Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
J Biol Chem ; 300(6): 107313, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38657864

RESUMEN

Sortilin-related receptor 1 (SORL1) is an intracellular sorting receptor genetically implicated in Alzheimer's disease (AD) that impacts amyloid precursor protein trafficking. The objective of these studies was to test the hypothesis that SORL1 binds tau, modulates its cellular trafficking and impacts the aggregation of cytoplasmic tau induced by pathological forms of tau. Using surface plasmon resonance measurements, we observed high-affinity binding of tau to SORL1 and the vacuolar protein sorting 10 domain of SORL1. Interestingly, unlike LDL receptor-related protein 1, SORL1 binds tau at both pH 7.4 and pH 5.5, revealing its ability to bind tau at endosomal pH. Immunofluorescence studies confirmed that exogenously added tau colocalized with SORL1 in H4 neuroglioma cells, while overexpression of SORL1 in LDL receptor-related protein 1-deficient Chinese hamster ovary (CHO) cells resulted in a marked increase in the internalization of tau, indicating that SORL1 can bind and mediate the internalization of monomeric forms of tau. We further demonstrated that SORL1 mediates tau seeding when tau RD P301S FRET biosensor cells expressing SORL1 were incubated with high molecular weight forms of tau isolated from the brains of patients with AD. Seeding in H4 neuroglioma cells is significantly reduced when SORL1 is knocked down with siRNA. Finally, we demonstrate that the N1358S mutant of SORL1 significantly increases tau seeding when compared to WT SORL1, identifying for the first time a potential mechanism that connects this specific SORL1 mutation to Alzheimer's disease. Together, these studies identify SORL1 as a receptor that contributes to trafficking and seeding of pathogenic tau.


Asunto(s)
Cricetulus , Proteínas Relacionadas con Receptor de LDL , Proteínas de Transporte de Membrana , Proteínas tau , Humanos , Proteínas tau/metabolismo , Proteínas tau/genética , Animales , Células CHO , Proteínas Relacionadas con Receptor de LDL/metabolismo , Proteínas Relacionadas con Receptor de LDL/genética , Proteínas de Transporte de Membrana/metabolismo , Proteínas de Transporte de Membrana/genética , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Línea Celular Tumoral , Unión Proteica , Transporte de Proteínas
2.
Arterioscler Thromb Vasc Biol ; 40(6): 1479-1490, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32268785

RESUMEN

OBJECTIVE: Enhanced expression of PAI-1 (plasminogen activator inhibitor-1) has been implicated in atherosclerosis formation in humans with obesity and metabolic syndrome. However, little is known about the effects of pharmacological targeting of PAI-1 on atherogenesis. This study examined the effects of pharmacological PAI-1 inhibition on atherosclerosis formation in a murine model of obesity and metabolic syndrome. Approach and Results: LDL receptor-deficient (ldlr-/-) mice were fed a Western diet high in cholesterol, fat, and sucrose to induce obesity, metabolic dysfunction, and atherosclerosis. Western diet triggered significant upregulation of PAI-1 expression compared with normal diet controls. Addition of a pharmacological PAI-1 inhibitor (either PAI-039 or MDI-2268) to Western diet significantly inhibited obesity and atherosclerosis formation for up to 24 weeks without attenuating food consumption. Pharmacological PAI-1 inhibition significantly decreased macrophage accumulation and cell senescence in atherosclerotic plaques. Recombinant PAI-1 stimulated smooth muscle cell senescence, whereas a PAI-1 mutant defective in LRP1 (LDL receptor-related protein 1) binding did not. The prosenescent effect of PAI-1 was blocked by PAI-039 and R2629, a specific anti-LRP1 antibody. PAI-039 significantly decreased visceral adipose tissue inflammation, hyperglycemia, and hepatic triglyceride content without altering plasma lipid profiles. CONCLUSIONS: Pharmacological targeting of PAI-1 inhibits atherosclerosis in mice with obesity and metabolic syndrome, while inhibiting macrophage accumulation and cell senescence in atherosclerotic plaques, as well as obesity-associated metabolic dysfunction. PAI-1 induces senescence of smooth muscle cells in an LRP1-dependent manner. These results help to define the role of PAI-1 in atherosclerosis formation and suggest a new plasma-lipid-independent strategy for inhibiting atherogenesis.


