Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Int J Mol Sci ; 23(3)2022 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-35163205

RESUMEN

Acute Respiratory Distress Syndrome is the most common cause of respiratory failure among critically ill patients, and its importance has been heightened during the COVID-19 pandemic. Even with the best supportive care, the mortality rate in the most severe cases is 40-50%, and the only pharmacological agent shown to be of possible benefit has been steroids. Mesenchymal stromal cells (MSCs) have been tested in several pre-clinical models of lung injury and been found to have significant therapeutic benefit related to: (a) potent immunomodulation; (b) secretion of epithelial and endothelial growth factors; and (c) augmentation of host defense to infection. Initial translational efforts have shown signs of promise, but the results have not yielded the anticipated outcomes. One potential reason is the relatively low survival of MSCs in inflammatory conditions as shown in several studies. Therefore, strategies to boost the survival of MSCs are needed to enhance their therapeutic effect. Protease-activated receptors (PARs) may represent one such possibility as they are G-protein coupled receptors expressed by MSCs and control several facets of cell behavior. This review summarizes some of the existing literature about PARs and MSCs and presents possible future areas of investigation in order to develop potential, PAR-modified MSCs with enhanced therapeutic efficiency.


Asunto(s)
Supervivencia de Injerto/genética , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Receptores Proteinasa-Activados/fisiología , Síndrome de Dificultad Respiratoria/terapia , Animales , COVID-19/genética , COVID-19/patología , COVID-19/terapia , Supervivencia Celular/genética , Enfermedad Crítica/terapia , Humanos , Células Madre Mesenquimatosas/fisiología , Receptores Proteinasa-Activados/genética , Receptores Proteinasa-Activados/metabolismo , Síndrome de Dificultad Respiratoria/genética , Síndrome de Dificultad Respiratoria/metabolismo , Síndrome de Dificultad Respiratoria/virología , SARS-CoV-2/fisiología , Transducción de Señal/fisiología , Transfección , Resultado del Tratamiento
2.
Am J Pathol ; 192(2): 361-378, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35144762

RESUMEN

As per the classical view of the coagulation system, it functions solely in plasma to maintain hemostasis. An experimental approach modeling vascular reconstitution was used to show that vascular endothelial cells (ECs) endogenously synthesize coagulation factors during angiogenesis. Intracellular thrombin generated from this synthesis promotes the mitotic function of vascular endothelial cell growth factor A (VEGF-A). The thrombin concurrently cleaves C5a from EC-synthesized complement component C5 and unmasks the tethered ligand for EC-expressed protease-activated receptor 4 (PAR4). The two ligands jointly trigger EC C5a receptor-1 (C5ar1) and PAR4 signaling, which together promote VEGF receptor 2 growth signaling. C5ar1 is functionally associated with PAR4, enabling C5a or thrombin to elicit Gαi and/or Gαq signaling. EC coagulation factor and EC complement component synthesis concurrently down-regulate with contact inhibition. The connection of these processes with VEGF receptor 2 signaling provides new insights into mechanisms underlying angiogenesis. Knowledge of endogenous coagulation factor/complement component synthesis and joint PAR4/C5ar1 signaling could be applied to other cell types.


Asunto(s)
Factores de Coagulación Sanguínea/biosíntesis , Células Endoteliales/metabolismo , Neovascularización Fisiológica , Receptor de Anafilatoxina C5a/metabolismo , Receptores Proteinasa-Activados/metabolismo , Transducción de Señal , Animales , Factores de Coagulación Sanguínea/genética , Femenino , Masculino , Ratones , Ratones Noqueados , Receptor de Anafilatoxina C5a/genética , Receptores Proteinasa-Activados/genética
3.
Tohoku J Exp Med ; 255(1): 1-8, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34511578

RESUMEN

Endothelial nitric oxide synthase (eNOS) dysfunction is known to exacerbate the progression and prognosis of diabetic kidney disease (DKD). One of the mechanisms through which this is achieved is that low eNOS levels are associated with hypercoagulability, which promotes kidney injury. In the extrinsic coagulation cascade, the tissue factor (factor III) and downstream coagulation factors, such as active factor X (FXa), exacerbate inflammation through activation of the protease-activated receptors (PARs). Recently, it has been shown that the lack of or reduced eNOS expression in diabetic mice, as a model of advanced DKD, increases renal tissue factor levels and PAR1 and 2 expression in their kidneys. Furthermore, pharmaceutical inhibition or genetic deletion of coagulation factors or PARs ameliorated inflammation in DKD in mice lacking eNOS. In this review, we summarize the relationship between eNOS, coagulation, and PARs and propose a novel therapeutic option for the management of patients with DKD.


