized by thrombin trans-activating AT1R and the synergistic responses of the AT1R-PAR1 complex. Here, we investigated the other face of the coin by examining the effect of AT1R on PAR1 activity using BRET. AngII/AT1R did not promote PAR1 activation in the absence of thrombin. However, the combination of thrombin and AngII resulted in their synergistic/allosteric action. Moreover, AngII/AT1R potentiated the maximal thrombin responses, suggesting specific conformational changes within the AT1R-PAR1 complex. Overall, our data confirm the functional AT1R-PAR1 interplay and further support the implication of both AT1R and PAR1 protomers in their synergistic interaction as previously reported.
- Published
- 2021
- Full Text
- View/download PDF
21. Protease-Activated Receptors in Gastrointestinal Function and Disease
- Author
-
Bunnett, Nigel W., Cottrell, Graeme S., Hooper, Nigel M., editor, and Lendeckel, Uwe, editor
- Published
- 2006
- Full Text
- View/download PDF
22. Role and Regulation of PAR-1 in Melanoma Progression
- Author
-
Tellez, Carmen, Bar-Eli, Menashe, Hearing, Vincent J., editor, and Leong, Stanley P. L., editor
- Published
- 2006
- Full Text
- View/download PDF
23. Platelet Physiology
- Author
-
Quinn, Martin, Cannon, Christopher P., editor, Quinn, Martin, editor, and Fitzgerald, Desmond, editor
- Published
- 2005
- Full Text
- View/download PDF
24. Role of Proteinase-Activated Receptors in Brain Function : Thrombin signaling and its interacting players in physiologic and pathophysiologic processes
- Author
-
Festoff, Barry W., Lendeckel, Uwe, editor, and Hooper, Nigel M., editor
- Published
- 2005
- Full Text
- View/download PDF
25. The hemostatic system in angiogenesis
- Author
-
van Hinsbergh, Victor W.M., Koolwijk, Pieter, Hoekman, Klaas, Clauss, Matthias, editor, and Breier, Georg, editor
- Published
- 2005
- Full Text
- View/download PDF
26. Coordinated Pre- and Postsynaptic Changes Involved in Developmental Activity-Dependent Synapse Elimination
- Author
-
Nelson, Phillip G., Jia, Min, Li, Min-Xu, Gizaw, Rahel, Lanuza, Maria A., Tomas, Josep, Stanton, Patrick K., editor, Bramham, Clive, editor, and Scharfman, Helen E., editor
- Published
- 2005
- Full Text
- View/download PDF
27. Pleiotropic actions of factor Xa inhibition in cardiovascular prevention - mechanistic insights and implications for anti-thrombotic treatment
- Subjects
ACUTE CORONARY SYNDROMES ,Anticoagulant ,Thrombin ,FIBRINOGEN DEFICIENCY ,Atherosclerosis ,Cardiovascular ,SHEDDING IN-VITRO ,PROCOAGULANT ACTIVITY ,FACTOR-VII ,THROMBIN RECEPTOR ,TISSUE FACTOR ,HUMAN SOLUBLE THROMBOMODULIN ,Factor Xa ,ENDOTHELIAL BARRIER PROTECTION ,PROTEIN-C RECEPTOR - Abstract
Atherosclerosis is a chronic inflammatory disease in which atherothrombotic complications lead to cardiovascular morbidity and mortality. At advanced stages, myocardial infarction, ischemic stroke and peripheral artery disease (PAD), including major adverse limb events (MALE), are caused either by acute occlusive atherothrombosis, or by thromboembolism. Endothelial dysfunction, vascular smooth muscle cell activation and vascular inflammation are essential in the development of acute cardiovascular events. Effects of the coagulation system on vascular biology extend beyond thrombosis. Under physiological conditions, coagulation proteases in blood are pivotal in maintaining hemostasis and vascular integrity. Under pathological conditions, including atherosclerosis, the same coagulation proteases (including factor Xa, factor VIIa and thrombin) become drivers of atherothrombosis, working in concert with platelets and vessel wall components. While initially atherothrombosis was attributed primarily to platelets, recent advances indicate the critical role of fibrin clot and plasma coagulation factors. Mechanisms of atherothrombosis and hypercoagulability vary depending on plaque erosion or plaque rupture. In addition to contributing to thrombus formation, factor Xa and thrombin can affect endothelial dysfunction, oxidative stress, vascular smooth muscle cell function as well as immune cell activation and vascular inflammation. By these mechanisms they promote atherosclerosis and contribute to plaque instability. In this review, we first discuss the postulated vasoprotective mechanisms of protease activated receptor (PARs) signaling induced by coagulation enzymes under physiological conditions. Next, we discuss preclinical studies linking coagulation with endothelial cell dysfunction, thromboinflammation and atherogenesis. Understanding these mechanisms is pivotal for the introduction of novel strategies in cardiovascular prevention and therapy. We therefore translate these findings to clinical studies of direct oral anticoagulant (DOAC) drugs and discuss the potential relevance of dual pathway inhibition for atherothrombosis prevention and vascular protection.
