Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Adipocyte ; 9(1): 170-178, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32272863

RESUMEN

Adipose tissue is an important metabolic organ, and transplantation of white adipose tissue plays crucial roles in glucose homoeostasis and energy metabolism. However, how adipose tissue affects glucose utilization is poorly understood. PAI-1-knockout (PAI-1KO) mice were previously shown to be resistant to a high-fat diet and obesity. We used microPET/CT (positron emission tomography/computed tomography), gene microarray, and biochemical assays to measure changes in systemic and myocardial glucose metabolism in mice subjected to transplantation of adipose tissue from PAI-1KO and wild-type mice. Here, we show that transplanting subcutaneous white adipose tissue (scWAT) from PAI-1KO mice into high-fat diet (HFD)-fed mice reduced levels of serum total cholesterol and triglycerides, and improved glucose tolerance in the HFD-fed mice. microPET/CT imaging revealed that cardiac glucose uptake was increased in the heart but not in the liver, hindlimb muscles, or abdominal subcutaneous white adipose tissue in HFD-fed mice transplanted with PAI-1KO scWAT, suggesting that the transplanted PAI-1KO scWAT exerted endocrine effects in the heart. In addition, transplantation of scWAT from PAI-1KO mice upregulated mitochondrial gene expression in cardiac muscle, increased the expression of glucose transporters 1 and 4 in cardiac tissues and was associated with an increased NAD+/NADH ratio. Together, these findings suggest that modulating PAI-1 in scWAT may provide a promising approach for intervening in glucose metabolism.


Asunto(s)
Tejido Adiposo/metabolismo , Glucosa/metabolismo , Miocardio/metabolismo , Obesidad/metabolismo , Serpina E2/metabolismo , Animales , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Prueba de Tolerancia a la Glucosa , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocardio/patología , Obesidad/inducido químicamente , Serpina E2/deficiencia
2.
Am J Respir Cell Mol Biol ; 62(3): 319-330, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31513752

RESUMEN

Senescence of alveolar type II (ATII) cells, progenitors of the alveolar epithelium, is a pathological feature and contributes importantly to the pathogenesis of idiopathic pulmonary fibrosis. Despite recognition of the importance of ATII cell senescence in idiopathic pulmonary fibrosis pathogenesis, how ATII cell senescence is regulated and how senescent ATII cells contribute to lung fibrogenesis remain unclear. In this study, we show that TGF-ß1 (transforming growth factor-ß1), a most ubiquitous and potent profibrotic cytokine, induces plasminogen activator inhibitor-1 (PAI-1), a cell senescence and fibrosis mediator, and p16 as well as senescence, but not apoptosis, in primary mouse ATII cells. We also found that senescent ATII cells secrete various cytokines and chemokines, including IL-4 and IL-13, which stimulate the expression of genes associated with a profibrotic phenotype in alveolar macrophages. Similar responses were also observed in TGF-ß1-treated rat ATII (L2) and rat macrophage NR8383 cells. Deletion of PAI-1 or inhibition of PAI-1 activity with a small molecule PAI-1 inhibitor, however, blocks TGF-ß1-induced senescence as well as a senescence-associated secretory phenotype in ATII and L2 cells and, consequently, the stimulatory effects of the conditioned medium from senescent ATII/L2 cells on macrophages. Moreover, we show that silencing p16 ameliorates PAI-1 protein-induced ATII cell senescence and secretion of profibrotic mediators. Our data suggest that PAI-1 mediates TGF-ß1-induced ATII cell senescence and secretion of profibrotic mediators through inducing p16, and they also suggest that senescent ATII cells contribute to lung fibrogenesis in part by activating alveolar macrophages through secreting profibrotic and proinflammatory mediators.


Asunto(s)
Células Epiteliales Alveolares/citología , Senescencia Celular/fisiología , Activación de Macrófagos/fisiología , Macrófagos Alveolares/fisiología , Serpina E2/fisiología , Factor de Crecimiento Transformador beta1/fisiología , Células Epiteliales Alveolares/metabolismo , Animales , Células Cultivadas , Quimiocinas/metabolismo , Medios de Cultivo Condicionados/farmacología , Inhibidor p16 de la Quinasa Dependiente de Ciclina/fisiología , Citocinas/metabolismo , Genes p16 , Ratones , Ratones Noqueados , Fibrosis Pulmonar/patología , Interferencia de ARN , ARN Interferente Pequeño/genética , Ratas , Serpina E2/deficiencia , Serpina E2/genética
3.
Transl Stroke Res ; 10(4): 372-380, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-29978354

