Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Thromb Haemost ; 22(7): 2039-2051, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38670314

RESUMEN

BACKGROUND: Activated protein C (APC) has anticoagulant and cytoprotective cell-signaling activities, which often require protease-activated receptor (PAR) 1 and PAR3 and PAR cleavages at noncanonical sites (R46-N47 and R41-G42, respectively). Some PAR1-derived (P1) peptides and PAR3-derived (P3) peptides, eg, P1-47-66 and P3-42-65, mimic APC's cell signaling. In anti-inflammatory assays, these 2 peptides at low concentrations synergistically attenuate cellular inflammation. OBJECTIVES: To determine whether a P1 peptide covalently linked to a P3 peptide mimics APC's anti-inflammatory and endothelial barrier stabilization activities. METHODS: Anti-inflammatory assays employed stimulated THP-1 cells and caspase-1 measurements. Cultured human EA.hy926 or murine aortic endothelial cells (ECs) exposed to thrombin were monitored for transendothelial electrical resistance. Bivalent covalently linked P1:P3 peptides were studied for APC-like activities. RESULTS: In anti-inflammatory assays, P1-47-55 was as active as P1-47-66 and some P3 peptides (eg, P3-44-54 and P3-51-65) were as active as P3-42-65. The bivalent P1:P3 peptide comprising P1-47-55-(Gly[10 residues])-P3-51-65 (designated "G10 peptide") was more potently anti-inflammatory than the P1 or P3 peptide alone. In transendothelial electrical resistance studies of thrombin-challenged ECs, P1-47-55 and the G10 peptide mimicked APC's protective actions. In dose-response studies, the G10 peptide was more potent than the P1-47-55 peptide. In murine EC studies, the murine PAR-sequence-derived G10 peptide mimicked murine APC's activity. Anti-PAR1 and anti-PAR3 antibodies, but not anti-endothelial protein C receptor antibodies, abated G10's cytoprotection, showing that G10's actions involve PAR1:PAR3. G10 significantly increased survival in murine endotoxemia. CONCLUSION: The PAR-sequence-derived G10 peptide is a bivalent agonist that mimics APC's cytoprotective, anti-inflammatory, and endothelial barrier-stabilizing actions and APC's protection against endotoxemic mortality.


Asunto(s)
Células Endoteliales , Proteína C , Receptor PAR-1 , Proteína C/metabolismo , Proteína C/química , Humanos , Animales , Receptor PAR-1/agonistas , Receptor PAR-1/metabolismo , Células Endoteliales/metabolismo , Células Endoteliales/efectos de los fármacos , Ratones , Antiinflamatorios/farmacología , Antiinflamatorios/química , Ratones Endogámicos C57BL , Células THP-1 , Trombina/metabolismo , Receptor de Proteína C Endotelial/metabolismo , Receptores de Trombina/agonistas , Receptores de Trombina/metabolismo , Transducción de Señal , Receptores Proteinasa-Activados/agonistas , Receptores Proteinasa-Activados/metabolismo , Péptidos/farmacología , Péptidos/química , Endotoxemia/tratamiento farmacológico , Endotoxemia/metabolismo , Fragmentos de Péptidos/farmacología , Masculino , Modelos Animales de Enfermedad
2.
J Biol Chem ; 300(2): 105614, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38159863

RESUMEN

The activation and mobilization of immune cells play a crucial role in immunotherapy. Existing therapeutic interventions, such as cytokines administration, aim to enhance immune cell activity. However, these approaches usually result in modest effectiveness and toxic side effects, thereby restricting their clinical application. Protease-activated receptors (PARs), a subfamily of G protein-coupled receptors, actively participate in the immune system by directly activating immune cells. The activation of PARs by proteases or synthetic ligands can modulate immune cell behavior, signaling, and responses to treat immune-related diseases, suggesting the significance of PARs agonism in immunotherapy. However, the agonism of PARs in therapeutical applications remains rarely discussed, since it has been traditionally considered that PARs activation facilitates disease progressions. This review aims to comprehensively summarize the activation, rather than inhibition, of PARs in immune-related physiological responses and diseases. Additionally, we will discuss the emerging immunotherapeutic potential of PARs agonism, providing a new strategic direction for PARs-mediated immunotherapy.