- Published
- 2021
- Full Text
- View/download PDF
28. Dapagliflozin reduces thrombin generation and platelet activation: implications for cardiovascular risk reduction in type 2 diabetes mellitus
- Author
-
Margitta Elvers, Stefan Lehr, Stephen Gerfer, Petra Keul, Sören Twarock, Irena Krüger, Ulrich Flögel, Christina Kohlmorgen, Meike Klier, Sabine Kahl, Maria Grandoch, Michael Roden, Bodo Levkau, Jens W. Fischer, Sonja Hartwig, Carolin Helten, Kathrin Feldmann, Malte Kelm, and Amin Polzin
- Subjects
Blood Glucose ,Male ,Endocrinology, Diabetes and Metabolism ,Coronary Artery Disease ,Type 2 diabetes ,Cardiovascular ,chemistry.chemical_compound ,Glucosides ,Platelet ,Dapagliflozin ,medicine.diagnostic_test ,Thrombin ,Middle Aged ,Flow Cytometry ,Immunohistochemistry ,Healthy Volunteers ,HDL-cholesterol ,P-Selectin ,Cardiovascular Diseases ,Sodium–glucose cotransporter 2 (SGLT2) inhibitors ,Female ,medicine.drug ,Adult ,Blood Platelets ,Platelets ,medicine.medical_specialty ,Heart failure ,Real-Time Polymerase Chain Reaction ,Article ,Bleeding time ,Diabetes mellitus ,Internal medicine ,Thrombin receptor ,P-Selectin (CD62P) ,Internal Medicine ,medicine ,Animals ,Humans ,Platelet activation ,Benzhydryl Compounds ,Sodium-Glucose Transporter 2 Inhibitors ,Platelet Count ,business.industry ,Cholesterol, HDL ,Platelet Activation ,Atherosclerosis ,medicine.disease ,Mice, Inbred C57BL ,Endocrinology ,Diabetes Mellitus, Type 2 ,chemistry ,business ,Risk Reduction Behavior - Abstract
Aims/hypothesis People with diabetes have an increased cardiovascular risk with an accelerated development of atherosclerosis and an elevated mortality rate after myocardial infarction. Therefore, cardioprotective effects of glucose-lowering therapies are of major importance for the pharmacotherapy of individuals with type 2 diabetes. For sodium–glucose cotransporter 2 inhibitors (SGLT2is), in addition to a reduction in blood glucose, beneficial effects on atherosclerosis, obesity, renal function and blood pressure have been observed. Recent results showed a reduced risk of worsening heart failure and cardiovascular deaths under dapagliflozin treatment irrespective of the diabetic state. However, the underlying mechanisms are yet unknown. Platelets are known drivers of atherosclerosis and atherothrombosis and disturbed platelet activation has also been suggested to occur in type 2 diabetes. Therefore, the present study investigates the impact of the SGLT2i dapagliflozin on the interplay between platelets and inflammation in atherogenesis. Methods Male, 8-week-old LDL-receptor-deficient (Ldlr−/−) mice received a high-fat, high-sucrose diabetogenic diet supplemented without (control) or with dapagliflozin (5 mg/kg body weight per day) for two time periods: 8 and 25 weeks. In a first translational approach, eight healthy volunteers received 10 mg dapagliflozin/day for 4 weeks. Results Dapagliflozin treatment ameliorated atherosclerotic lesion development, reduced circulating platelet–leucocyte aggregates (glycoprotein [GP]Ib+CD45+: 29.40 ± 5.94 vs 17.00 ± 5.69 cells, p +lymphocyte antigen 6 complex, locus G+ (Ly6G): 8.00 ± 2.45 vs 4.33 ± 1.75 cells, p 3 cells/aorta, p Ldlr−/− mice fed a diabetogenic diet (3.78 ± 1.20% vs 2.83 ± 1.06%, p p = 0.78). While blood glucose was only moderately affected, dapagliflozin further reduced endogenous thrombin generation (581.4 ± 194.6 nmol/l × min) × 10−9 thrombin vs 254.1 ± 106.4 (nmol/l × min) × 10−9 thrombin), thereby decreasing one of the most important platelet activators. We observed a direct inhibitory effect of dapagliflozin on isolated platelets. In addition, dapagliflozin increased HDL-cholesterol levels. Importantly, higher HDL-cholesterol levels (1.70 ± 0.58 vs 3.15 ± 1.67 mmol/l, p p p Conclusions/interpretation We demonstrate that dapagliflozin-mediated atheroprotection in mice is driven by elevated HDL-cholesterol and ameliorated thrombin–platelet-mediated inflammation without interfering with haemostasis. This glucose-independent mechanism likely contributes to dapagliflozin’s beneficial cardiovascular risk profile. Graphical abstract
- Published
- 2021
- Full Text
- View/download PDF
29. Effects of the Tryptase Receptor Activating Peptide and Antibodies Against the Tryptase Receptor Par-2 on Neonatal Rat Cardiomyocytes in Culture
- Author
-
Wallukat, Gerd, Morwinski, Rosemarie, Nissen, Eberhard, Müller, Johannes, Luft, Friedrich C., and Matsumori, Akira, editor
- Published
- 2003
- Full Text
- View/download PDF
30. Receptor-Mediated Regulation of the Cardiac Sarcolemmal Na+/H+ Exchanger : Mechanisms And (Patho)Physiological Significance
- Author
-
Haworth, Robert S., Avkiran, Metin, Karmazyn, Morris, editor, Avkiran, Metin, editor, and Fliegel, Larry, editor
- Published
- 2003
- Full Text
- View/download PDF
31. Thrombin Receptor
- Author
-
Choi, Sangdun, editor
- Published
- 2018
- Full Text
- View/download PDF