RESUMEN

After stroke, secondary brain damage is influenced by the extent of fibrin clot formation. This is counteracted by the endogenous fibrinolysis. Of major interest are the key players of the fibrinolytic plasminogen activator system including the urokinase plasminogen activator (uPA), the tissue-type plasminogen activator (tPA), and their endogenous inhibitors plasminogen activator inhibitor 1 (PAI-1) and PAI-2. The role of PAI-1 in brain injury is well established, whereas the importance of PAI-2 is unknown at present. The objectives of the present were twofold: first, to characterize the time-dependent cerebral mRNA expression of the plasminogen activator system (PAS) after brain ischemia and second, to investigate the impact of PAI-1 and PAI-2 on brain infarct volume using gene-deficient mice. Adult C57Bl/6J mice were subjected to unilateral transient middle cerebral artery occlusion (MCAO) followed by reperfusion for 3, 24, 72, or 120 h. Quantitative PCR revealed that brain mRNA expression levels of the PAS components, and particularly of PAI-1 (237-fold) and PAI-2 (19-fold), peaked at 24 h after stroke. Accordingly, PAI-1 plasma activity was strongly increased. Brain infarct volume in TTC (2,3,5-triphenyltetrazolium chloride)-stained brain sections was significantly smaller 24 h after MCAO in PAI-1-deficient mice (- 31%), but not in PAI-2-deficient mice (- 6%). Thus, endogenous upregulation of PAI-1, but not of PAI-2, might contribute to increased brain damage after acute ischemic stroke. The present study therefore shows that PAI-2 is induced by brain ischemia, but does not play an important or relevant role for secondary brain damage after brain injury.


Asunto(s)
Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/prevención & control , Inhibidor 2 de Activador Plasminogénico/deficiencia , Serpina E2/deficiencia , Accidente Cerebrovascular/metabolismo , Animales , Lesiones Encefálicas/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Inhibidor 2 de Activador Plasminogénico/genética , Serpina E2/genética , Accidente Cerebrovascular/genética
4.
Sci Rep ; 8(1): 5810, 2018 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-29643421

RESUMEN

Injuries to flexor tendons can be complicated by fibrotic adhesions, which severely impair the function of the hand. Adhesions have been associated with TGF-ß1, which causes upregulation of PAI-1, a master suppressor of protease activity, including matrix metalloproteinases (MMP). In the present study, the effects of inhibiting PAI-1 in murine zone II flexor tendon injury were evaluated utilizing knockout (KO) mice and local nanoparticle-mediated siRNA delivery. In the PAI-1 KO murine model, reduced adherence of injured tendon to surrounding subcutaneous tissue and accelerated recovery of normal biomechanical properties compared to wild type controls were observed. Furthermore, MMP activity was significantly increased in the injured tendons of the PAI-1 KO mice, which could explain their reduced adhesions and accelerated remodeling. These data demonstrate that PAI-1 mediates fibrotic adhesions in injured flexor tendons by suppressing MMP activity. In vitro siRNA delivery to silence Serpine1 expression after treatment with TGF-ß1 increased MMP activity. Nanoparticle-mediated delivery of siRNA targeting Serpine1 in injured flexor tendons significantly reduced target gene expression and subsequently increased MMP activity. Collectively, the data demonstrate that PAI-1 can be a druggable target for treating adhesions and accelerating the remodeling of flexor tendon injuries.


Asunto(s)
Metaloproteinasas de la Matriz/metabolismo , Inhibidor 1 de Activador Plasminogénico/metabolismo , Inhibidores de Serina Proteinasa/metabolismo , Traumatismos de los Tendones/patología , Animales , Modelos Animales de Enfermedad , Fibrosis/patología , Técnicas de Silenciamiento del Gen , Ratones , Ratones Noqueados , Serpina E2/deficiencia
5.
FASEB J ; 32(1): 276-288, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28899878

RESUMEN

Endothelial cells (ECs) in the tumor microenvironment have been reported to play a more active role in solid tumor growth and metastatic dissemination than simply providing the physical structure to form conduits for blood flow; however, the involvement of ECs in the process of triple-negative breast cancer (TNBC) metastasis has not been addressed. Here, we demonstrate that ECs-when mixed with TNBC cells-could increase TNBC cell metastatic potency. After treatment with TGF-ß to induce endothelial-mesenchymal transition (EMT), TNBC cells could produce plasminogen activator inhibitor-1 (PAI-1) and stimulate the expression and secretion of the chemokine, CCL5, from ECs, which then acts in a paracrine fashion on TNBC cells to enhance their migration, invasion, and metastasis. CCL5, in turn, accelerates TNBC cell secretion of PAI-1 and promotes TNBC cell metastasis, thus forming a positive feedback loop. Moreover, this enhanced metastatic ability is reversible and dependent on CCL5 signaling via the chemokine receptor, CCR5. Of importance, key features of this pathway are manifested in patients with TNBC and in The Cancer Genome Atlas database. Taken together, our results suggest that ECs enhance EMT-induced TNBC cell metastasis via PAI-1 and CCL5 signaling and illustrate the potential of developing new PAI-1- and CCL5-targeting therapy for patients with TNBC.-Zhang, W., Xu, J., Fang, H., Tang, L., Chen, W., Sun, Q., Zhang, Q., Yang, F., Sun, Z., Cao, L., Wang, Y., Guan, X. Endothelial cells promote triple-negative breast cancer cell metastasis via PAI-1 and CCL5 signaling.