Asunto(s)
Inmunoterapia , Receptores Proteinasa-Activados , Péptido Hidrolasas/metabolismo , Receptores Acoplados a Proteínas G , Receptores Proteinasa-Activados/agonistas , Receptores Proteinasa-Activados/metabolismo , Transducción de Señal , Neoplasias/inmunología , Neoplasias/terapia , Enfermedades del Sistema Inmune/inmunología , Enfermedades del Sistema Inmune/terapia , Humanos , Animales
3.
Thromb Haemost ; 121(10): 1337-1344, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33690868

RESUMEN

Platelet activation is characterized by shape change, granule secretion, activation of fibrinogen receptor (glycoprotein IIb/IIIa) sustaining platelet aggregation, and externalization of negatively charged aminophospholipids contributing to platelet procoagulant activity. Epinephrine (EPI) alone is a weak platelet activator. However, it is able to potentiate platelet activation initiated by other agonists. In this work, we investigated the role of EPI in the generation of procoagulant platelets. Human platelets were activated with convulxin (CVX), thrombin (THR) or protease-activated receptor (PAR) agonists, EPI, and combination thereof. Platelet aggregation was assessed by light transmission aggregometry or with PAC-1 binding by flow cytometry. Procoagulant collagen-and-THR (COAT) platelets, induced by combined activation with CVX-and-THR, were visualized by flow cytometry as Annexin-V-positive and PAC-1-negative platelets. Cytosolic calcium fluxes were monitored by flow cytometry using Fluo-3 indicator. EPI increased platelet aggregation induced by all agonist combinations tested. On the other hand, EPI dose-dependently reduced the formation of procoagulant COAT platelets generated by combined CVX-and-THR activation. We observed a decreased Annexin-V-positivity and increased binding of PAC-1 with the triple activation (CVX + THR + EPI) compared with CVX + THR. Calcium mobilization with triple activation was decreased with the higher EPI dose (1,000 µM) compared with CVX + THR calcium kinetics. In conclusion, when platelets are activated with CVX-and-THR, the addition of increasing concentrations of EPI (triple stimulation) modulates platelet response reducing cytosolic calcium mobilization, decreasing procoagulant activity, and enhancing platelet aggregation.


Asunto(s)
Coagulación Sanguínea/efectos de los fármacos , Plaquetas/efectos de los fármacos , Coagulantes/farmacología , Epinefrina/farmacología , Agregación Plaquetaria/efectos de los fármacos , Adolescente , Adulto , Anciano , Plaquetas/metabolismo , Señalización del Calcio , Venenos de Crotálidos/farmacología , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Cinética , Lectinas Tipo C , Masculino , Persona de Mediana Edad , Glicoproteínas de Membrana Plaquetaria/agonistas , Glicoproteínas de Membrana Plaquetaria/metabolismo , Receptores Proteinasa-Activados/agonistas , Receptores Proteinasa-Activados/metabolismo , Trombina/farmacología , Adulto Joven
4.
J Leukoc Biol ; 105(4): 729-740, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30690783

RESUMEN

Tissue-type plasminogen activator (tPA) activates fibrinolysis and also suppresses innate immune system responses to LPS in bone marrow-derived macrophages (BMDMs) and in vivo in mice. The objective of this study was to assess the activity of tPA as a regulator of macrophage physiology in the presence of plasmin. Enzymatically active and enzymatically inactive (EI) tPA appeared to comprehensively block the response to LPS in BMDMs, including expression of proinflammatory cytokines such as TNF-α and IL-1ß and anti-inflammatory cytokines such as IL-10 and IL-1 receptor antagonist. The activity of EI-tPA as an LPS response modifier was conserved in the presence of plasminogen. By contrast, in BMDMs treated with tPA and plasminogen or preactivated plasmin, in the presence or absence of LPS, increased proinflammatory cytokine expression was observed and tPA failed to reverse the response. Plasmin independently activated NF-κB, ERK1/2, c-Jun N-terminal kinase, and p38 mitogen-activated protein kinase in BMDMs, which is characteristic of proinflammatory stimuli. Plasmin-induced cytokine expression was blocked by ε-aminocaproic acid, aprotinin, and inhibitors of the known plasmin substrate, Protease-activated receptor-1 (PAR-1), but not by N-methyl-d-aspartate receptor inhibitor, which blocks the effects of tPA on macrophages. Cytokine expression by BMDMs treated with the PAR-1 agonist, TFLLR, was not inhibited by EI-tPA, possibly explaining why EI-tPA does not inhibit macrophage responses to plasmin and providing evidence for specificity in the ability of tPA to oppose proinflammatory stimuli. Regulation of innate immunity by the fibrinolysis system may reflect the nature of the stimulus and a balance between the potentially opposing activities of tPA and plasmin.