32. Sarsasapogenin ameliorates diabetes‐associated memory impairment and neuroinflammation through down‐regulation of <scp>PAR</scp> ‐1 receptor
- Author
-
Li Kong, Teng-Fei Ma, Yu-Meng Zhang, Ling-Shan Gou, Yue Liu, Yao-Wu Liu, and Yu Li
- Subjects
Male ,Inflammasomes ,Down-Regulation ,Morris water navigation task ,Hippocampus ,Pharmacology ,Neuroprotection ,Streptozocin ,Cell Line ,Diabetes Mellitus, Experimental ,Rats, Sprague-Dawley ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Memory ,Thrombin receptor ,Spirostans ,medicine ,Animals ,Humans ,Memory impairment ,Receptor, PAR-1 ,Neuroinflammation ,Memory Disorders ,0303 health sciences ,Chemistry ,030302 biochemistry & molecular biology ,NF-kappa B ,Inflammasome ,Sarsasapogenin ,Rats ,030220 oncology & carcinogenesis ,Signal Transduction ,medicine.drug - Abstract
Sarsasapogenin (Sar), a natural steroidal compound, shows neuroprotection, cognition-enhancement, antiinflammation, antithrombosis effects, and so on. However, whether Sar has ameliorative effects on diabetes-associated cognitive impairment remains unknown. In this study, we found that Sar ameliorated diabetes-associated memory impairment in streptozotocin-induced diabetic rats, evidenced by increased numbers of crossing platform and percentage of time spent in the target quadrant in Morris water maze tests, and suppressed the nucleotide-binding domain and leucine-rich repeat containing protein 1 (NLRP1) inflammasome in hippocampus and cerebral cortex. Furthermore, Sar inhibited advanced glycation end-products and its receptor (AGEs/RAGE) axis and suppressed up-regulation of thrombin receptor protease-activated receptor 1 (PAR-1) in cerebral cortex. On the other hand, Sar mitigated high glucose-induced neuronal damages, NLRP1 inflammasome activation, and PAR-1 up-regulation in high glucose-cultured SH-SY5Y cells, but did not affect thrombin activity. Moreover, the effects of Sar were similar to those of a selective PAR-1 antagonist vorapaxar. Further studies indicated that activation of the NLRP1 inflammasome and NF-κB mediated the effect of PAR-1 up-regulation in high glucose condition by using PAR-1 knockdown assay. In summary, this study demonstrated that Sar prevented memory impairment caused by diabetes, which was achieved through suppressing neuroinflammation from activated NLRP1 inflammasome and NF-κB regulated by cerebral PAR-1. HIGHLIGHTS: Sarsasapogenin ameliorated memory impairment caused by diabetes in rats. Sarsasapogenin mitigated neuronal damages and neuroinflammation by down-regulating cerebral PAR-1. The NLRP1 inflammasome and NF-κB signaling mediated the pro-inflammatory effects of PAR-1. Sarsasapogenin was a pleiotropic neuroprotective agent and memory enhancer in diabetic rodents.
- Published
- 2021
- Full Text
- View/download PDF
33. Targeting platelet inhibition receptors for novel therapies: PECAM-1 and G6b-B
- Author
-
Eva M Soriano Jerez, Craig E. Hughes, and Jonathan M. Gibbins
- Subjects
Blood Platelets ,0301 basic medicine ,030204 cardiovascular system & hematology ,Pharmacology ,Ligands ,Collagen receptor ,Mice ,03 medical and health sciences ,0302 clinical medicine ,P2Y12 ,Thrombin receptor ,Animals ,Humans ,Medicine ,Platelet ,Platelet activation ,Receptors, Immunologic ,Receptor ,Mice, Knockout ,business.industry ,Hematology ,General Medicine ,Platelet Activation ,Platelet Endothelial Cell Adhesion Molecule-1 ,030104 developmental biology ,GPVI ,Signal transduction ,business - Abstract
While current oral antiplatelet therapies benefit many patients, they deregulate the haemostatic balance leaving patients at risk of systemic side-effects such as haemorrhage. Dual antiplatelet treatment is the standard approach, combining aspirin with P2Y12 blockers. These therapies mainly target autocrine activation mechanisms (TxA2, ADP) and, more recently, the use of thrombin or thrombin receptor antagonists have been added to the available approaches. Recent efforts to develop new classes of anti-platelet drugs have begun to focus on primary platelet activation pathways such as through the immunoreceptor tyrosine-based activation motif (ITAM)-containing collagen receptor GPVI/FcR-chain complex. There are already encouraging results from targeting GPVI, with reduced aggregation and smaller arterial thrombi, without major bleeding complications, likely due to overlapping activation signalling pathways with other receptors such as the GPIb–V–IX complex.\ud \ud An alternative approach to reduce platelet activation could be to inhibit this signalling pathway by targeting the inhibitory pathways intrinsic to platelets. Stimulation of endogenous negative modulators could provide more specific inhibition of platelet function, but is this feasible? In this review, we explore the potential of the two major platelet immunoreceptor tyrosine-based inhibitory motif (ITIM)-containing inhibitory receptors, G6b-B and PECAM-1, as antithrombotic targets.