Asunto(s)
Aterosclerosis/prevención & control , Síndrome Metabólico/tratamiento farmacológico , Inhibidor 1 de Activador Plasminogénico/efectos de los fármacos , Animales , Senescencia Celular/efectos de los fármacos , Dieta Occidental , Modelos Animales de Enfermedad , Ácidos Indolacéticos/administración & dosificación , Macrófagos/efectos de los fármacos , Macrófagos/patología , Síndrome Metabólico/patología , Síndrome Metabólico/prevención & control , Ratones , Ratones Noqueados , Obesidad/etiología , Obesidad/prevención & control , Placa Aterosclerótica/patología , Inhibidor 1 de Activador Plasminogénico/fisiología , Receptores de LDL/deficiencia , Receptores de LDL/genética
3.
Sci Transl Med ; 11(482)2019 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-30842312

RESUMEN

There is a major unmet clinical need to identify pathways in inflammatory bowel disease (IBD) to classify patient disease activity, stratify patients that will benefit from targeted therapies such as anti-tumor necrosis factor (TNF), and identify new therapeutic targets. In this study, we conducted global transcriptome analysis to identify IBD-related pathways using colon biopsies, which highlighted the coagulation gene pathway as one of the most enriched gene sets in patients with IBD. Using this gene-network analysis across 14 independent cohorts and 1800 intestinal biopsies, we found that, among the coagulation pathway genes, plasminogen activator inhibitor-1 (PAI-1) expression was highly enriched in active disease and in patients with IBD who did not respond to anti-TNF biologic therapy and that PAI-1 is a key link between the epithelium and inflammation. Functionally, PAI-1 and its direct target, the fibrinolytic protease tissue plasminogen activator (tPA), played an important role in regulating intestinal inflammation. Intestinal epithelial cells produced tPA, which was protective against chemical and mechanical-mediated colonic injury in mice. In contrast, PAI-1 exacerbated mucosal damage by blocking tPA-mediated cleavage and activation of anti-inflammatory TGF-ß, whereas the inhibition of PAI-1 reduced both mucosal damage and inflammation. This study identifies an immune-coagulation gene axis in IBD where elevated PAI-1 may contribute to more aggressive disease.


Asunto(s)
Colitis/metabolismo , Colitis/patología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Inhibidor 1 de Activador Plasminogénico/metabolismo , Animales , Factores Biológicos/farmacología , Factores Biológicos/uso terapéutico , Coagulación Sanguínea , Proliferación Celular/efectos de los fármacos , Citrobacter/efectos de los fármacos , Colitis/inmunología , Colitis/microbiología , Colon/patología , Citocinas/metabolismo , Inflamación/patología , Enfermedades Inflamatorias del Intestino/sangre , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Enfermedades Inflamatorias del Intestino/patología , Interleucina-17/metabolismo , Ratones , Índice de Severidad de la Enfermedad , Bibliotecas de Moléculas Pequeñas/farmacología , Células Th17/inmunología , Activador de Tejido Plasminógeno/metabolismo , Transcripción Genética , Factor de Crecimiento Transformador beta/metabolismo
4.
Acta Neuropathol ; 134(4): 585-604, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28725968