Asunto(s)
Fibrinolisina/metabolismo , Inflamación/patología , Lipopolisacáridos/metabolismo , Receptores Proteinasa-Activados/metabolismo , Activador de Tejido Plasminógeno/metabolismo , Animales , Células CHO , Cricetinae , Cricetulus , Citocinas/metabolismo , Humanos , Mediadores de Inflamación/metabolismo , Masculino , Ratones Endogámicos C57BL , Péptidos/farmacología , Receptores Proteinasa-Activados/agonistas , Receptores Proteinasa-Activados/antagonistas & inhibidores , Transducción de Señal
5.
Physiol Rep ; 5(24)2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29263120

RESUMEN

Macrophage migration inhibitory factor (MIF) mediates pain although the mechanisms are not well understood. Urothelial activation of protease activated receptor 4 (PAR4) results in urothelial MIF release, urothelial high mobility group box 1 (HMGB1) release and bladder pain in mice without bladder inflammation. All three effects are prevented by MIF inhibition while intravesical disulfide HMGB1 alone can induce bladder pain. This study utilizes genetic MIF deletion to determine whether MIF mediates PAR4-induced bladder pain and is upstream of HMGB1-induced bladder pain. Wild type (C57/BL6) and MIF knockout (KO) mice were treated with intravesical PAR4 activating peptide or disulfide HMGB1 and tested for abdominal mechanical hypersensitivity at baseline (before treatment) and 24 h after injection. Micturition parameters and bladder histology were examined after behavioral test. Real-time PCR and western blotting measured HMGB1 mRNA and protein levels in the bladders of naïve wild type and MIF KO mice, while immunofluorescence measured HMGB1 protein levels in the urothelium of both strains. Intravesical PAR4 activation resulted in abdominal mechanical hypersensitivity in wild-type mice but not MIF KO mice. Intravesical disulfide HMGB1 induced abdominal mechanical hypersensitivity in both strains. Neither treatment resulted in significant changes in micturition or bladder histology in either strain. HMGB1 mRNA and protein levels were higher in MIF KO mouse bladders and the urothelium of MIF KO bladder had greater immunostaining than the wild-type strain. MIF is a pivotal molecule mediating PAR4-induced bladder pain and regulating urothelial HMGB1 production and release to elicit bladder pain.


Asunto(s)
Hiperalgesia/metabolismo , Oxidorreductasas Intramoleculares/genética , Factores Inhibidores de la Migración de Macrófagos/genética , Vejiga Urinaria/metabolismo , Urotelio/metabolismo , Animales , Proteína HMGB1/genética , Proteína HMGB1/metabolismo , Hiperalgesia/etiología , Oxidorreductasas Intramoleculares/metabolismo , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Oligopéptidos/toxicidad , Receptores Proteinasa-Activados/agonistas , Tacto
6.
J Immunol Res ; 2017: 5193572, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28503577

RESUMEN

Proteinase-activated receptors 1 (PAR1) and 2 (PAR2) are the most highly expressed members of the PAR family in the periodontium. These receptors regulate periodontal inflammatory and repair processes through their activation by endogenous and bacterial enzymes. PAR1 is expressed by the periodontal cells such as human gingival fibroblasts, gingival epithelial cells, periodontal ligament cells, osteoblasts, and monocytic cells and can be activated by thrombin, matrix metalloproteinase 1 (MMP-1), MMP-13, fibrin, and gingipains from Porphyromonas gingivalis. PAR2 is expressed by neutrophils, osteoblasts, oral epithelial cells, and human gingival fibroblasts, and its possible activators in the periodontium are gingipains, neutrophil proteinase 3, and mast cell tryptase. The mechanisms through which PARs can respond to periodontal enzymes and result in appropriate immune responses have until recently been poorly understood. This review discusses recent findings that are beginning to identify a cardinal role for PAR1 and PAR2 on periodontal tissue metabolism.