Asunto(s)
Quimiocina CCL5/metabolismo , Inhibidor 1 de Activador Plasminogénico/metabolismo , Neoplasias de la Mama Triple Negativas/metabolismo , Animales , Línea Celular Tumoral , Movimiento Celular , Quimiocina CCL5/genética , Células Endoteliales/metabolismo , Células Endoteliales/patología , Transición Epitelial-Mesenquimal , Retroalimentación Fisiológica , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Invasividad Neoplásica , Neovascularización Patológica , Comunicación Paracrina , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores CCR5/genética , Receptores CCR5/metabolismo , Serpina E2/deficiencia , Serpina E2/genética , Serpina E2/metabolismo , Transducción de Señal , Neoplasias de la Mama Triple Negativas/patología , Neoplasias de la Mama Triple Negativas/secundario , Microambiente Tumoral
6.
J Thromb Haemost ; 15(12): 2451-2460, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29028290

RESUMEN

Essentials Vitronectin (VN) is produced by smooth muscle cells (SMCs) and promotes neointima formation. We studied the regulation of vascular VN expression by plasminogen activator inhibitor-1 (PAI-1). PAI-1 stimulates VN gene expression in SMCs by binding LDL receptor-related protein 1. Stimulation of VN gene expression may be a mechanism by which PAI-1 controls vascular remodeling. SUMMARY: Background Increased expression of vitronectin (VN) by smooth muscle cells (SMCs) promotes neointima formation after vascular injury, and may contribute to chronic vascular diseases, such as atherosclerosis. However, the molecular regulation of vascular VN expression is poorly defined. Given the overlapping expression profiles and functions of VN and plasminogen activator inhibitor (PAI)-1, we hypothesized that PAI-1 regulates vascular VN expression. Objectives To determine whether PAI-1 regulates VN expression in SMCs and in vivo. Methods The effects of genetic alterations in PAI-1 expression, pharmacologic PAI-1 inhibition and recombinant PAI-1 on SMC VN expression were studied, and vascular VN expression in wild-type (WT) and PAI-1-deficient mice was assessed. Results VN expression was significantly lower in PAI-1-deficient SMCs and significantly increased in PAI-1-overexpressing SMCs. PAI-1 small interfering RNA and pharmacologic PAI-1 inhibition significantly decreased SMC VN expression. Recombinant PAI-1 stimulated VN expression by binding LDL receptor-related protein-1 (LRP1), but another LRP1 ligand, α2 -macroglobulin, did not. As compared with WT controls, carotid artery VN expression was significantly lower in PAI-1-deficient mice and significantly higher in PAI-1-transgenic mice. In a vein graft (VG) model of intimal hyperplasia, VN expression was significantly attenuated in PAI-1-deficient VGs as compared with WT controls. The plasma VN concentration was significantly decreased in PAI-1-deficient mice versus WT controls at 4 weeks, but not at 5 days or 8 weeks, after surgery. Conclusions PAI-1 stimulates SMC VN expression by binding LRP1, and controls vascular VN expression in vivo. Autocrine regulation of vascular VN expression by PAI-1 may play important roles in vascular homeostasis and pathologic vascular remodeling.


Asunto(s)
Músculo Liso Vascular/metabolismo , Serpina E2/metabolismo , Vitronectina/metabolismo , Animales , Células Cultivadas , Regulación de la Expresión Génica , Humanos , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/metabolismo , Neointima/etiología , Neointima/genética , Neointima/metabolismo , ARN Interferente Pequeño/genética , Receptores de LDL/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Serpina E2/deficiencia , Serpina E2/genética , Proteínas Supresoras de Tumor/metabolismo , Remodelación Vascular , Vitronectina/deficiencia , Vitronectina/genética
7.
BMC Musculoskelet Disord ; 18(1): 392, 2017 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-28893232

RESUMEN

BACKGROUND: Subchondral osteopenia is important for the pathophysiology of osteoarthritis (OA). Although previous studies suggest that plasminogen activator inhibitor-1 (PAI-1), an inhibitor of fibrinolysis, is related to bone metabolism, its role in OA remains unknown. We therefore investigated the roles of PAI-1 in the subchondral bone in OA model mice. METHODS: Wild type (WT) and PAI-1-deficient (KO) mice were ovariectomized (OVX), and then destabilization of the medial meniscus (DMM) surgery was performed. RESULTS: DMM and OVX significantly decreased the trabecular bone mineral density of the subchondral bone evaluated by quantitative computed tomography in PAI-1 KO mice. The effects of OVX and/or PAI-1 deficiency on the OARSI score for the evaluation of the progression of knee degeneration were not significant. PAI-1 deficiency significantly augmented receptor activator nuclear factor κB ligand mRNA levels enhanced by IL-1ß in mouse primary osteoblasts, although it did not affect osteoblast differentiation. Moreover, PAI-1 deficiency significantly increased osteoclast formation from mouse bone marrow cells. CONCLUSION: We showed that PAI-1 deficiency accelerates the subchondral osteopenia after induction of OA in mice. PAI-1 might suppress an enhancement of bone resorption and subsequent subchondral osteopenia after induction of OA in mice.