Asunto(s)
Nefropatías Diabéticas/etiología , Óxido Nítrico Sintasa de Tipo III/deficiencia , Receptores Proteinasa-Activados/metabolismo , Animales , Anticuerpos Neutralizantes/administración & dosificación , Coagulación Sanguínea , Nefropatías Diabéticas/sangre , Nefropatías Diabéticas/fisiopatología , Modelos Animales de Enfermedad , Inhibidores del Factor Xa/farmacología , Humanos , Riñón/efectos de los fármacos , Riñón/metabolismo , Ratones , Ratones Noqueados , Óxido Nítrico Sintasa de Tipo III/genética , Receptores Proteinasa-Activados/deficiencia , Receptores Proteinasa-Activados/genética , Transducción de Señal , Tromboplastina/antagonistas & inhibidores , Tromboplastina/metabolismo
4.
Int J Mol Sci ; 22(16)2021 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-34445691

RESUMEN

The essential role of G-protein coupled receptors (GPCRs) in tumor growth is recognized, yet a GPCR based drug in cancer is rare. Understanding the molecular path of a tumor driver gene may lead to the design and development of an effective drug. For example, in members of protease-activated receptor (PAR) family (e.g., PAR1 and PAR2), a novel PH-binding motif is allocated as critical for tumor growth. Animal models have indicated the generation of large tumors in the presence of PAR1 or PAR2 oncogenes. These tumors showed effective inhibition when the PH-binding motif was either modified or were inhibited by a specific inhibitor targeted to the PH-binding motif. In the second part of the review we discuss several aspects of some cardinal GPCRs in tumor angiogenesis.


Asunto(s)
Neoplasias/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores Proteinasa-Activados/metabolismo , Expresión Génica/genética , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Neoplasias/genética , Neovascularización Patológica/metabolismo , Neovascularización Patológica/fisiopatología , Dominios Homólogos a Pleckstrina/genética , Dominios Homólogos a Pleckstrina/fisiología , Dominios Proteicos/genética , Dominios Proteicos/fisiología , Receptor PAR-1/metabolismo , Receptor PAR-2/metabolismo , Receptores Acoplados a Proteínas G/fisiología , Receptores Proteinasa-Activados/genética , Transducción de Señal/fisiología
5.
FEBS J ; 288(8): 2697-2726, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33742547

RESUMEN

Proteinase-activated receptors (PARs) are a four-member family of G protein-coupled receptors defined by their irreversible proteolytic mechanism of activation. PARs have emerged as important regulators of various physiological responses and are implicated in numerous pathological conditions. Importantly, PAR1 and PAR4 are critical regulators of platelet function, while PAR2 is well established as a driver of inflammatory responses. PAR-targeted drug development efforts are therefore of great interest. In this review, we provide an overview of recent advances in our understanding of molecular mechanisms underlying PAR activation, effector interaction, and signaling. We also provide an overview of the diverse proteolytic enzymes that are now established as PAR regulators and describe the ability of different enzymes to elicit biased signaling through PARs. Finally, we highlight recent advances in the development of PAR-targeted pharmacological agents and discuss recent structure-activity relationship studies.


Asunto(s)
Receptor PAR-1/genética , Receptor PAR-2/genética , Receptores Acoplados a Proteínas G/genética , Receptores de Trombina/genética , Antineoplásicos/farmacología , Humanos , Péptido Hidrolasas/genética , Receptores Proteinasa-Activados/genética , Transducción de Señal/genética
6.
Physiol Rev ; 101(2): 545-567, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33124941

RESUMEN

Evolving information has identified disease mechanisms and dysregulation of host biology that might be targeted therapeutically in coronavirus disease 2019 (COVID-19). Thrombosis and coagulopathy, associated with pulmonary injury and inflammation, are emerging clinical features of COVID-19. We present a framework for mechanisms of thrombosis in COVID-19 that initially derive from interaction of SARS-CoV-2 with ACE2, resulting in dysregulation of angiotensin signaling and subsequent inflammation and tissue injury. These responses result in increased signaling by thrombin (proteinase-activated) and purinergic receptors, which promote platelet activation and exert pathological effects on other cell types (e.g., endothelial cells, epithelial cells, and fibroblasts), further enhancing inflammation and injury. Inhibitors of thrombin and purinergic receptors may, thus, have therapeutic effects by blunting platelet-mediated thromboinflammation and dysfunction in other cell types. Such inhibitors include agents (e.g., anti-platelet drugs) approved for other indications, and that could be repurposed to treat, and potentially improve the outcome of, COVID-19 patients. COVID-19, caused by the SARS-CoV-2 virus, drives dysregulation of angiotensin signaling, which, in turn, increases thrombin-mediated and purinergic-mediated activation of platelets and increase in inflammation. This thromboinflammation impacts the lungs and can also have systemic effects. Inhibitors of receptors that drive platelet activation or inhibitors of the coagulation cascade provide opportunities to treat COVID-19 thromboinflammation.