Asunto(s)
Periodontitis/metabolismo , Periodontitis/fisiopatología , Periodoncio/metabolismo , Receptor PAR-1/metabolismo , Receptores Proteinasa-Activados/metabolismo , Adhesinas Bacterianas/metabolismo , Animales , Células Cultivadas , Cisteína Endopeptidasas/metabolismo , Células Epiteliales , Fibroblastos , Regulación de la Expresión Génica , Cisteína-Endopeptidasas Gingipaínas , Encía/citología , Encía/metabolismo , Humanos , Metaloproteinasa 1 de la Matriz/genética , Metaloproteinasa 1 de la Matriz/metabolismo , Ratones , Periodontitis/genética , Porphyromonas gingivalis , Receptor PAR-1/agonistas , Receptor PAR-1/antagonistas & inhibidores , Receptor PAR-1/genética , Receptores Proteinasa-Activados/agonistas , Receptores Proteinasa-Activados/antagonistas & inhibidores , Receptores Proteinasa-Activados/genética
7.
FEBS Lett ; 588(24): 4604-12, 2014 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-25447533

RESUMEN

Phospholipase A2 (PLA2) is an important component in snake venoms. Here, an acidic PLA2, designated PA2-Vb was isolated from the Trimeresurus stejnegeri snake venom. PA2-Vb acts on a protease-activated receptor (PAR-1) to evoke Ca(2+) release through the inositol 1,4,5-trisphosphate receptor (IP3R) and induces mouse aorta contraction. PAR-1, phospholipase C and IP3R inhibitors suppressed PA2-Vb-induced aorta contraction. The crystal structure reveals that PA2-Vb has the typical fold of most snake venom PLA2. Several PEG molecules bond to a positively charged pocket. The finding offers a novel pharmacological basis of the structure for investigating the PAR-1 receptor and suggests potential applications for PA2-Vb in the vascular system.


Asunto(s)
Venenos de Crotálidos/enzimología , Fosfolipasas A2/química , Fosfolipasas A2/farmacología , Receptores Proteinasa-Activados/agonistas , Trimeresurus , Secuencia de Aminoácidos , Animales , Calcio/metabolismo , Cristalografía por Rayos X , Concentración de Iones de Hidrógeno , Modelos Moleculares , Datos de Secuencia Molecular , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/fisiología , Fosfolipasas A2/aislamiento & purificación , Multimerización de Proteína , Estructura Cuaternaria de Proteína , Vasoconstricción/efectos de los fármacos
8.
Int J Mol Sci ; 15(4): 6169-83, 2014 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-24733067

RESUMEN

Protease-activated receptors (PARs) are a family of four G protein-coupled receptors that exhibit increasingly appreciated differences in signaling and regulation both within and between the receptor class. By nature of their proteolytic self-activation mechanism, PARs have unique processes of receptor activation, "ligand" binding, and desensitization/resensitization. These distinctive aspects have presented both challenges and opportunities in the targeting of PARs for therapeutic benefit-the most notable example of which is inhibition of PAR1 on platelets for the prevention of arterial thrombosis. However, more recent studies have uncovered further distinguishing features of PAR-mediated signaling, revealing mechanisms by which identical proteases elicit distinct effects in the same cell, as well as how distinct proteases produce different cellular consequences via the same receptor. Here we review this differential signaling by PARs, highlight how important distinctions between PAR1 and PAR4 are impacting on the progress of a new class of anti-thrombotic drugs, and discuss how these more recent insights into PAR signaling may present further opportunities for manipulating PAR activation and signaling in the development of novel therapies.


Asunto(s)
Receptores Proteinasa-Activados/metabolismo , Plaquetas/efectos de los fármacos , Plaquetas/metabolismo , Dimerización , Fibrinolíticos/uso terapéutico , Humanos , Proteína C/metabolismo , Receptores Proteinasa-Activados/agonistas , Receptores Proteinasa-Activados/antagonistas & inhibidores , Receptores de Trombina/antagonistas & inhibidores , Receptores de Trombina/metabolismo , Sepsis/terapia , Transducción de Señal , Trombosis/prevención & control , Trombosis/terapia
9.
Mediators Inflamm ; 2013: 165453, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24453410

RESUMEN

Serine proteinases have been recognized as playing an important role in inflammation via proteinase activated receptors (PARs). However, little is known about the influence of serine proteinases and PARs on TNF secretion from highly purified T cells. We challenged T cells from human peripheral blood with serine proteinases and agonist peptides of PARs and measured the levels of TNF in culture supernatants by ELISA. The results showed that thrombin and trypsin, but not tryptase, stimulated approximately up to 2.5-fold increase in TNF release from T cells following 16 h incubation. Proteinase inhibitors and PAR-1 antagonist SCH 79797 almost completely abolished thrombin- and trypsin-induced TNF release from T cells. Agonist peptides of PAR-1, but not PAR-2 induced TNF release from T cells. Moreover, trypsin- and thrombin-induced upregulated expression of TNF was observed in CD4+, IL-4+, or CD25+ T cells, but not in IFN+ or IL-17+ T cells. The signaling pathways MAPK/ERK and PI3K/Akt are involved in the thrombin- and trypsin-induced TNF release from T cells. In conclusion, thrombin and trypsin can induce TNF release from IL-4+ and CD25+ T cells through activation of PAR-1 and therefore contribute to regulation of immune response and inflammation of the body.