Asunto(s)
Enfermedades Óseas Metabólicas/metabolismo , Enfermedades Óseas Metabólicas/patología , Osteoartritis/metabolismo , Osteoartritis/patología , Serpina E2/deficiencia , Animales , Enfermedades Óseas Metabólicas/etiología , Femenino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Osteoartritis/complicaciones , Distribución Aleatoria
8.
Thromb Haemost ; 114(2): 379-89, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25925849

RESUMEN

The tumour suppressor phosphatase and tensin homologue (PTEN), mutated or lost in many human cancers, is a major regulator of angiogenesis. However, the cellular mechanism of PTEN regulation in endothelial cells so far remains elusive. Here, we characterise the urokinase receptor (uPAR, CD87) and its tumour-derived soluble form, suPAR, as a key molecule of regulating PTEN in endothelial cells. We observed uPAR-deficient endothelial cells to express enhanced PTEN mRNA- and protein levels. Consistently, uPAR expression in endogenous negative uPAR cells, down-regulated PTEN and activated the PI3K/Akt pathway. Additionally, we found that integrin adhesion receptors act as trans-membrane signaling partners for uPAR to repress PTEN transcription in a NF-κB-dependent manner. Functional in vitro assays with endothelial cells, derived from uPAR-deficient and PTEN heterozygous crossbred mice, demonstrated the impact of uPAR-dependent PTEN regulation on cell motility and survival. In an in vivo murine angiogenesis model uPAR-deficient PTEN heterozygous animals increased the impaired angiogenic phenotype of uPAR knockout mice and were able to reverse the high invasive potential of PTEN heterozygots. Our data provide first evidence that endogenous as well as exogenous soluble uPAR down-regulated PTEN in endothelial cells to support angiogenesis. The uPAR-induced PTEN regulation might represent a novel target for drug interference, and may lead to the development of new therapeutic strategies in anti-angiogenic treatment.


Asunto(s)
Células Endoteliales/metabolismo , Regulación de la Expresión Génica , Neovascularización Fisiológica/genética , Fosfohidrolasa PTEN/biosíntesis , Receptores del Activador de Plasminógeno Tipo Uroquinasa/fisiología , Animales , Movimiento Celular , Células Cultivadas , Regulación hacia Abajo , Activación Enzimática , Células HEK293 , Humanos , Integrinas/metabolismo , Pulmón/irrigación sanguínea , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Fosfohidrolasa PTEN/deficiencia , Fosfohidrolasa PTEN/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores del Activador de Plasminógeno Tipo Uroquinasa/deficiencia , Receptores del Activador de Plasminógeno Tipo Uroquinasa/genética , Proteínas Recombinantes de Fusión/metabolismo , Serpina E2/deficiencia , Transducción de Señal , Migración Transendotelial y Transepitelial , Transfección
9.
Andrologia ; 47(10): 1147-52, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25557984

RESUMEN

Myoendothelial junctions are specialised projections of cell : cell contact through the internal elastic lamina between endothelial cells and vascular smooth muscle cells. These junctions allow for endothelial cells and vascular smooth muscle cells to make direct membrane apposition and are involved in cell : cell communication. In this study, we evaluated for the presence of myoendothelial junctions in murine corporal tissue and used plasminogen activator inhibitor (PAI)-1-deficient mice, which lack myoendothelial junctions, to determine whether myoendothelial junctions affect erectile function. Transmission electron microscopy demonstrated the presence of myoendothelial junctions in the corporal tissue of wild-type mice and confirmed the decreased junction numbers in the tissue of PAI-1(-/-) mice. A potential role for myoendothelial junctions in tumescence was established; in that, PAI-1(-/-) mice demonstrated a significantly longer time to achieve maximal intracavernous pressure. Treatment of PAI-1(-/-) mice with recombinant PAI-1 restored the number of myoendothelial junctions in the corporal tissue and also induced a significant decrease in time to maximal corporal pressures. Myoendothelial junctions were similarly identified in the human corporal tissue. These results suggest a critical role for myoendothelial junctions in erectile pathophysiology and therapies aimed at restoring myoendothelial junction numbers in the corporal tissue may provide a novel therapy for erectile dysfunction.