Asunto(s)
COVID-19/complicaciones , Inflamación/etiología , Receptores Proteinasa-Activados/metabolismo , Receptores Purinérgicos/metabolismo , SARS-CoV-2 , Trombosis/etiología , Humanos , Inflamación/tratamiento farmacológico , Antagonistas Purinérgicos/farmacología , Receptores Proteinasa-Activados/antagonistas & inhibidores , Receptores Proteinasa-Activados/genética , Receptores Purinérgicos/genética , Trombosis/prevención & control
7.
Mol Vis ; 26: 540-562, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32818017

RESUMEN

Purpose: Previous research in our laboratory indicated that prothrombin and other coagulation enzymes required to activate prothrombin to thrombin are synthesized by the cornea and that apoptotic human corneal stromal cells can provide a surface for prothrombin activation through the intrinsic and extrinsic coagulation pathways. The purpose of the work reported here is to study the role of thrombin activity in the regulation of matricellular protein Cyr61 (CCN1) produced by wounded phenotype human corneal stromal fibroblasts and myofibroblasts. Methods: Stromal cells from human donor corneas were converted to defined wounded phenotype fibroblasts and myofibroblasts with fetal bovine serum, followed by basic fibroblast growth factor (bFGF) and transforming growth factor beta-1 (TGFß-1), respectively, and stimulated with varying concentrations (0-10.0 units (U)/ml) of thrombin from 1-7 h. Cyr61 transcript levels were determined using reverse transcriptase-PCR (RT-PCR) and quantitative PCR (qPCR) while protein forms were analyzed using western blot data. Protease activities were characterized via protease class-specific inhibitors and western blot analysis. Thrombin activity was quantified using the fluorogenic peptide Phe-Pro-Arg-AFC. Protease-activated receptor (PAR) agonist peptides-1 and -4 were used to determine whether cells increased Cyr61 through PAR signaling pathways. The PAR-1 antagonist SCH 79797 was used to block the thrombin cleavage of the receptor. PCR data were analyzed using MxPro software and western blot data were analyzed using Image Lab™ and Image J software. Student t test and one- and two-way ANOVA (with or without ranking, depending on sample distribution), together with Dunnett's test or Tukey comparison tests for post-hoc analysis, were used to determine statistical significance.Results: Full-length Cyr61 is expressed by human corneal stromal fibroblasts and myofibroblasts and is significantly upregulated by active thrombin stimulation at the message (p<0.03) and protein (p<0.03) levels for fibroblasts and myofibroblasts. Inhibition by the allosteric thrombin-specific inhibitor hirudin prevented the thrombin-associated increase in the Cyr61 protein expression, indicating that the proteolytic activity of thrombin is required for the increase of the Cyr61 protein level. PAR-1 agonist stimulation of fibroblasts and myofibroblasts significantly increased cell-associated Cyr61 protein levels (p<0.04), and PAR-1 antagonist SCH 79797 significantly inhibited the thrombin stimulated increase of Cyr61 in fibroblasts but not in myofibroblasts. In the fibroblast and myofibroblast conditioned media, Cyr61 was detected as the full-length 40 kDa protein in the absence of thrombin, and mainly at 24 kDa in the presence of thrombin at ≥0.5 U/ml, using an antibody directed toward the internal linker region between the von Willebrand factor type C and thrombospondin type-1 domains. Although known to undergo alternative splicing, Cyr61 that is synthesized by corneal fibroblasts and myofibroblasts is not alternatively spliced in response to thrombin stimulation nor is Cyr61 directly cleaved by thrombin to generate its 24 kDa form; instead, Cyr61 is proteolytically processed into 24 kDa N- and 16 kDa C-terminal fragments by a thrombin activated leupeptin-sensitive protease present in conditioned media with activity distinct from the proteolytic activity of thrombin. Conclusions: In cultured human corneal stromal fibroblasts and myofibroblasts, thrombin regulates Cyr61 through two mechanisms: 1) thrombin increases the Cyr61 expression at the message and protein levels, and 2) thrombin increases the activation of a leupeptin-sensitive protease that stimulates the cleavage of Cyr61 into N- and C-terminal domain populations in or near the thrombospondin type-1 domain. Generation of Cyr61 peptides during corneal injury stimulation may reveal additional functions of the protein, which modulate corneal wound healing activities or decrease activities of the full-length Cyr61 form.


Asunto(s)
Proteína 61 Rica en Cisteína/genética , Fibroblastos/efectos de los fármacos , Miofibroblastos/efectos de los fármacos , Receptores Proteinasa-Activados/genética , Células del Estroma/efectos de los fármacos , Trombina/farmacología , Empalme Alternativo , Diferenciación Celular , Sustancia Propia/citología , Sustancia Propia/metabolismo , Medios de Cultivo Condicionados/farmacología , Proteína 61 Rica en Cisteína/antagonistas & inhibidores , Proteína 61 Rica en Cisteína/metabolismo , Factor 2 de Crecimiento de Fibroblastos/farmacología , Fibroblastos/citología , Fibroblastos/metabolismo , Hirudinas/farmacología , Humanos , Leupeptinas/farmacología , Miofibroblastos/citología , Miofibroblastos/metabolismo , Cultivo Primario de Células , Proteolisis , Pirroles/farmacología , Quinazolinas/farmacología , ARN Mensajero/antagonistas & inhibidores , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores Proteinasa-Activados/metabolismo , Transducción de Señal , Células del Estroma/citología , Células del Estroma/metabolismo , Trombina/metabolismo , Factor de Crecimiento Transformador beta1/farmacología
8.
FEBS J ; 287(4): 645-658, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31495063