Asunto(s)
Receptores Proteinasa-Activados/agonistas , Subgrupos de Linfocitos T/inmunología , Factor de Necrosis Tumoral alfa/biosíntesis , Cromonas/farmacología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Flavonoides/farmacología , Humanos , Morfolinas/farmacología , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Trombina/farmacología , Tripsina/farmacología , Factor de Necrosis Tumoral alfa/genética
10.
Genes Dev ; 26(19): 2138-43, 2012 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-22972936

RESUMEN

The Hippo signaling pathway plays a crucial role in tissue growth and tumorigenesis. Core components of the Hippo pathway include the MST1/2 and Lats1/2 kinases. Acting downstream from the Hippo pathway are the YAP/TAZ transcription coactivators, which are inhibited through phosphorylation by Lats. However, upstream signals that regulate the Hippo pathway have not been well delineated. Here we report that stimulation of protease-activated receptors (PARs) activates YAP/TAZ by decreasing phosphorylation and increasing nuclear localization. PAR1 acts through G(12/13) and Rho GTPase to inhibit the Lats1/2 kinase. Our observations establish thrombin as a physiological signal for the Hippo pathway and implicate Hippo-YAP as a key downstream signaling branch of PAR activation.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Nucleares/metabolismo , Receptores Proteinasa-Activados/metabolismo , Factores de Transcripción/metabolismo , Citoesqueleto de Actina , Aciltransferasas , Proteínas de Ciclo Celular , Línea Celular Tumoral , Núcleo Celular/enzimología , Activación Enzimática/fisiología , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas Nucleares/genética , Oligopéptidos/metabolismo , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Transporte de Proteínas , ARN Interferente Pequeño/metabolismo , Receptores Proteinasa-Activados/agonistas , Factores de Transcripción/genética
11.
J Pharmacol Sci ; 119(1): 40-51, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22641126

RESUMEN

The present study was undertaken to investigate the function of protease-activated receptor (PAR) in endometriotic lesions using a mouse model of endometriosis. Unilateral ovariectomy (uOVX) was performed on female nude mice followed by intraperitoneal transplantation of a suspension mixture of immortalized human endometrial epithelial cells (EM-1) and stromal cells (EtsT-499). Endometriosis-like lesions were observed mostly around the dissection site after transplantation. The expression of interleukin (IL)-6 and cyclooxygenase-2 in the lesions was enhanced in uOVX mice compared to non-uOVX animals. In non-uOVX mice, IL-6 mRNA levels were higher in lesions formed with cells that were pretreated with PAR1/2 agonists (thrombin, 10 U/ml and PAR2-activating peptide, 30 µM) compared to untreated, intact cells. Peritoneal IL-6 concentrations were also increased in the PAR1/2 agonists-treated group. IL-6 expression induced by PAR activation was blocked by the treatment of cells with serine protease inhibitors. In cultured endometrial cells, simultaneous treatment with PAR1 and PAR2 agonists significantly increased the expression of IL-6. These results suggest that peritoneal bleeding may accelerate IL-6 expression in endometriotic lesions in part through the activation of PAR.


Asunto(s)
Endometriosis/metabolismo , Endometriosis/patología , Interleucina-6/biosíntesis , Interleucina-6/genética , Receptores Proteinasa-Activados/agonistas , Receptores Proteinasa-Activados/metabolismo , Animales , Apoptosis/efectos de los fármacos , Células Cultivadas , Ciclooxigenasa 2/metabolismo , Dinoprostona/farmacología , Modelos Animales de Enfermedad , Endometriosis/genética , Endometrio/metabolismo , Endometrio/patología , Células Epiteliales/metabolismo , Femenino , Humanos , Indometacina/farmacología , Interleucina-6/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , ARN Mensajero/metabolismo , Inhibidores de Serina Proteinasa/farmacología , Células del Estroma/metabolismo , Trombina/farmacología , Factor A de Crecimiento Endotelial Vascular/metabolismo
13.
Curr Pharm Des ; 18(2): 161-74, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22229576