Asunto(s)
Endotelio Vascular/efectos de los fármacos , Disfunción Eréctil/tratamiento farmacológico , Uniones Intercelulares/efectos de los fármacos , Músculo Liso Vascular/efectos de los fármacos , Erección Peniana/efectos de los fármacos , Serpina E2/deficiencia , Animales , Comunicación Celular , Endotelio Vascular/fisiología , Endotelio Vascular/ultraestructura , Disfunción Eréctil/etiología , Uniones Intercelulares/fisiología , Uniones Intercelulares/ultraestructura , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Electrónica de Transmisión , Músculo Liso Vascular/fisiología , Músculo Liso Vascular/ultraestructura , Proteínas Recombinantes , Serpina E2/uso terapéutico
10.
J Neuropathol Exp Neurol ; 73(5): 387-402, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24709679

RESUMEN

Intracerebral-intraventricular hemorrhages (ICH/IVH) in very preterm neonates are responsible for high mortality and subsequent disabilities. In humans, tissue plasminogen activator (t-PA) initiates fibrinolysis and activates endoluminal-endothelial receptors; dysfunction of the t-PA inhibitor (PAI-1) results in recurrent hemorrhages. We used PAI-1 knockout (PAI-1) mice to examine the role of t-PA in age-dependent intracranial hemorrhages as a possible model of preterm ICH/IVH. Intracortical injection of 2 µL of phosphate-buffered saline produced a small traumatic injury and a high rate of hemorrhage in PAI-1 pups at postnatal day 3 (P3) or P5, whereas it had no effect in wild-type neonates. This resulted in white matter and cortical lesions, ventricle enlargement, hyperlocomotion, and altered cortical levels of serotonin and dopamine in the adult PAI mice. N-methyl-D-aspartate receptor blockers, plasmin- and matrix metalloproteinases inhibitors reduced hemorrhage and tissue lesions. In contrast to P3 to P5, no significant hemorrhages were induced in P10 PAI-1 pups and there were no behavioral or neurochemical alterations in adulthood. These data suggest that microvascular immaturity up to P5 in mice is a determinant factor required for t-PA-dependent vascular rupture. Neonatal PAI-1 mice could be a useful ICH/IVH model for studying the ontogenic window of vascular immaturity and vascular protection against later neurodisabilities.


Asunto(s)
Envejecimiento/fisiología , Hemorragia Cerebral/metabolismo , Hemorragia Cerebral/fisiopatología , Serpina E2/deficiencia , Factores de Edad , Ácido Aminocaproico/administración & dosificación , Animales , Animales Recién Nacidos , Aprotinina/administración & dosificación , Hemorragia Cerebral/genética , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Actividad Motora/efectos de los fármacos , Actividad Motora/fisiología , Fenotipo , Serpina E2/antagonistas & inhibidores , Serpina E2/fisiología
11.
Int J Mol Sci ; 14(8): 16719-31, 2013 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-23949634

RESUMEN

The serine protease thrombin plays a role in signalling ischemic neuronal death in the brain. Paradoxically, endogenous neuroprotective mechanisms can be triggered by preconditioning with thrombin (thrombin preconditioning, TPC), leading to tolerance to cerebral ischemia. Here we studied the role of thrombin's endogenous potent inhibitor, protease nexin-1 (PN-1), in ischemia and in tolerance to cerebral ischemia induced by TPC. Cerebral ischemia was modelled in vitro in organotypic hippocampal slice cultures from rats or genetically engineered mice lacking PN-1 or with the reporter gene lacZ knocked into the PN-1 locus PN-1HAPN-1-lacZ/HAPN-1-lacZ (PN-1 KI) exposed to oxygen and glucose deprivation (OGD). We observed increased thrombin enzyme activity in culture homogenates 24 h after OGD. Lack of PN-1 increased neuronal death in the CA1, suggesting that endogenous PN-1 inhibits thrombin-induced neuronal damage after ischemia. OGD enhanced ß-galactosidase activity, reflecting PN-1 expression, at one and 24 h, most strikingly in the stratum radiatum, a glial cell layer adjacent to the CA1 layer of ischemia sensitive neurons. TPC, 24 h before OGD, additionally increased PN-1 expression 1 h after OGD, compared to OGD alone. TPC failed to induce tolerance in cultures from PN-1(-/-) mice confirming PN-1 as an important TPC target. PN-1 upregulation after TPC was blocked by the c-Jun N-terminal kinase (JNK) inhibitor, L-JNKI1, known to block TPC. This work suggests that PN-1 is an endogenous neuroprotectant in cerebral ischemia and a potential target for neuroprotection.