RESUMEN

Treatment of acute ischemic stroke with the thrombolytic tissue plasminogen activator (tPA) can significantly improve neurological outcomes; however, thrombolytic therapy is associated with an increased risk of intra-cerebral hemorrhage (ICH). Previously, we demonstrated that during stroke tPA acting on the parenchymal side of the neurovascular unit (NVU) can increase blood-brain barrier (BBB) permeability and ICH through activation of latent platelet-derived growth factor-CC (PDGF-CC) and signaling by the PDGF receptor-α (PDGFRα). However, in vitro, activation of PDGF-CC by tPA is very inefficient and the mechanism of PDGF-CC activation in the NVU is not known. Here, we show that the integrin Mac-1, expressed on brain microglia/macrophages (denoted microglia throughout), acts together with the endocytic receptor LRP1 in the NVU to promote tPA-mediated activation of PDGF-CC. Mac-1-deficient mice (Mac-1-/-) are protected from tPA-induced BBB permeability but not from permeability induced by intracerebroventricular injection of active PDGF-CC. Immunofluorescence analysis demonstrates that Mac-1, LRP1, and the PDGFRα all localize to the NVU of arterioles, and following middle cerebral artery occlusion (MCAO) Mac-1-/- mice show significantly less PDGFRα phosphorylation, BBB permeability, and infarct volume compared to wild-type mice. Bone-marrow transplantation studies indicate that resident CD11b+ cells, but not bone-marrow-derived leukocytes, mediate the early activation of PDGF-CC by tPA after MCAO. Finally, using a model of thrombotic stroke with late thrombolysis, we show that wild-type mice have an increased incidence of spontaneous ICH following thrombolysis with tPA 5 h after MCAO, whereas Mac-1-/- mice are resistant to the development of ICH even with late tPA treatment. Together, these results indicate that Mac-1 and LRP1 act as co-factors for the activation of PDGF-CC by tPA in the NVU, and suggest a novel mechanism for tightly regulating PDGFRα signaling in the NVU and controlling BBB permeability.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Isquemia Encefálica/metabolismo , Permeabilidad Capilar/fisiología , Linfocinas/metabolismo , Microglía/metabolismo , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Accidente Cerebrovascular/metabolismo , Animales , Arteriolas/efectos de los fármacos , Arteriolas/metabolismo , Arteriolas/patología , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/patología , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/patología , Antígeno CD11b/metabolismo , Permeabilidad Capilar/efectos de los fármacos , Células Cultivadas , Hemorragia Cerebral/inducido químicamente , Hemorragia Cerebral/metabolismo , Hemorragia Cerebral/patología , Modelos Animales de Enfermedad , Femenino , Fibrinolíticos/efectos adversos , Fibrinolíticos/farmacología , Leucocitos/metabolismo , Leucocitos/patología , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad , Antígeno de Macrófago-1/genética , Antígeno de Macrófago-1/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/patología , Receptores de LDL/metabolismo , Accidente Cerebrovascular/tratamiento farmacológico , Accidente Cerebrovascular/patología , Activador de Tejido Plasminógeno/efectos adversos , Activador de Tejido Plasminógeno/farmacología , Proteínas Supresoras de Tumor/metabolismo
5.
Pharmacol Ther ; 167: 108-119, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27524729

RESUMEN

Neurological disorders account for a majority of non-malignant disability in humans and are often associated with dysfunction of the blood-brain barrier (BBB). Recent evidence shows that despite apparent variation in the origin of neural damage, the central nervous system has a common injury response mechanism involving platelet-derived growth factor (PDGF)-CC activation in the neurovascular unit and subsequent dysfunction of BBB integrity. Inhibition of PDGF-CC signaling with imatinib in mice has been shown to prevent BBB dysfunction and have neuroprotective effects in acute damage conditions, including traumatic brain injury, seizures or stroke, as well as in neurodegenerative diseases that develop over time, including multiple sclerosis and amyotrophic lateral sclerosis. Stroke and traumatic injuries are major risk factors for age-associated neurodegenerative disorders and we speculate that restoring BBB properties through PDGF-CC inhibition might provide a common therapeutic opportunity for treatment of both acute and progressive neuropathology in humans. In this review we will summarize what is known about the role of PDGF-CC in neurovascular signaling events and the variety of seemingly different neuropathologies it is involved in. We will also discuss the pharmacological means of therapeutic interventions for anti-PDGF-CC therapy and ongoing clinical trials. In summary: inhibition of PDGF-CC signaling can be protective for immediate injury and decrease the long-term neurodegenerative consequences.