RESUMEN

The Protease-Activated Receptors (PARs) are G-protein-coupled receptors (GPCRs) characterized by a unique mechanism of activation. They carry built in their extended N-terminal structure their own activating agonist, in the form of a cryptic tethered ligand, unmasked by an irreversible proteolytic cleavage. Besides, PARs display several other particular properties, that converge and create interacting and intertwined layers of molecular processes regulating receptor's selective signaling with important biological and pharmacological consequences. These include the operation of multiple proteases, co-factors and protease inhibitors expressed in many types of cells and tissues, creating a dynamic balance between activators and inhibitors of PAR function in a tissue specific way. Membrane microdomain compartmentalization and allosteric modulation through intermolecular interactions between PARs adds further complexity to the receptor signaling and desensitization. Furthermore, molecular components interacting with thrombin and PARs take on new roles. In particular, activated protein C (APC) forms a significant negative feedback loop for thrombin with anticoagulant properties. In addition, APC exerts anti-inflammatory and direct neuroprotective effects in vivo and in vitro. This has informed the pharmacological dissection of anticoagulant from the anti-inflammatory and neuroprotective actions of APC and the generation of engineered APC mutations with diminished risk of serious bleeding, while preserving the cytoprotective effects of APC on cells. Even more important, these advances have made possible a paradigm shift, away from a "neurocentric" and towards a "vasculo-neuronal-inflammatory model of action", which supports novel pharmacological strategies targeting multiple disease mechanisms.


Asunto(s)
Proteína C/metabolismo , Receptores Proteinasa-Activados/metabolismo , Transducción de Señal , Regulación Alostérica , Animales , Humanos , Microdominios de Membrana , Péptido Hidrolasas/metabolismo , Inhibidores de Proteasas/metabolismo , Proteína C/farmacología , Receptores Proteinasa-Activados/agonistas , Trombina/metabolismo
14.
Nat Rev Drug Discov ; 11(1): 69-86, 2012 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-22212680

RESUMEN

Proteinase-activated receptors (PARs), a family of four seven-transmembrane G protein-coupled receptors, act as targets for signalling by various proteolytic enzymes. PARs are characterized by a unique activation mechanism involving the proteolytic unmasking of a tethered ligand that stimulates the receptor. Given the emerging roles of these receptors in cancer as well as in disorders of the cardiovascular, musculoskeletal, gastrointestinal, respiratory and central nervous system, PARs have become attractive targets for the development of novel therapeutics. In this Review we summarize the mechanisms by which PARs modulate cell function and the roles they can have in physiology and diseases. Furthermore, we provide an overview of possible strategies for developing PAR antagonists.


Asunto(s)
Sistemas de Liberación de Medicamentos/tendencias , Preparaciones Farmacéuticas/administración & dosificación , Preparaciones Farmacéuticas/metabolismo , Receptores Proteinasa-Activados/metabolismo , Animales , Sistemas de Liberación de Medicamentos/métodos , Guanidinas/administración & dosificación , Guanidinas/química , Guanidinas/metabolismo , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Neoplasias/metabolismo , Oligopéptidos/administración & dosificación , Oligopéptidos/química , Oligopéptidos/metabolismo , Preparaciones Farmacéuticas/química , Receptores Proteinasa-Activados/agonistas , Receptores Proteinasa-Activados/antagonistas & inhibidores
15.
Exp Hematol ; 40(2): 131-42.e4, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22024107

RESUMEN

The pathobiological role of p53 has been widely studied, however, its role in normophysiology is relatively unexplored. We previously showed that p53 knock-down increased ploidy in megakaryocytic cultures. This study aims to examine the effect of p53 loss on in vivo megakaryopoiesis, platelet production, and function, and to investigate the basis for greater ploidy in p53(-/-) megakaryocytic cultures. Here, we used flow cytometry to analyze ploidy, DNA synthesis, and apoptosis in murine cultured and bone marrow megakaryocytes following thrombopoietin administration and to analyze fibrinogen binding to platelets in vitro. Culture of p53(-/-) marrow cells for 6 days with thrombopoietin gave rise to 1.7-fold more megakaryocytes, 26.1% ± 3.6% of which reached ploidy classes ≥64 N compared to 8.2% ± 0.9% of p53(+/+) megakaryocytes. This was due to 30% greater DNA synthesis in p53(-/-) megakaryocytes and 31% greater apoptosis in p53(+/+) megakaryocytes by day 4 of culture. Although the bone marrow and spleen steady-state megakaryocytic content and ploidy were similar in p53(+/+) and p53(-/-) mice, thrombopoietin administration resulted in increased megakaryocytic polyploidization in p53(-/-) mice. Although their platelet counts were normal, p53(-/-) mice exhibited significantly longer bleeding times and p53(-/-) platelets were less sensitive than p53(+/+) platelets to agonist-induced fibrinogen binding and P-selectin secretion. In summary, our in vivo and ex vivo studies indicate that p53 loss leads to increased polyploidization during megakaryopoiesis. Our findings also suggest for the first time a direct link between p53 loss and the development of fully functional platelets resulting in hemostatic deficiencies.