- Published
- 2021
- Full Text
- View/download PDF
34. A Novel Proteolytic Mechanism for Termination of the Ca2+ Signalling Evoked by Proteinase-Activated Receptor-1 (PAR-1) in Rat Astrocytes
- Author
-
Ubl, Joachim J., Reiser, Georg, Back, Nathan, editor, Cohen, Irun R., editor, Kritchevsky, David, editor, Lajtha, Abel, editor, Paoletti, Rodolfo, editor, Langner, Jürgen, editor, and Ansorge, Siegfried, editor
- Published
- 2002
- Full Text
- View/download PDF
35. Vascular Endothelium
- Author
-
Yang, Zhihong, Lüscher, Thomas F., Lanzer, Peter, editor, and Topol, Eric J., editor
- Published
- 2002
- Full Text
- View/download PDF
36. Interface Between Inflammation and Coagulation
- Author
-
Altieri, Dario C. and Ley, Klaus, editor
- Published
- 2001
- Full Text
- View/download PDF
37. Protease-activated receptor-4 (PAR4) variant influences on platelet reactivity induced by PAR4-activating peptide through altered Ca2+ mobilization and ERK phosphorylation in healthy Japanese subjects.
- Author
-
Morikawa, Yoichiro, Kato, Hisashi, Kashiwagi, Hirokazu, Nishiura, Nobuko, Akuta, Keigo, Honda, Shigenori, Kanakura, Yuzuru, and Tomiyama, Yoshiaki
- Subjects
- *
PROTEASE-activated receptors , *THROMBIN receptors , *BLOOD platelet aggregation , *EXTRACELLULAR signal-regulated kinases , *CALCIUM ions - Abstract
Background Thrombin belongs to the most potent platelet agonists and activates human platelets through GPIbα and two protease activated receptors (PARs), PAR1 and PAR4. However, the details of thrombin receptor system, especially the role of PAR4 on human platelet activation is still not clear. Objectives We found a significant difference in PAR4-activating peptide (PAR4-AP)-induced, but not PAR1-AP, platelet aggregation between healthy Japanese subjects. Sequencing analysis revealed a single nucleotide change in PAR4 gene F2RL3 (SNP rs773902) leading to Ala120Thr variant. To elucidate the role of PAR4 in human platelet activation, we examined if platelet activation induced by PAR4-AP may be associated with PAR4 genotype. Methods Platelets from 202 healthy Japanese volunteers were genetically analyzed and determined the genotype frequency of rs773902. Agonist induced platelet aggregation, integrin αIIbβ3 activation, granule release, Ca 2+ mobilization, and activation of ERK and MLC were evaluated. The specificity of effects observed in platelets was confirmed in 293T cells transfected PAR4-Thr120 or Ala120. Results The frequencies of PAR4 variant Thr/Thr120, Ala/Thr120, and Ala/Ala 120 were 5.9, 37.1, and 57.0%, respectively. Platelets with Thr/Thr120 showed significantly higher reactivity in PAR4-AP-induced platelet aggregation, αIIbβ3 activation and granule release compared to platelets with Ala/Ala120. PAR4-AP induced higher Ca 2+ mobilization and ERK activation in platelets with Thr/Thr120 than Ala/Ala120. Ca 2+ mobilization and ERK activation were also increased in 293T cells transfected with PAR4-Thr120 compared to Ala120. Conclusion Our data suggested that PAR4-AP-induced platelet reactivity between PAR4 rs773902 was associated with altered intensity of Ca 2+ mobilization and ERK activation. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