RESUMEN

Protease-activated receptors (PARs) are a unique class of G-protein-coupled transmembrane receptors, which revolutionized the perception of proteases from degradative enzymes to context-specific signaling factors. Although PARs are traditionally known to affect several vascular responses, recent investigations have started to pinpoint the functional role of PAR signaling in the gastrointestinal (GI) tract. This organ is exposed to the highest number of proteases, either from the gut lumen or from the mucosa. Luminal proteases include the host's digestive enzymes and the proteases released by the commensal microbiota, while mucosal proteases entail extravascular clotting factors and the enzymes released from resident and infiltrating immune cells. Active proteases and, in case of a disrupted gut barrier, even entire microorganisms are capable to translocate the intestinal epithelium, particularly under inflammatory conditions. Especially PAR-1 and PAR-2, expressed throughout the GI tract, impact gut permeability regulation, a major factor affecting intestinal physiology and metabolic inflammation. In addition, PARs are critically involved in the onset of inflammatory bowel diseases, irritable bowel syndrome, and tumor progression. Due to the number of proteases involved and the multiple cell types affected, selective regulation of intestinal PARs represents an interesting therapeutic strategy. The analysis of tissue/cell-specific knockout animal models will be of crucial importance to unravel the intrinsic complexity of this signaling network. Here, we provide an overview on the implication of PARs in intestinal permeability regulation under physiologic and disease conditions.


Asunto(s)
Neoplasias Gastrointestinales/genética , Tracto Gastrointestinal/metabolismo , Enfermedades Inflamatorias del Intestino/genética , Péptido Hidrolasas/genética , Receptores Proteinasa-Activados/genética , Receptores Proteinasa-Activados/metabolismo , Transducción de Señal/genética , Animales , Traslocación Bacteriana , Modelos Animales de Enfermedad , Microbioma Gastrointestinal/fisiología , Neoplasias Gastrointestinales/metabolismo , Neoplasias Gastrointestinales/microbiología , Neoplasias Gastrointestinales/patología , Tracto Gastrointestinal/citología , Tracto Gastrointestinal/microbiología , Regulación de la Expresión Génica , Humanos , Enfermedades Inflamatorias del Intestino/metabolismo , Enfermedades Inflamatorias del Intestino/microbiología , Enfermedades Inflamatorias del Intestino/patología , Mucosa Intestinal/citología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Péptido Hidrolasas/metabolismo , Permeabilidad , Simbiosis/fisiología
9.
Curr Drug Targets ; 21(7): 647-656, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31755385

RESUMEN

The major proteases that constitute the fibrinolysis system are tightly regulated. Protease inhibitors target plasmin, the protease responsible for fibrin degradation, and the proteases that convert plasminogen into plasmin, including tissue-type plasminogen activator (tPA) and urokinase-type plasminogen activator (uPA). A second mechanism by which fibrinolysis is regulated involves exosite interactions, which localize plasminogen and its activators to fibrin, extracellular matrix (ECM) proteins, and cell surfaces. Once plasmin is generated in association with cell surfaces, it may cleave transmembrane proteins, activate growth factors, release growth factors from ECM proteins, remodel ECM, activate metalloproteases, and trigger cell-signaling by cleaving receptors in the Proteaseactivated Receptor (PAR) family. These processes are all implicated in cancer. It is thus not surprising that a family of structurally diverse but functionally similar cell-surface proteins, called Plasminogen Receptors (PlgRs), which increase the catalytic efficiency of plasminogen activation, have received attention for their possible function in cancer and as targets for anticancer drug development. In this review, we consider four previously described PlgRs, including: α-enolase, annexin-A2, Plg-RKT, and cytokeratin-8, in human cancer. To compare the PlgRs, we mined transcriptome profiling data from The Cancer Genome Atlas (TCGA) and searched for correlations between PlgR expression and patient survival. In glioma, the expression of specific PlgRs correlates with tumor grade. In a number of malignancies, including glioblastoma and liver cancer, increased expression of α-enolase or annexin-A2 is associated with an unfavorable prognosis. Whether these correlations reflect the function of PlgRs as receptors for plasminogen or other activities is discussed.


Asunto(s)
Neoplasias/genética , Neoplasias/metabolismo , Receptores del Activador de Plasminógeno Tipo Uroquinasa/biosíntesis , Receptores del Activador de Plasminógeno Tipo Uroquinasa/genética , Animales , Fibrinólisis/fisiología , Humanos , Terapia Molecular Dirigida , Clasificación del Tumor , Neoplasias/terapia , Pronóstico , Receptores Proteinasa-Activados/biosíntesis , Receptores Proteinasa-Activados/genética
10.
Int J Mol Sci ; 19(11)2018 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-30400241