Asunto(s)
Plaquetas/fisiología , Trombopoyesis/fisiología , Proteína p53 Supresora de Tumor/fisiología , Animales , Cruzamiento , Tamaño de la Célula , Hemostasis , Masculino , Ratones , Selectina-P/metabolismo , Receptores Proteinasa-Activados/agonistas
16.
Biochem J ; 438(2): 359-67, 2011 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-21627585

RESUMEN

hPAR(2) (human proteinase-activated receptor-2) is a member of the novel family of proteolytically activated GPCRs (G-protein-coupled receptors) termed PARs (proteinase-activated receptors). Previous pharmacological studies have found that activation of hPAR(2) by mast cell tryptase can be regulated by receptor N-terminal glycosylation. In order to elucidate other post-translational modifications of hPAR(2) that can regulate function, we have explored the functional role of the intracellular cysteine residue Cys(361). We have demonstrated, using autoradiography, that Cys(361) is the primary palmitoylation site of hPAR(2). The hPAR(2)C361A mutant cell line displayed greater cell-surface expression compared with the wt (wild-type)-hPAR(2)-expressing cell line. hPAR(2)C361A also showed a decreased sensitivity and efficacy (intracellular calcium signalling) towards both trypsin and SLIGKV. In stark contrast, hPAR(2)C361A triggered greater and more prolonged ERK (extracellular-signal-regulated kinase) phosphorylation compared with that of wt-hPAR(2) possibly through Gi, since pertussis toxin inhibited the ability of this receptor to activate ERK. Finally, flow cytometry was utilized to assess the rate and extent of receptor internalization following agonist challenge. hPAR(2)C361A displayed faster internalization kinetics following trypsin activation compared with wt-hPAR(2), whereas SLIGKV had a negligible effect on internalization for either receptor. In conclusion, palmitoylation plays an important role in the regulation of PAR(2) expression, agonist sensitivity, desensitization and internalization.


Asunto(s)
Señalización del Calcio , Endocitosis , Lipoilación , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Receptores Proteinasa-Activados/metabolismo , Señalización del Calcio/efectos de los fármacos , Endocitosis/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Humanos , Espacio Intracelular/efectos de los fármacos , Espacio Intracelular/metabolismo , Lipoilación/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Modelos Moleculares , Proteínas Mutantes/metabolismo , Toxina del Pertussis/farmacología , Fosforilación/efectos de los fármacos , Receptores Proteinasa-Activados/agonistas , Tripsina/farmacología
17.
J Neurophysiol ; 105(6): 2811-7, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21430273

RESUMEN

Chronic itch is symptomatic of many skin conditions and systemic diseases. Little is known about pathophysiological alterations in itch-signaling neural pathways associated with chronic itch. We used a mouse model of hindpaw chronic dry skin itch to investigate properties of presumptive itch-signaling neurons. Neurons in the lumbar superficial dorsal horn ipsilateral to hindpaw dry skin treatment exhibited a high level of spontaneous activity that was inhibited by scratching the plantar surface. Most spontaneously active units exhibited further increases in firing rate following intradermal injection of an agonist of the protease-activated receptor PAR-2, or histamine. The large majority of pruritogen-responsive units also responded to capsaicin and allyl isothiocyanate. For neurons ipsilateral to dry skin treatment, responses elicited by the PAR-2 agonist, but not histamine or mechanical stimuli, were significantly larger compared with neurons ipsilateral to vehicle (water) treatment or neurons recorded in naïve (untreated) mice. The spontaneous activity may signal ongoing itch, while enhanced PAR-2 agonist-evoked responses may underlie hyperknesis (enhanced itch), both of which are symptomatic of many chronic itch conditions. The enhancement of neuronal responses evoked by the PAR-2 agonist, but not by histamine or mechanical stimuli, implies that the dry skin condition selectively sensitized PAR-2 agonist-sensitive primary afferent pruriceptors.