38. Platelet function and microparticle levels in atrial fibrillation: Changes during the acute episode.
- Author
-
Pourtau, Line, Sellal, Jean Marc, Lacroix, Romaric, Poncelet, Philippe, Bernus, Olivier, Clofent-Sanchez, Gisèle, Hocini, Mélèze, Haïssaguerre, Michel, Dignat-George, Françoise, Sacher, Frédéric, and Nurden, Paquita
- Subjects
- *
ATRIAL fibrillation , *PLATELET function tests , *THROMBOSIS risk factors , *HEMOSTASIS , *FIBRINOLYTIC agents - Abstract
Background Thrombotic risk constitutes a major complication of atrial fibrillation (AF). Platelets and microparticles (MPs) are important for hemostasis and thrombosis, however their participation during AF is not well known. The aim of this study was to characterize platelet function and MPs procoagulant and fibrinolytic activity in AF patients and to determine the effects of an acute-AF episode. Methods Blood was collected from paroxysmal (21) and persistent (16) AF patients referred for AF catheter ablation. Ten patients in sinus rhythm for 10 days were induced in AF allowing comparisons of left atrium samples before and after induction. Platelet aggregation with ADP, TRAP, collagen, and ristocetin was studied. Platelet surface expression of PAR-1, αIIbβ3, GPIb and P-selectin were evaluated by flow cytometry, and MPs-associated procoagulant and fibrinolytic activity levels were determined by functional assays. Results A specific reduction in platelet aggregation to TRAP, activating the thrombin receptor PAR-1, was found in all AF patients. No differences in platelet receptor expression were found. Yet, after acute-induced AF, the platelet response was improved. Furthermore, a significant decrease of left atrium tissue factor-dependent procoagulant activity of MPs was observed. Conclusion Acute episodes of AF results in a decrease in MPs-associated tissue factor activity, possibly corresponding to consumption, which in turn favors coagulation and the local production of thrombin. A decreased platelet basal aggregation to TRAP may result from PAR1 desensitization, whereas the improved response after an induced episode of AF suggests activation of coagulation and PAR1 re-sensitization. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
39. On the Mechanism(s) of Thrombin Induced Angiogenesis
- Author
-
Maragoudakis, Michael E., Tsopanoglou, Nikos E., and Maragoudakis, Michael E., editor
- Published
- 2000
- Full Text
- View/download PDF
40. T
- Author
-
Bikfalvi, Andreas and Bikfalvi, Andreas
- Published
- 2000
- Full Text
- View/download PDF
41. Elevated expression of protease-activated receptor 1 via ΔNp63 down-regulation contributes to nodal metastasis in oral squamous cell carcinoma
- Author
-
M. Hiwatashi, Eiki Hamada, Naoki Kaneko, Seiji Nakamura, Taiji Sakamoto, Ryoji Kitamura, Yurie Mikami, Taichi Hattori, Shintarou Kawano, Shoichi Tanaka, Yasuyuki Maruse, Kazunari Oobu, Ryota Matsubara, Yuma Hashiguchi, Tamotsu Kiyoshima, and Masahiko Morioka
- Subjects
Stromal cell ,Down-Regulation ,Metastasis ,03 medical and health sciences ,0302 clinical medicine ,Thrombin receptor ,Humans ,Medicine ,Receptor, PAR-1 ,Receptor ,Survival rate ,Retrospective Studies ,Predictive marker ,Squamous Cell Carcinoma of Head and Neck ,business.industry ,Cancer ,030206 dentistry ,medicine.disease ,Otorhinolaryngology ,Head and Neck Neoplasms ,030220 oncology & carcinogenesis ,Cancer cell ,Carcinoma, Squamous Cell ,cardiovascular system ,Cancer research ,Mouth Neoplasms ,Surgery ,Oral Surgery ,business - Abstract
Protease-activated receptor 1 (PAR1) is known as a thrombin receptor. Recent studies have reported PAR1 expression in various malignancies; however, its role in oral squamous cell carcinoma (OSCC) requires clarification. A previous study showed that down-regulation of ΔNp63, a homolog of p53, augments PAR1 expression in OSCC. In the present study, the association of PAR1 expression with clinicopathological findings in OSCC was examined retrospectively. Expression of PAR1, thrombin, and ΔNp63 was examined immunohistochemically in OSCC specimens. Patients were divided into three groups based on the expression pattern of PAR1 at the invasive front: group A, PAR1-negative in both cancer and stromal cells; group B, positive in stromal cells but negative in cancer cells; group C, positive in both cancer and stromal cells. Histologically high-grade tumours were significantly more common in group C. Patients in group C had the highest incidence rate of nodal metastasis (P
- Published
- 2021
- Full Text
- View/download PDF
42. Monitoring Platelet Aggregation
- Author
-
Kleiman, Neal S., Mazur, Wojciech, Graziadei, Nikki, Cannon, Christopher P., editor, Lincoff, A. Michael, editor, and Topol, Eric J., editor
- Published
- 1999
- Full Text
- View/download PDF
43. The Coagulation Pathway and Antithrombotic Strategies
- Author
-
Leblond, L., Winocour, P. D., Uprichard, Andrew C. G., editor, and Gallagher, Kim P., editor
- Published
- 1999
- Full Text
- View/download PDF
44. Human Platelet Thrombin Receptors and the Two Receptor Model for Platelet Activation
- Author
-
Jamieson, G. A., Tandon, Narendra N., Greco, Nicholas J., and Rao, Gundu H. R., editor
- Published
- 1999
- Full Text
- View/download PDF
45. Protease-activated receptor 1 as a potential therapeutic target for COVID-19
- Author
-
Marinella Holzhausen, Tomaz Alves, and Emanuel da Silva Rovai
- Subjects
0301 basic medicine ,coronavirus ,Receptors, Cell Surface ,Inflammation ,Disease ,030204 cardiovascular system & hematology ,General Biochemistry, Genetics and Molecular Biology ,Fibrin ,coagulopathy ,Proinflammatory cytokine ,03 medical and health sciences ,0302 clinical medicine ,Thrombin ,Protease-activated receptor 1 ,Thrombin receptor ,medicine ,Humans ,Receptor, PAR-1 ,anticoagulation ,Blood Coagulation ,Venous Thrombosis ,biology ,SARS-CoV-2 ,business.industry ,COVID-19 ,Anticoagulants ,Disseminated Intravascular Coagulation ,medicine.disease ,Blood Coagulation Factors ,COVID-19 Drug Treatment ,Venous thrombosis ,030104 developmental biology ,Coagulation ,Immunology ,biology.protein ,proteases ,Minireview ,medicine.symptom ,Pulmonary Embolism ,business ,medicine.drug - Abstract