RESUMEN

G protein-coupled receptors (GPCRs), the largest family of cell receptors, act as important regulators of diverse signaling pathways. Our understanding of the impact of GPCRs in tumors is emerging, yet there is no therapeutic platform based on GPCR driver genes. As cancer progresses, it disrupts normal epithelial organization and maintains the cells outside their normal niche. The dynamic and flexible microenvironment of a tumor contains both soluble and matrix-immobilized proteases that contribute to the process of cancer advancement. An example is the activation of cell surface protease-activated receptors (PARs). Mammalian PARs are a subgroup of GPCRs that form a family of four members, PAR1⁻4, which are uniquely activated by proteases found in the microenvironment. PAR1 and PAR2 play central roles in tumor biology, and PAR3 acts as a coreceptor. The significance of PAR4 in neoplasia is just beginning to emerge. PAR1 has been shown to be overexpressed in malignant epithelia, in direct correlation with tumor aggressiveness, but there is no expression in normal epithelium. In this review, the involvement of key transcription factors such as Egr1, p53, Twist, AP2, and Sp1 that control PAR1 expression levels specifically, as well as hormone transcriptional regulation by both estrogen receptors (ER) and androgen receptors (AR) are discussed. The cloning of the human protease-activated receptor 2; Par2 (hPar2) promoter region and transcriptional regulation of estrogen (E2) via binding of the E2⁻ER complex to estrogen response elements (ERE) are shown. In addition, evidence that TEA domain 4 (TEAD4) motifs are present within the hPar2 promoter is presented since the YAP oncogene, which plays a central part in tumor etiology, acts via the TEAD4 transcription factor. As of now, no information is available on regulation of the hPar3 promoter. With regard to hPar4, only data showing CpG methylation promoter regulation is available. Characterization of the PAR transcriptional landscape may identify powerful targets for cancer therapies.


Asunto(s)
Células Epiteliales/patología , Neoplasias/genética , Neoplasias/patología , Animales , Secuencia de Bases , Humanos , Regiones Promotoras Genéticas , Receptores Proteinasa-Activados/genética , Receptores Proteinasa-Activados/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética
11.
Int J Mol Sci ; 19(7)2018 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-29954076

RESUMEN

G protein-coupled receptors (GPCRs) are a large diverse family of cell surface signaling receptors implicated in various types of cancers. Several studies indicate that GPCRs control many aspects of cancer progression including tumor growth, invasion, migration, survival and metastasis. While it is known that GPCR activity can be altered in cancer through aberrant overexpression, gain-of-function activating mutations, and increased production and secretion of agonists, the precise mechanisms of how GPCRs contribute to cancer progression remains elusive. Protease-activated receptors (PARs) are a unique class of GPCRs implicated in cancer. PARs are a subfamily of GPCRs comprised of four members that are irreversibly activated by proteolytic cleavage induced by various proteases generated in the tumor microenvironment. Given the unusual proteolytic irreversible activation of PARs, expression of receptors at the cell surface is a key feature that influences signaling responses and is exquisitely controlled by endocytic adaptor proteins. Here, we discuss new survey data from the Cancer Genome Atlas and the Genotype-Tissue Expression projects analysis of expression of all PAR family member expression in human tumor samples as well as the role and function of the endocytic sorting machinery that controls PAR expression and signaling of PARs in normal cells and in cancer.


Asunto(s)
Neoplasias/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores Proteinasa-Activados/metabolismo , Animales , Arrestinas/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Femenino , Humanos , Neoplasias/genética , Receptores Acoplados a Proteínas G/genética , Receptores Proteinasa-Activados/genética , Transducción de Señal/genética , Transducción de Señal/fisiología
12.
Int J Cardiol ; 252: 163-166, 2018 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-29249425

RESUMEN

BACKGROUND: Thrombin promotes cardiac fibroblast proliferation and fibrosis via protease-activated receptor (PAR-1). PAR-4 is reportedly absent in cardiac fibroblasts. In smooth muscle cells, PAR-4 expression is also low but increases upon hyperglycemia and contributes to vascular remodelling in diabetic mice. We examined if PAR-4 is a glucose-responsive gene with remodelling-related functions in cardiac fibroblasts. METHODS AND RESULTS: Cardiac PAR-4 increased in mice with streptozotocin- or diabetogenic diet (DD)-induced diabetes. PAR-4 mRNA and protein were detectable in cardiac fibroblasts from chow-fed mice and increased in high (HG, 25mM) vs. low glucose (LG; 5.5mM) cultures. Conversely PAR-4 mRNA was higher in fibroblasts from DD-fed mice but reduced in LG cultures. Cardiac fibroblasts in HG culture responded more strongly to thrombin or PAR-4 activating peptide in terms of migration (wound-scratch assay), remodelling-associated gene expression (interleukin 6, alpha smooth muscle actin) and oxidative stress (dihydroethidium fluorescence). CONCLUSION: PAR-4 is expressed in mouse cardiac fibroblasts and is dynamically regulated by extracellular glucose in vitro and diabetes in vivo, thereby impacting on fibroblast functions relevant for cardiac remodelling. These findings add further evidence for the usefulness of the recently developed PAR-4 antagonists in clinical settings.