Asunto(s)
Linfocinas/metabolismo , Enfermedades del Sistema Nervioso/tratamiento farmacológico , Enfermedades Neurodegenerativas/tratamiento farmacológico , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Animales , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/patología , Diseño de Fármacos , Humanos , Ratones , Terapia Molecular Dirigida , Enfermedades del Sistema Nervioso/etiología , Enfermedades del Sistema Nervioso/fisiopatología , Enfermedades Neurodegenerativas/etiología , Enfermedades Neurodegenerativas/fisiopatología , Fármacos Neuroprotectores/farmacología , Factores de Riesgo , Transducción de Señal
6.
Blood ; 127(9): 1085-96, 2016 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-26647393

RESUMEN

Extravascular fibrin deposition accompanies many human diseases and causes chronic inflammation and organ damage, unless removed in a timely manner. Here, we used intravital microscopy to investigate how fibrin is removed from extravascular space. Fibrin placed into the dermis of mice underwent cellular endocytosis and lysosomal targeting, revealing a novel intracellular pathway for extravascular fibrin degradation. A C-C chemokine receptor type 2 (CCR2)-positive macrophage subpopulation constituted the majority of fibrin-uptaking cells. Consequently, cellular fibrin uptake was diminished by elimination of CCR2-expressing cells. The CCR2-positive macrophage subtype was different from collagen-internalizing M2-like macrophages. Cellular fibrin uptake was strictly dependent on plasminogen and plasminogen activator. Surprisingly, however, fibrin endocytosis was unimpeded by the absence of the fibrin(ogen) receptors, αMß2 and ICAM-1, the myeloid cell integrin-binding site on fibrin or the endocytic collagen receptor, the mannose receptor. The study identifies a novel fibrin endocytic pathway engaged in extravascular fibrin clearance and shows that interstitial fibrin and collagen are cleared by different subsets of macrophages employing distinct molecular pathways.


Asunto(s)
Endocitosis , Fibrina/metabolismo , Macrófagos/metabolismo , Receptores CCR2/metabolismo , Animales , Bioensayo , Receptor 1 de Quimiocinas CX3C , Proliferación Celular , Fibrinolisina/metabolismo , Ratones , Células Mieloides/metabolismo , Plasminógeno/metabolismo , Activadores Plasminogénicos/metabolismo , Proteolisis , Receptores de Quimiocina/metabolismo , Receptores de Péptidos/metabolismo
7.
Arterioscler Thromb Vasc Biol ; 35(1): 111-20, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25378411

RESUMEN

OBJECTIVE: Plasminogen activator inhibitor-1 (PAI-1) regulates angiogenesis via effects on extracellular matrix proteolysis and cell adhesion. However, no previous study has implicated PAI-1 in controlling vascular endothelial growth factor (VEGF) signaling. We tested the hypothesis that PAI-1 downregulates VEGF receptor-2 (VEGFR-2) activation by inhibiting a vitronectin-dependent cooperative binding interaction between VEGFR-2 and αVß3. APPROACH AND RESULTS: We studied effects of PAI-1 on VEGF signaling in human umbilical vein endothelial cells. PAI-1 inhibited VEGF-induced phosphorylation of VEGFR-2 in human umbilical vein endothelial cells grown on vitronectin, but not on fibronectin or collagen. PAI-1 inhibited the binding of VEGFR-2 to ß3 integrin, VEGFR-2 endocytosis, and intracellular signaling pathways downstream of VEGFR-2. The anti-VEGF effect of PAI-1 was mediated by 2 distinct pathways, one requiring binding to vitronectin and another requiring binding to very low-density lipoprotein receptor. PAI-1 inhibited VEGF-induced angiogenesis in vitro and in vivo, and pharmacological inhibition of PAI-1 promoted collateral arteriole development and recovery of hindlimb perfusion after femoral artery interruption. CONCLUSIONS: PAI-1 inhibits activation of VEGFR-2 by VEGF by disrupting a vitronectin-dependent proangiogenic binding interaction involving αVß3 and VEGFR-2. These results broaden our understanding of the roles of PAI-1, vitronectin, and endocytic receptors in regulating VEGFR-2 activation and suggest novel therapeutic strategies for regulating VEGF signaling.