Acute respiratory disease caused by a novel coronavirus (SARS-CoV-2) has spread all over the world, since its discovery in 2019, Wuhan, China. This disease is called COVID-19 and already killed over 1 million people worldwide. The clinical symptoms include fever, dry cough, dyspnea, headache, dizziness, generalized weakness, vomiting, and diarrhea. Unfortunately, so far, there is no validated vaccine, and its management consists mainly of supportive care. Venous thrombosis and pulmonary embolism are highly prevalent in patients suffering from severe COVID-19. In fact, a prothrombotic state seems to be present in most fatal cases of the disease. SARS-CoV-2 leads to the production of proinflammatory cytokines, causing immune-mediated tissue damage, disruption of the endothelial barrier, and uncontrolled thrombogenesis. Thrombin is the key regulator of coagulation and fibrin formation. In severe COVID-19, a dysfunctional of physiological anticoagulant mechanisms leads to a progressive increase of thrombin activity, which is associated with acute respiratory distress syndrome development and a poor prognosis. Protease-activated receptor type 1 (PAR1) is the main thrombin receptor and may represent an essential link between coagulation and inflammation in the pathophysiology of COVID-19. In this review, we discuss the potential role of PAR1 inhibition and regulation in COVID-19 treatment.
- Published
- 2020
- Full Text
- View/download PDF
46. Protease‐activated receptors: An illustrated review
- Author
-
Xu Han, Bryce A. Kerlin, and Marvin T. Nieman
- Subjects
Illustrated Review ,Cell type ,Proteases ,protease‐activated receptors ,lcsh:RC633-647.5 ,G‐protein–coupled receptors ,Cell ,antithrombotic therapies ,Hematology ,lcsh:Diseases of the blood and blood-forming organs ,Biology ,Endocytosis ,Cell biology ,medicine.anatomical_structure ,Thrombin ,Thrombin receptor ,platelets ,medicine ,Receptor ,signaling ,thrombosis ,G protein-coupled receptor ,medicine.drug - Abstract
Proteases are important regulators of cell behavior, survival, and apoptosis. They communicate to cells directly through a special class of G‐protein–coupled receptors known as protease‐activated receptors (PARs). N‐terminal PAR proteolysis unmasks a neo‐N‐terminus, which serves as a tethered ligand to activate PARs. Using this unique irreversible activation mechanism, PARs relay information across cell membranes. The year 2020 is the 30th year since discovery of the first member of this family, PAR1. In this illustrated review, we highlight achievements in the PAR field over the past 3 decades. Additionally, the known expression profiles of PARs in human tissues and across species are portrayed. We also illustrate the tethered ligand activation mechanism, which is unique to PARs, and PAR regulatory mechanisms. PAR1 was originally named “thrombin receptor” because thrombin was the first protease identified to activate PAR1. However, over the past 30 years, a growing number of proteases have been found to cleave PARs and trigger differential downstream signaling depending on cleavage site, cell type, and species. We exemplify the diversity of PAR1‐mediated signaling outcomes in platelets and endothelial cells as pertinent examples to the hemostasis, thrombosis, and vascular biology fields. Further, the termination and regulation of PAR signaling via endocytosis and currently available pharmacologic approaches are depicted. We conclude with portrayal of clinically translational aspects of PAR biology including pharmacologic manipulation and single‐nucleotide polymorphisms.
- Published
- 2020
47. Flow Cytometric Assessment of AKT Signaling in Platelet Activation: An Alternative Diagnostic Tool for Small Volumes of Blood
- Author
-
Irene Marini, Lisann Pelzl, Karina Althaus, Tamam Bakchoul, and M. Wagner
- Subjects
Blood Platelets ,0301 basic medicine ,030204 cardiovascular system & hematology ,Pharmacology ,Flow cytometry ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Thrombin receptor ,medicine ,Humans ,Platelet ,Platelet activation ,Protein kinase B ,medicine.diagnostic_test ,Chemistry ,Hematology ,Flow Cytometry ,Platelet Activation ,Healthy Volunteers ,Adenosine diphosphate ,030104 developmental biology ,Hemostasis ,Phosphorylation ,Proto-Oncogene Proteins c-akt ,Signal Transduction - Abstract
Introduction The diagnosis of platelet function disorder in children is challenging. Light transmission aggregometry is the gold standard for platelet function disorders. However, large blood volumes are required. Currently, there are no existing tools for the diagnosis of platelet function disorders that use small blood volumes. AKT signaling plays a central role in platelet activation during hemostasis and might be visualized by flow cytometry. Methods Platelet-rich plasma obtained by centrifugation of citrated blood from healthy volunteers was activated with arachidonic acid, thrombin receptor activating peptide-6 (TRAP-6), collagen, adenosine diphosphate ADP, collagen-related peptide (CRP), and epinephrine. After platelet activation, the phosphorylation of AKT was assessed by flow cytometer using a Navios cytometer. Results Healthy volunteers showed a reproducible phosphorylation of AKT upon activation. In comparison to nonactivated platelets, we documented an increase in pAKT expression with all agonists. Especially TRAP-6 and CRP caused considerable increase in percentage of pAKT expression throughout all the tested healthy volunteers. Conclusion An activation of the AKT-signal pathway by different agonists can clearly be detected on the flow cytometer, indicating that the visualization of signaling in platelets by flow cytometry might be an efficient alternative for light transmission aggregometry to test platelet function in children.