Asunto(s)
Fibroblastos/metabolismo , Glucosa/metabolismo , Miocitos Cardíacos/metabolismo , Receptores Proteinasa-Activados/biosíntesis , Trombina/biosíntesis , Animales , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Fibroblastos/efectos de los fármacos , Expresión Génica , Glucosa/toxicidad , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Miocitos Cardíacos/efectos de los fármacos , Receptores Proteinasa-Activados/genética , Trombina/genética
13.
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
14.
J Pharmacol Sci ; 133(3): 156-161, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28314697

RESUMEN

Activated factor X (FXa) plays an important role in thrombin generation and inflammation. Factor X is not converted constitutively to FXa, but only after intrinsic clotting factors are activated and/or cellular injury occurs. Although rivaroxaban is one of direct FXa inhibitors, its function in the inactivated coagulation cascade is unclear. In human umbilical vein endothelial cells that natively express protease-activated receptor-1 and -2, high dose rivaroxaban did not alter gene transcripts including pro-inflammatory genes in DNA microarray. Upon FXa stimulation, the expressions of pro-inflammatory genes such as monocyte chemoattractant protein-1 (MCP-1), intracellular adhesion molecule-1, and interleukin-8 were maximally increased at 4 h after stimulation, and were suppressed by rivaroxaban. To confirm these results, quantitative polymerase chain reaction and enzyme-linked immunosorbent assay (ELISA) for MCP-1 were performed. FXa evoked the expression of MCP-1 maximally at 4 h after stimulation, whereas MCP-1 displayed a different temporal activation in ELISA. Interestingly, rivaroxaban inhibited both time courses of MCP-1 expression. These results suggest that rivaroxaban may not influence gene modulation in the inactivated coagulation state, but can attenuate the endothelial damage evoked by FXa and pro-inflammatory cytokine genes.


Asunto(s)
Inhibidores del Factor Xa/farmacología , Factor Xa/farmacología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Rivaroxabán/farmacología , Células Cultivadas , Quimiocina CCL2/genética , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Molécula 1 de Adhesión Intercelular/genética , Interleucina-8/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Receptores Proteinasa-Activados/genética
15.
Gynecol Oncol ; 145(1): 167-175, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28148395

RESUMEN

OBJECTIVE: Enhanced tissue factor (TF) expression in epithelial ovarian cancer (EOC) is associated with aggressive disease. Our objective was to evaluate the role of the TF-factor VIIa-protease-activated receptor-2 (PAR-2) pathway in human EOC. METHODS: TCGA RNAseq data from EOC databases were analyzed for PAR expression. Cell and microparticle (MP) associated TF protein expression (Western blot) and MP-associated coagulant activity were determined in human EOC (SKOV-3, OVCAR-3 and CaOV-3) and control cell lines. PAR-1 and PAR-2 protein expressions were similarly examined. The PAR dependence of VEGF-A release (ELISA) and chemotactic migration in response to FVIIa and cellular proliferation in response to thrombin was evaluated with small molecule antagonists. RESULTS: Relative mRNA expression consistently demonstrated PAR-2>PAR-1≫PAR-3/4 in multiple EOC datasets. Human EOC cell line lysates confirmed expression of TF, PAR-1 and PAR-2 proteins. MPs isolated from EOC cell lines demonstrated markedly enhanced (4-10 fold) TF coagulant activity relative to control cell lines. FVIIa induced a dose-dependent increase in VEGF-A release (2.5-3 fold) from EOC cell lines that was abrogated by the PAR-2 antagonist ENMD-1068. FVIIa treatment of CaOV-3 and OVCAR-3 cells resulted in increased chemotactic migration that was abolished by ENMD-1068. Thrombin induced dose-dependent EOC cell line proliferation was completely reversed by the PAR-1 antagonist vorapaxar. Small molecule antagonists had no effect on these phenotypes without protease present. CONCLUSIONS: Enhanced activity of the TF-FVIIa-PAR-2 axis may contribute to the EOC progression via PAR-2 dependent signaling that supports an angiogenic and invasive phenotype and local thrombin generation supporting PAR-1 dependent proliferation.


Asunto(s)
Movimiento Celular , Factor VIIa/metabolismo , Neoplasias Glandulares y Epiteliales/genética , Neoplasias Ováricas/genética , ARN Mensajero/metabolismo , Receptor PAR-1/genética , Receptor PAR-2/genética , Tromboplastina/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Western Blotting , Carcinoma Epitelial de Ovario , Línea Celular Tumoral , Proliferación Celular , Quimiotaxis , Femenino , Humanos , Invasividad Neoplásica , Neoplasias Glandulares y Epiteliales/metabolismo , Neoplasias Glandulares y Epiteliales/patología , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Receptor PAR-1/metabolismo , Receptor PAR-2/metabolismo , Receptores Proteinasa-Activados/genética , Receptores Proteinasa-Activados/metabolismo , Transducción de Señal , Trombina/metabolismo
16.
Am J Rhinol Allergy ; 31(1): 13-18, 2017 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-28234145