Asunto(s)
Isquemia Encefálica/metabolismo , Hipoxia de la Célula/fisiología , Glucosa/deficiencia , Hipocampo/metabolismo , Serpina E2/metabolismo , Animales , Encéfalo/metabolismo , Muerte Celular , Técnicas de Sustitución del Gen , Glucosa/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/metabolismo , Péptidos/farmacología , Ratas , Ratas Sprague-Dawley , Serpina E2/deficiencia , Serpina E2/genética , Accidente Cerebrovascular/metabolismo , Trombina/antagonistas & inhibidores , Trombina/metabolismo
12.
Arterioscler Thromb Vasc Biol ; 33(7): 1647-54, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23661674

RESUMEN

OBJECTIVE: Human protein C is a plasma serine protease that plays a key role in hemostasis, and activated protein C (aPC) is known to elicit protective responses in vascular endothelial cells. This cytoprotective activity requires the interaction of the protease with its cell membrane receptor, endothelial protein C receptor. However, the mechanisms regulating the beneficial cellular effects of aPC are not well known. We aimed to determine whether a serine protease inhibitor called protease nexin-1 (PN-1) or serpinE2, expressed by vascular cells, can modulate the effect of aPC on endothelial cells. APPROACH AND RESULTS: We found that vascular barrier protective and antiapoptotic activities of aPC were reduced both in endothelial cells underexpressing PN-1 and in endothelial cells whose PN-1 function was blocked by a neutralizing antibody. Our in vitro data were further confirmed in vivo. Indeed, we found that vascular endothelial growth factor-mediated hyperpermeability in the skin of mice was markedly reduced by local intradermal injection of aPC in wild-type mice but not in PN-1-deficient mice. Furthermore, we demonstrated a previously unknown protective role of endothelial PN-1 on endothelial protein C receptor shedding. We provided evidence that PN-1 inhibits furin, a serine protease that activates a disintegrin and metalloproteinase 17 involved in the shedding of endothelial protein C receptor. We indeed evidenced a direct interaction between PN-1 and furin in endothelial cells. CONCLUSIONS: Our results thus demonstrate an original role of PN-1 as a furin convertase inhibitor, providing new insights for understanding the regulation of endothelial protein C receptor-dependent aPC endothelial protective effects.


Asunto(s)
Proteínas ADAM/metabolismo , Antígenos CD/metabolismo , Células Endoteliales/enzimología , Furina/metabolismo , Glicoproteínas/metabolismo , Receptores de Superficie Celular/metabolismo , Serpina E2/metabolismo , Piel/irrigación sanguínea , Proteínas ADAM/antagonistas & inhibidores , Proteínas ADAMTS , Animales , Anticuerpos Neutralizantes/farmacología , Antígenos CD/genética , Apoptosis , Permeabilidad Capilar , Línea Celular , Células Endoteliales/efectos de los fármacos , Receptor de Proteína C Endotelial , Regulación de la Expresión Génica , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Interferencia de ARN , ARN Mensajero/metabolismo , Receptores de Superficie Celular/genética , Serpina E2/antagonistas & inhibidores , Serpina E2/deficiencia , Serpina E2/genética , Transducción de Señal , Transfección
13.
Am J Pathol ; 180(6): 2321-9, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22507835

RESUMEN

Hepatic fibrin(ogen) has been noted to occur after acetaminophen (APAP)-induced liver injury in mice. Deficiency in plasminogen activator inhibitor-1 (PAI-1), an endogenous inhibitor of fibrinolysis, increases APAP-induced liver injury in mice. However, the roles of fibrinogen and fibrinolysis in APAP-induced liver injury are not known. We tested the hypothesis that hepatic fibrin(ogen) deposition reduces severity of APAP-induced liver injury. APAP-induced (300 mg/kg) liver injury in mice was accompanied by thrombin generation, consumption of plasma fibrinogen, and deposition of hepatic fibrin. Neither fibrinogen depletion with ancrod nor complete fibrinogen deficiency [via knockout of the fibrinogen alpha chain gene (Fbg(-/-))] affected APAP-induced liver injury. PAI-1 deficiency (PAI-1(-/-)) increased APAP-induced liver injury and hepatic fibrin deposition 6 hours after APAP administration, which was followed by marked hemorrhage at 24 hours. As in PAI-1(-/-) mice, administration of recombinant tissue plasminogen activator (tenecteplase, 5 mg/kg) worsened APAP-induced liver injury and hemorrhage in wild-type mice. In contrast, APAP-induced liver injury was reduced in both plasminogen-deficient mice and in wild-type mice treated with tranexamic acid, an inhibitor of plasminogen activation. Activation of matrix metalloproteinase 9 (MMP-9) paralleled injury, but MMP-9 deficiency did not affect APAP-induced liver injury. The results indicate that fibrin(ogen) does not contribute to development of APAP-induced liver injury and suggest rather that plasminogen activation contributes to APAP-induced liver injury.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas/fisiopatología , Fibrina/fisiología , Activadores Plasminogénicos/fisiología , Acetaminofén , Alanina Transaminasa/sangre , Animales , Antitrombina III , Coagulación Sanguínea/fisiología , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Sinergismo Farmacológico , Fibrinógeno/metabolismo , Fibrinólisis/fisiología , Hemorragia/inducido químicamente , Hígado/metabolismo , Masculino , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Péptido Hidrolasas/sangre , Serpina E2/deficiencia , Trombina/biosíntesis , Activador de Tejido Plasminógeno
14.
Br J Haematol ; 157(3): 291-8, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22360729