- Published
- 2020
- Full Text
- View/download PDF
48. Podocyte Integrin-β 3 and Activated Protein C Coordinately Restrict RhoA Signaling and Ameliorate Diabetic Nephropathy
- Author
-
Silke Zimmermann, Alireza R. Rezaie, Marcus J. Moeller, Sumra Nazir, Liliana Schaefer, Shruthi Krishnan, Shrey Kohli, Ihsan Gadi, Wei Dong, Sanchita Ghosh, Rajiv Rana, Wolfram Ruf, Dheerendra Gupta, Thati Madhusudhan, Jochen Reiser, Stoyan Stoyanov, Hongjie Wang, Jinyang Zeng-Brouwers, Charles T. Esmon, Ronald Biemann, Berend Isermann, Moh'd Mohanad Al-Dabet, Ahmed Elwakiel, and Akash Mathew
- Subjects
0301 basic medicine ,RHOA ,physiology [Podocytes] ,physiology [Protein C] ,Protein subunit ,Integrin ,prevention & control [Diabetic Nephropathies] ,030204 cardiovascular system & hematology ,GTP-Binding Protein alpha Subunits, G12-G13 ,Podocyte ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Thrombin ,Thrombin receptor ,medicine ,Animals ,Humans ,Diabetic Nephropathies ,Receptor, PAR-1 ,ddc:610 ,physiology [Endothelial Protein C Receptor] ,physiology [Receptor, PAR-1] ,Receptor ,physiology [GTP-Binding Protein alpha Subunits, G12-G13] ,biology ,Podocytes ,Chemistry ,physiology [rhoA GTP-Binding Protein] ,Integrin beta3 ,Endothelial Protein C Receptor ,General Medicine ,physiology [Integrin beta3] ,Cell biology ,Mice, Inbred C57BL ,Basic Research ,HEK293 Cells ,030104 developmental biology ,medicine.anatomical_structure ,Cytoprotection ,Nephrology ,biology.protein ,rhoA GTP-Binding Protein ,Protein C ,medicine.drug - Abstract
BACKGROUND: Diabetic nephropathy (dNP), now the leading cause of ESKD, lacks efficient therapies. Coagulation protease–dependent signaling modulates dNP, in part via the G protein–coupled, protease-activated receptors (PARs). Specifically, the cytoprotective protease-activated protein C (aPC) protects from dNP, but the mechanisms are not clear. METHODS: A combination of in vitro approaches and mouse models evaluated the role of aPC-integrin interaction and related signaling in dNP. RESULTS: The zymogen protein C and aPC bind to podocyte integrin-β(3), a subunit of integrin-α(v)β(3). Deficiency of this integrin impairs thrombin-mediated generation of aPC on podocytes. The interaction of aPC with integrin-α(v)β(3) induces transient binding of integrin-β(3) with G(α13) and controls PAR-dependent RhoA signaling in podocytes. Binding of aPC to integrin-β(3) via its RGD sequence is required for the temporal restriction of RhoA signaling in podocytes. In podocytes lacking integrin-β(3), aPC induces sustained RhoA activation, mimicking the effect of thrombin. In vivo, overexpression of wild-type aPC suppresses pathologic renal RhoA activation and protects against dNP. Disrupting the aPC–integrin-β(3) interaction by specifically deleting podocyte integrin-β(3) or by abolishing aPC’s integrin-binding RGD sequence enhances RhoA signaling in mice with high aPC levels and abolishes aPC’s nephroprotective effect. Pharmacologic inhibition of PAR1, the pivotal thrombin receptor, restricts RhoA activation and nephroprotects RGE-aPC(high) and wild-type mice. Conclusions aPC–integrin-α(v)β(3) acts as a rheostat, controlling PAR1-dependent RhoA activation in podocytes in diabetic nephropathy. These results identify integrin-α(v)β(3) as an essential coreceptor for aPC that is required for nephroprotective aPC-PAR signaling in dNP.