RESUMEN

BACKGROUND: Nasal epithelial cells and infiltrating eosinophils express tissue factor, and high thrombin activity and excess fibrin deposition are found in nasal secretion and in nasal polyp from patients with chronic rhinosinusitis with nasal polyp (CRSwNP). Activated coagulation factors play important roles not only in thrombosis but also in inflammation through interaction with protease-activated receptors (PAR). However, little is known about the effects of activated coagulation factors on the release of cytokines and extracellular matrix from nasal polyp fibroblasts (NPF). PURPOSE: The purpose of this study was to analyze the expression of PARs, which are receptors for activated coagulation factors, on NPFs and to determine the roles of thrombin and activated coagulation factor X (FXa) in the release of cytokines and fibronectin from NPFs. METHODS: NPFs were obtained from patients with CRSwNP, and the messenger RNA (mRNA) and protein expression of PARs in these NPFs were examined. We then investigated whether thrombin or FXa stimulates the release of transforming growth factor (TGF) beta 1, fibronectin, eotaxin-1, interleukin (IL) 6, or IL-8 from cultured NPFs. The effects of PAR agonists on the release of cytokines and fibronectin were also examined. RESULTS: NPFs expressed the mRNA and proteins of all four PARs: PAR-1, PAR-2, PAR-3, and PAR-4. Both thrombin and FXa significantly stimulated the release of TGF beta 1, fibronectin, eotaxin-1, IL-6, and IL-8 from cultured NPFs. PAR-1 and PAR-2 agonists stimulated the secretion of TGF beta 1, fibronectin, eotaxin-1, IL-6, and IL-8. PAR-3 agonist stimulated the release of TGF beta 1, fibronectin, and eotaxin-1. PAR-4 agonist did not induce the release of these molecules. CONCLUSION: NPFs play important roles in the pathophysiology of CRSwNP such as in nasal polyp formation and inflammatory cell infiltration by releasing cytokines and extracellular matrix proteins. Activated coagulation factors, thrombin and FXa, stimulate the release of these cytokines and fibronectin from NPFs via PARs.


Asunto(s)
Factor Xa/metabolismo , Fibroblastos/metabolismo , Pólipos Nasales/inmunología , Rinitis/inmunología , Sinusitis/inmunología , Trombina/metabolismo , Células Cultivadas , Enfermedad Crónica , Citocinas/metabolismo , Fibroblastos/inmunología , Fibroblastos/patología , Fibronectinas/metabolismo , Regulación de la Expresión Génica , Humanos , Pólipos Nasales/patología , Receptores Proteinasa-Activados/genética , Receptores Proteinasa-Activados/metabolismo , Rinitis/patología , Sinusitis/patología
17.
PLoS One ; 11(10): e0165565, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27788223

RESUMEN

Anti-platelet drugs are the mainstay of pharmacotherapy for heart attack and stroke prevention, yet improvements are continually sought. Thrombin is the most potent activator of platelets and targeting platelet thrombin receptors (protease-activated receptors; PARs) is an emerging anti-thrombotic approach. Humans express two PARs on their platelets-PAR1 and PAR4. The first PAR1 antagonist was recently approved for clinical use and PAR4 antagonists are in early clinical development. However, pre-clinical studies examining platelet PAR function are challenging because the platelets of non-primates do not accurately reflect the PAR expression profile of human platelets. Mice, for example, express Par3 and Par4. To address this limitation, we aimed to develop a genetically modified mouse that would express the same repertoire of platelet PARs as humans. Here, human PAR1 preceded by a lox-stop-lox was knocked into the mouse Par3 locus, and then expressed in a platelet-specific manner (hPAR1-KI mice). Despite correct targeting and the predicted loss of Par3 expression and function in platelets from hPAR1-KI mice, no PAR1 expression or function was detected. Specifically, PAR1 was not detected on the platelet surface nor internally by flow cytometry nor in whole cell lysates by Western blot, while a PAR1-activating peptide failed to induce platelet activation assessed by either aggregation or surface P-selectin expression. Platelets from hPAR1-KI mice did display significantly diminished responsiveness to thrombin stimulation in both assays, consistent with a Par3-/- phenotype. In contrast to the observations in hPAR1-KI mouse platelets, the PAR1 construct used here was successfully expressed in HEK293T cells. Together, these data suggest ectopic PAR1 expression is not tolerated in mouse platelets and indicate a different approach is required to develop a small animal model for the purpose of any future preclinical testing of PAR antagonists as anti-platelet drugs.


Asunto(s)
Plaquetas/metabolismo , Moléculas de Adhesión Celular/metabolismo , Perfilación de la Expresión Génica , Receptores Proteinasa-Activados/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Proteínas de Ciclo Celular , Ratones , Receptores Proteinasa-Activados/genética
18.
Int J Mol Med ; 37(3): 593-602, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26820116

RESUMEN

Belonging to the G protein-coupled receptor (GPcr) family, the protease-activated receptors (Pars) consist of 4 members, PAR1-4. PARs mediate the activation of cells via thrombin, serine and other proteases. Such protease-triggered signaling events are thought to be critical for hemostasis, thrombosis and other normal pathological processes. In the present study, we examined the evolution of PARs by analyzing phylogenetic trees, chromosome location, selective pressure and functional divergence based on the 169 functional gene alignment sequences from 57 vertebrate gene sequences. We found that the 4 Pars originated from 4 invertebrate ancestors by phylogenetic trees analysis. The selective pressure results revealed that only PAR1 appeared by positive selection during its evolution, while the other PAR members did not. In addition, we noticed that although these PARs evolved separately, the results of functional divergence indicated that their evolutional rates were similar and their functions did not significantly diverge. The findings of our study provide valuable insight into the evolutionary history of the vertebrate PAR family.