RESUMEN

Plasminogen activator inhibitor-1 (PAI-1, also known as SERPINE1) is a member of the serine protease inhibitor (SERPIN) superfamily and is the primary physiological regulator of urokinase-type plasminogen activator (uPA) and tissue-type plasminogen activator (tPA) activity. Although the principal function of PAI-1 is the inhibition of fibrinolysis, PAI-1 possesses pleiotropic functions besides haemostasis. In the quarter century since its discovery, a number of studies have focused on improving our understanding of PAI-1 functions in vivo and in vitro. The use of Serpine1-deficient mice has particularly enhanced our understanding of the functions of PAI-1 in various physiological and pathophysiological conditions. In this review, the results of recent studies on PAI-1 and its role in clinical conditions are discussed.


Asunto(s)
Hemostasis/fisiología , Inhibidor 1 de Activador Plasminogénico/fisiología , Animales , Aterosclerosis/fisiopatología , Modelos Animales de Enfermedad , Fibrosis/fisiopatología , Humanos , Inflamación/fisiopatología , Síndrome Metabólico/fisiopatología , Ratones , Ratones Noqueados , Neoplasias/fisiopatología , Serpina E2/deficiencia
15.
Blood ; 118(8): 2313-21, 2011 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-21734232

RESUMEN

Plasminogen activator inhibitor-1 (PAI-1) is increased in the lungs of patients with pulmonary fibrosis, and animal studies have shown that experimental manipulations of PAI-1 levels directly influence the extent of scarring that follows lung injury. PAI-1 has 2 known properties that could potentiate fibrosis, namely an antiprotease activity that inhibits the generation of plasmin, and a vitronectin-binding function that interferes with cell adhesion to this extracellular matrix protein. To determine the relative importance of each PAI-1 function in lung fibrogenesis, we administered mutant PAI-1 proteins that possessed either intact antiprotease or vitronectin-binding activity to bleomycin-injured mice genetically deficient in PAI-1. We found that the vitronectin-binding capacity of PAI-1 was the primary determinant required for its ability to exacerbate lung scarring induced by intratracheal bleomycin administration. The critical role of the vitronectin-binding function of PAI-1 in fibrosis was confirmed in the bleomycin model using mice genetically modified to express the mutant PAI-1 proteins. We conclude that the vitronectin-binding function of PAI-1 is necessary and sufficient in its ability to exacerbate fibrotic processes in the lung.


Asunto(s)
Fibrosis Pulmonar/etiología , Fibrosis Pulmonar/metabolismo , Serpina E2/metabolismo , Vitronectina/metabolismo , Animales , Bleomicina/administración & dosificación , Bleomicina/toxicidad , Líquido del Lavado Bronquioalveolar/química , Colágeno/metabolismo , Modelos Animales de Enfermedad , Humanos , Hidroxiprolina/metabolismo , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Inhibidor 1 de Activador Plasminogénico/genética , Inhibidor 1 de Activador Plasminogénico/metabolismo , Unión Proteica , Fibrosis Pulmonar/patología , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Serpina E2/sangre , Serpina E2/deficiencia , Serpina E2/genética , Vitronectina/sangre
16.
Arterioscler Thromb Vasc Biol ; 31(8): 1781-7, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21571686

RESUMEN

OBJECTIVE: The role of plasminogen activator inhibitor-1 (PAI-1) in vein graft (VG) remodeling is undefined. We examined the effect of PAI-1 on VG intimal hyperplasia and tested the hypothesis that PAI-1 regulates VG thrombin activity. METHODS AND RESULTS: VGs from wild-type (WT), Pai1(-/-), and PAI-1-transgenic mice were implanted into WT, Pai1(-/-), or PAI-1-transgenic arteries. VG remodeling was assessed 4 weeks later. Intimal hyperplasia was significantly greater in PAI-1-deficient mice than in WT mice. The proliferative effect of PAI-1 deficiency was retained in vitronectin-deficient mice, suggesting that PAI-1's antiproteolytic function plays a key role in regulating intimal hyperplasia. Thrombin-induced proliferation of PAI-1-deficient venous smooth muscle cells (SMC) was significantly greater than that of WT SMC, and thrombin activity was significantly higher in PAI-1-deficient VGs than in WT VGs. Increased PAI-1 expression, which has been associated with obstructive VG disease, did not increase intimal hyperplasia. CONCLUSIONS: Decreased PAI-1 expression (1) promotes intimal hyperplasia by pathways that do not require vitronectin and (2) increases thrombin activity in VG. PAI-1 overexpression, although it promotes SMC migration in vitro, did not increase intimal hyperplasia. These results challenge the concept that PAI-1 drives nonthrombotic obstructive disease in VG and suggest that PAI-1's antiproteolytic function, including its antithrombin activity, inhibits intimal hyperplasia.