Asunto(s)
Oligopéptidos/farmacología , Células del Asta Posterior/efectos de los fármacos , Prurito/patología , Receptores Proteinasa-Activados/agonistas , Acetona/efectos adversos , Potenciales de Acción/efectos de los fármacos , Análisis de Varianza , Animales , Antipruriginosos/uso terapéutico , Capsaicina/uso terapéutico , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Lateralidad Funcional , Histamina/farmacología , Isotiocianatos/farmacología , Ratones , Modelos Biológicos , Estimulación Física/efectos adversos , Prurito/inducido químicamente , Prurito/tratamiento farmacológico , Serotonina/farmacología , Solventes/efectos adversos , Médula Espinal/patología , Agua/efectos adversos
18.
J Physiol ; 588(Pt 12): 2255-67, 2010 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-20421285

RESUMEN

Purinergic regulation of airway innate defence activities is in part achieved by the release of nucleotides from epithelial cells. However, the mechanisms of airway epithelial nucleotide release are poorly understood. We have previously demonstrated that ATP is released from ionomycin-stimulated airway epithelial goblet cells coordinately with mucin exocytosis, suggesting that ATP is released as a co-cargo molecule from mucin-containing granules. We now demonstrate that protease-activated-receptor (PAR) agonists also stimulate the simultaneous release of mucins and ATP from airway epithelial cells. PAR-mediated mucin and ATP release were dependent on intracellular Ca(2+) and actin cytoskeleton reorganization since BAPTA AM, cytochalasin D, and inhibitors of Rho and myosin light chain kinases blocked both responses. To test the hypothesis that ATP is co-released with mucin from mucin granules, we measured the nucleotide composition of isolated mucin granules purified based on their MUC5AC and VAMP-8 content by density gradients. Mucin granules contained ATP, but the levels of ADP and AMP within granules exceeded by nearly 10-fold that of ATP. Consistent with this finding, apical secretions from PAR-stimulated cells contained relatively high levels of ADP/AMP, which could not be accounted for solely based on ATP release and hydrolysis. Thus, mucin granules contribute to ATP release and also are a source of extracellular ADP and AMP. Direct release of ADP/AMP from mucin granules is likely to provide a major source of airway surface adenosine to signal in a paracrine faction ciliated cell A(2b) receptors to activate ion/water secretion and appropriately hydrate goblet cell-released mucins.


Asunto(s)
Nucleótidos de Adenina/metabolismo , Exocitosis , Células Caliciformes/metabolismo , Mucina 5AC/metabolismo , Receptores Proteinasa-Activados/metabolismo , Mucosa Respiratoria/metabolismo , Vesículas Secretoras/metabolismo , Citoesqueleto de Actina/metabolismo , Adenosina Difosfato/metabolismo , Adenosina Monofosfato/metabolismo , Adenosina Trifosfato/metabolismo , Calcio/metabolismo , Línea Celular Tumoral , Quelantes/farmacología , Exocitosis/efectos de los fármacos , Células Caliciformes/efectos de los fármacos , Humanos , Hidrólisis , Quinasa de Cadena Ligera de Miosina/antagonistas & inhibidores , Quinasa de Cadena Ligera de Miosina/metabolismo , Péptidos/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas R-SNARE/metabolismo , Receptores Proteinasa-Activados/agonistas , Mucosa Respiratoria/efectos de los fármacos , Vesículas Secretoras/efectos de los fármacos , Trombina/farmacología , Quinasas Asociadas a rho/antagonistas & inhibidores , Quinasas Asociadas a rho/metabolismo
20.
Mol Interv ; 9(2): 87-96, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19401541

RESUMEN

Protease-activated receptors (PARs) are G protein-coupled receptors (GPCRs) that transmit cellular responses begun by the actions of extracellular proteases. The activation of a PAR occurs by a unique mechanism whereby the extracellular N-terminal segment of the inactive receptor undergoes proteolytic cleavage, resulting in irreversible activation--unlike most GPCRs that are reversibly activated. PARs mediate cellular responses to coagulant proteases in various cell types localized within the vasculature. Additionally, PARs are expressed in other cell types and respond to a plethora of proteases. Recent studies have revealed that different proteases elicit distinct responses through the activation of the same PAR. This phenomenon appears to involve stabilization of distinct active PAR conformations that facilitates selectively coupling to different effectors and is localized to caveolae, a subtype of lipid rafts.


Asunto(s)
Péptido Hidrolasas/metabolismo , Receptores Proteinasa-Activados/agonistas , Receptores Proteinasa-Activados/metabolismo , Animales , Humanos , Familia de Multigenes , Unión Proteica , Conformación Proteica , Transporte de Proteínas , Receptores Proteinasa-Activados/química , Receptores Proteinasa-Activados/genética , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...