- Published
- 2020
- Full Text
- View/download PDF
49. Good Platelets Gone Bad: The Effects of Trauma Patient Plasma on Healthy Platelet Aggregation
- Author
-
Alexander T. Fields, Rachael A. Callcut, Lucy Z. Kornblith, Zachary A. Matthay, Brenda Nunez-Garcia, Roland J. Bainton, and Ellicott C. Matthay
- Subjects
Blood Platelets ,Adult ,Male ,medicine.medical_specialty ,Physical Injury - Accidents and Adverse Effects ,Platelet Aggregation ,Clinical Sciences ,030204 cardiovascular system & hematology ,Cardiovascular ,Critical Care and Intensive Care Medicine ,Article ,Plasma ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Thrombin ,platelet function tests ,Clinical Research ,Internal medicine ,Thrombin receptor ,medicine ,Coagulopathy ,Humans ,Platelet ,platelet adhesiveness ,business.industry ,Shock ,030208 emergency & critical care medicine ,Hematology ,Blood Coagulation Disorders ,Middle Aged ,medicine.disease ,thrombin ,Emergency & Critical Care Medicine ,Pathophysiology ,trauma ,Endocrinology ,Shock (circulatory) ,Emergency Medicine ,Wounds and Injuries ,Injury Severity Score ,Base excess ,Platelet activation ,medicine.symptom ,business ,medicine.drug - Abstract
BackgroundAltered postinjury platelet behavior is recognized in the pathophysiology of trauma-induced coagulopathy (TIC), but the mechanisms remain largely undefined. Studies suggest that soluble factors released by injury may inhibit signaling pathways and induce structural changes in circulating platelets. Given this, we sought to examine the impact of treating healthy platelets with plasma from injured patients. We hypothesized that healthy platelets treated ex-vivo with plasma from injured patients with shock would impair platelet aggregation, while treatment with plasma from injured patients with significant injury burden, but without shock, would enhance platelet aggregation.MethodsPlasma samples were isolated from injured patients (pretransfusion) and healthy donors at a Level I trauma center and stored at -80°C. Plasma samples from four separate patients in each of the following stratified clinical groups were used: mild injury/no shock (injury severity score [ISS] 2-15, base excess [BE]>-6), mild injury/with shock (ISS 2-15, BE≤-6), severe injury/no shock (ISS>25, BE>-6), severe injury/with shock (ISS>25, BE≤-6), minimal injury (ISS 0/1, BE>-6), and healthy. Platelets were isolated from three healthy adult males and were treated with plasma for 30 min. Aggregation was stimulated with a thrombin receptor agonist and measured via multiple-electrode platelet aggregometry. Data were normalized to HEPES Tyrode's (HT) buffer-only treated platelets. Associations of plasma treatment groups with platelet aggregation measures were tested with Mann-Whitney U tests.ResultsPlatelets treated with plasma from patients with shock (regardless of degree of injury) had significantly impaired thrombin-stimulated aggregation compared with platelets treated with plasma from patients without shock (P = 0.002). Conversely, platelets treated with plasma from patients with severe injury, but without shock, had amplified thrombin-stimulated aggregation (P = 0.030).ConclusionShock-mediated soluble factors impair platelet aggregation, and tissue injury-mediated soluble factors amplify platelet aggregation. Future characterization of these soluble factors will support development of novel treatments of TIC.
- Published
- 2020
- Full Text
- View/download PDF
50. Coagulation factor 2 thrombin receptor promotes malignancy in glioma under SOX2 regulation
- Author
-
Xiaoling Zhang, Baozhe Jin, Ming Yang, Ge Dang, Xiang-Yang Wang, Jing Zhao, Guojun Gao, and Fan Wang
- Subjects
Aging ,Epithelial-Mesenchymal Transition ,proliferation ,SOX2 ,Datasets as Topic ,Kaplan-Meier Estimate ,Metastasis ,Downregulation and upregulation ,Cell Line, Tumor ,Glioma ,Thrombin receptor ,medicine ,metastasis ,Humans ,F2R ,Receptor, PAR-1 ,Wnt Signaling Pathway ,Cell Proliferation ,Brain Neoplasms ,Chemistry ,SOXB1 Transcription Factors ,Wnt signaling pathway ,Brain ,Cell Biology ,Prognosis ,medicine.disease ,Xenograft Model Antitumor Assays ,Up-Regulation ,Gene Expression Regulation, Neoplastic ,Gene Knockdown Techniques ,Cancer research ,Female ,Signal transduction ,Chromatin immunoprecipitation ,Research Paper - Abstract
Glioma is the most common human primary brain cancer with high mortality and unfavorable clinical outcome. Coagulation factor 2 thrombin receptor (F2R), is a key component in the thrombosis process and has been demonstrated upregulated in various cancers. However, the effect and molecular mechanisms of F2R in glioma remains unclear. In our study, we confirmed that the expression of F2R was upregulated in glioma and predicted poor prognosis. Gene Set Enrichment Analysis (GSEA) and function assays demonstrated that F2R overexpression promoted glioma cell proliferation, metastasis and epithelial-mesenchymal transition (EMT) in vitro and tumor growth in vivo. Then, we identified and validated F2R was the target gene of SRY-box 2 (SOX2) by dual luciferase reporter assay and chromatin immunoprecipitation assay. Besides, High expression of F2R in malignant glioma was associated with β-catenint signaling pathway activation. Our findings conclude that F2R promotes glioma cell proliferation and metastasis under SOX2 and actives WNT/β-catenin Signaling pathway, which provides novel insight to the therapeutic regimen in glioma.
- Published
- 2020
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.