Asunto(s)
Serpina E2/fisiología , Vena Cava Inferior/trasplante , Animales , Movimiento Celular , Proliferación Celular , Puente de Arteria Coronaria/efectos adversos , Fibrina/metabolismo , Fibrinógeno/metabolismo , Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Miocitos del Músculo Liso/patología , Miocitos del Músculo Liso/fisiología , Neointima/etiología , Neointima/patología , Neointima/fisiopatología , Serpina E2/deficiencia , Serpina E2/genética , Túnica Íntima/patología , Vena Cava Inferior/patología , Vitronectina/deficiencia
17.
J Thromb Haemost ; 9(6): 1200-6, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21486382

RESUMEN

BACKGROUND: Plasminogen activator inhibitor-1 (PAI-1) is the primary physiological regulator of urokinase plasminogen activator (uPA) and tissue plasminogen activator (tPA) activity. A number of studies have shown that elevated levels of PAI-1 are related to pathological states such as an increased risk of arterial thrombotic events and a poor prognosis for cancer patients; however, there are few reports about PAI-1 deficiency in humans because the disorder is very rare. OBJECTIVE: To understand the in vivo impact of a complete PAI-1 deficiency, Serpine1(-/-) mice were generated; a number of in vivo studies have been conducted to elucidate the function of PAI-1 using Serpine1(-/-) mice. The phenotypes demonstrated in Serpine1(-/-) mice, however, were quite different from those in humans. Therefore, it is necessary to find out and analyze SERPINE1 deficiency in humans. PATIENT AND METHODS: The patient is a 47-year-old woman who has had multiple episodes of major bleeding. Although most of the patient's blood coagulation factors were functionally normal, her PAI-1 antigen levels were undetectable. Therefore, DNA sequencing of the SERPINE1 gene were analyzed. RESULTS: The proband had a homozygous 1-bp duplication (C) at exon 3 (c.356dupC; p.Ile120AspfsX42). Both wild-type PAI-1 (42.7 kDa) and mutated (Mut) PAI-1 (14.7kDa) were expressed in COS-1 cells, although the level of Mut PAI-1 expressed in the cell lysates was much lower. Wild-type PAI-1 was observed in the culture supernatant, whereas no Mut PAI-1 was detected in the supernatant. CONCLUSIONS: Considering the results of the present study, the translation of mouse studies to humans must be performed with great care.


Asunto(s)
Hemorragia/etiología , Inhibidor 1 de Activador Plasminogénico/deficiencia , Serpina E2/deficiencia , Cicatrización de Heridas , Animales , Enfermedad Crítica , Femenino , Homocigoto , Humanos , Ratones , Ratones Noqueados , Persona de Mediana Edad , Mutación , Fenotipo , Inhibidor 1 de Activador Plasminogénico/genética , Análisis de Secuencia de ADN , Serpina E2/genética , Transfección
18.
Physiol Genomics ; 43(4): 188-98, 2011 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-21119013

RESUMEN

Murine placentation is associated with the invasion of maternal endometrium by trophoblasts and an extensive maternal and fetal angiogenesis. Plasminogen activator inhibitor-1 (PAI-1) is transiently produced by spongiotrophoblasts and trophoblast giant cells at 10.5-11.5 days postcoitum (dpc). Knowing the key contribution of PAI-1 in the regulation of angiogenesis, we have now analyzed the consequence of PAI-1 deficiency on murine placentation. Morphological and quantitative computer-assisted image analysis revealed abnormal placental morphology in PAI-1-/- mice at 10.5 and 12.5 dpc. At 10.5 dpc, the genetic ablation of PAI-1 resulted in a transient reduction of both maternal and fetal vascularizations in the placenta and increased trophoblast cell density. This was associated with a poorer development of the labyrinth and an extension of the decidua. A larger spongiotrophoblast layer appeared at 12.5 dpc in PAI-1-deficient mice. Placental morphology was normalized at 14.5 dpc. Microarray analyses performed on laser capture microdissected labyrinths revealed that 46 genes were differentially expressed between the two genotypes at 10.5 dpc. However, only 11 genes were still differently modulated at 14.5 dpc, when normalization of placental morphology had taken place. This transcriptomic profiling highlighted a dysregulation in the expression of placenta-related cathepsin family members. Altogether our data provide evidence for a transient impaired placental morphology in PAI-1-deficient mice that is then normalized, leading to normal embryonic development.


Asunto(s)
Neovascularización Fisiológica , Placenta/irrigación sanguínea , Serpina E2/deficiencia , Animales , Decidua/citología , Decidua/metabolismo , Implantación del Embrión/genética , Femenino , Feto/irrigación sanguínea , Feto/citología , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Procesamiento de Imagen Asistido por Computador , Ratones , Fenotipo , Placenta/citología , Placenta/metabolismo , Embarazo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Serpina E2/genética , Serpina E2/metabolismo , Trofoblastos/citología , Trofoblastos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...