Asunto(s)
Receptores Proteinasa-Activados/genética , Vertebrados/metabolismo , Animales , Humanos , Filogenia , Receptores Proteinasa-Activados/clasificación
19.
Auris Nasus Larynx ; 43(2): 166-70, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26254958

RESUMEN

OBJECTIVE: Fibroblasts are major supporting cells in nasal mucosa and can induce inflammatory process with recruitment of inflammatory cells. Airborne fungi have been suggested as an etiologic factor of chronic rhinosinusitis (CRS). The aim of this study was to investigate the interaction between airborne fungi and pattern recognition receptors (PRRs) in nasal fibroblasts. METHODS: Primary nasal polyp fibroblasts were cultured with Alternaria and Aspergillus for 48h. To determine the production of chemical mediators interleukine-6 (IL-6), IL-8, granulocyte-macrophage colony stimulating factor (GM-CSF), eotaxin, and regulated on activation normal T expressed and secreted (RANTES) were measured with enzyme immunoassay methods. PRRs for toll-like receptors (TLRs) and protease-activated receptors (PARs) mRNA were determined with reverse transcription polymerase chain reaction (RT-PCR). To determine the role of PRRs, fibroblasts were treated with small interfering RNA (siRNA). RESULTS: IL-6 and IL-8 productions were significantly increased by 50 and 100µg/ml of Alternaria. However, GM-CSF, eotaxin, and RANTES productions did not change. Aspergillus did not influence the production of chemical mediators from nasal polyp fibroblasts. TLR2 and TLR5 mRNA expressions were significantly increased by fungi and these two TLRs were associated with the production of IL-6 and IL-8. CONCLUSION: Alternaria interacts as a pathogen-associated molecular pattern with the PRRs, such as TLR2 and TLR5, which induce the production of inflammatory chemical mediators from nasal polyp fibroblasts. Airborne fungi enhance the innate immune defense mechanism and may be associated with the pathogenesis of nasal inflammatory diseases.


Asunto(s)
Fibroblastos/metabolismo , Micosis/genética , Pólipos Nasales/genética , ARN Mensajero/metabolismo , Receptores de Reconocimiento de Patrones/genética , Receptores Proteinasa-Activados/genética , Receptores Toll-Like/metabolismo , Adulto , Alternaria , Aspergillus , Células Cultivadas , Quimiocina CCL5/metabolismo , Quimiocinas/metabolismo , Enfermedad Crónica , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Humanos , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Masculino , Persona de Mediana Edad , Micosis/metabolismo , Micosis/microbiología , Pólipos Nasales/metabolismo , Pólipos Nasales/microbiología , ARN Interferente Pequeño , Receptores de Reconocimiento de Patrones/metabolismo , Receptores Proteinasa-Activados/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Rinitis/genética , Rinitis/metabolismo , Rinitis/microbiología , Sinusitis/genética , Sinusitis/metabolismo , Sinusitis/microbiología
20.
Haemophilia ; 22(1): 152-9, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26189554

RESUMEN

INTRODUCTION AND AIM: Joint bleeding results in blood-induced arthropathy. We investigate whether a joint bleed alters protease-activated receptor (PAR) expression, and whether treatment with small interfering RNA (siRNA) targeted against PAR1-4 attenuates synovitis and cartilage damage. METHODS: Protease-activated receptor expression was evaluated upon a joint bleed in haemophilic mice and in humans. In addition, mice with a joint bleed were randomized between treatment with PAR1-4 siRNA or control and evaluated for the presence of synovitis and cartilage damage. Also, human cartilage was transfected with PAR1-4 siRNA or control, and evaluated for plasmin-induced cartilage damage. RESULTS: Following a joint bleed, we observed an increase in synovial PAR1, -2 and -4 expression, and an increase in chondrocyte PAR2 and -3 expression in mice (all P < 0.05). Also an increase in synovial PAR1 and chondrocyte PAR4 expression in patients was observed (both P < 0.05). Treatment of a joint bleed in haemophilic mice with PAR1-4 siRNA attenuates synovitis and cartilage damage (both P < 0.01). Treatment of human cartilage tissue explants with PAR1-4 siRNA reduced plasmin-induced cartilage damage (P < 0.01). CONCLUSION: This study demonstrates that synovial and chondrocyte PAR expression is altered upon a joint bleed, and that treatment with PAR1-4 siRNA attenuates synovitis and plasmin-induced cartilage damage.


Asunto(s)
Cartílago Articular/patología , Silenciador del Gen , Hemofilia A/complicaciones , Hemorragia/complicaciones , Receptores Proteinasa-Activados/deficiencia , Receptores Proteinasa-Activados/genética , Sinovitis/genética , Animales , Cartílago Articular/efectos de los fármacos , Cartílago Articular/metabolismo , Condrocitos/efectos de los fármacos , Condrocitos/metabolismo , Fibrinolisina/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Hemorragia/genética , Hemorragia/patología , Humanos , Ratones , ARN Interferente Pequeño/genética , Sinovitis/complicaciones , Sinovitis/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...