Asunto(s)
Células Endoteliales/metabolismo , Integrina alfaVbeta3/metabolismo , Músculo Esquelético/irrigación sanguínea , Neovascularización Fisiológica , Inhibidor 1 de Activador Plasminogénico/metabolismo , Receptor Cross-Talk , Transducción de Señal , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Adhesión Celular , Movimiento Celular , Células Cultivadas , Modelos Animales de Enfermedad , Endocitosis , Células Endoteliales/efectos de los fármacos , Miembro Posterior , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Ácidos Indolacéticos/administración & dosificación , Isquemia/metabolismo , Isquemia/fisiopatología , Isquemia/prevención & control , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Neovascularización Fisiológica/efectos de los fármacos , Fosforilación , Inhibidor 1 de Activador Plasminogénico/genética , Interferencia de ARN , Receptor Cross-Talk/efectos de los fármacos , Receptores de LDL/metabolismo , Proteínas Recombinantes/metabolismo , Inhibidores de Serina Proteinasa/administración & dosificación , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Transfección , Vitronectina/deficiencia , Vitronectina/genética
8.
J Vasc Surg Venous Lymphat Disord ; 2(4): 441-450.e1, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25419511

RESUMEN

BACKGROUND: Treatment with low-molecular-weight heparin (LMWH) favorably alters the vein wall response to deep venous thrombosis (DVT), although the mechanisms remain unclear. Previous studies have suggested that LMWH alters the levels of circulating plasminogen activator inhibitor 1 (PAI-1), a known mediator of fibrosis, and may improve endogenous fibrinolysis. We hypothesized that LMWH favorably alters the vein wall response by binding of PAI-1 and acceleration of fibrinolysis. METHODS: Wild-type and PAI-1 -/- mice underwent treatment with LMWH after induction of occlusive DVT. Vein wall and plasma were harvested and analyzed by enzyme-linked immunosorbent assay, zymography, real-time polymerase chain reaction, and immunohistochemistry. RESULTS: Wild-type mice treated with LMWH exhibited diminished vein wall fibrosis (0.6 ± 0.6 vs 1.4 ± 0.2; P < .01; n = 5) and elevation of circulating PAI-1 (1776 ± 342 vs 567 ± 104 ρg/mL; P < .01; n = 5) compared with untreated controls after occlusive DVT. PAI-1-/- mice treated with LMWH were not similarly protected from fibrosis, despite improved thrombus resolution. Treatment with LMWH was associated with decreased intrathrombus interleukin-lß (68.6 ± 31.0 vs 223.4 ± 28.9 ρg/mg total protein; P < .01; n = 5) but did not alter inflammatory cell recruitment to the vein wall. PAI-1 -/- mice exhibited significantly elevated intrathrombus (257.2 ± 51.5 vs 4.3 ± 3.8 ρg/mg total protein; n = 5) and vein wall interleukin-13 (187.2 ± 57.6 vs 9.9 ± 1.1 ρg/mg total protein; P < .05; n = 5) as well as vein wall F4/80 positively staining monocytes (53 ± 11 vs 16 ± 2 cells/5 high-power fields; P < .05; n = 4). CONCLUSIONS: LMWH did not accelerate venous thrombosis resolution but did protect against vein wall fibrosis in a PAI-1-dependent manner in an occlusive DVT model. Lack of PAI-1 correlated with accelerated venous thrombosis resolution but no protection from fibrosis. PAI-1 inhibition as a treatment strategy for DVT is likely to accelerate clearance of the thrombus but may come at the expense of increased vein wall fibrosis. CLINICAL RELEVANCE: The pathophysiologic mechanism of post-thrombotic syndrome is not well understood clinically or experimentally. In this study, we evaluated the effect of the prominent fibrinolytic mechanism, plasminogen activator inhibitor 1 (PAI-1), and low-molecular-weight heparin (LMWH) on vein wall injury after thrombosis. We show here that LMWH is protective from vein wall fibrosis, but this is abrogated in PAI-1-deleted mice. This is also correlated with monocyte vein wall influx. These data support the clinical observation that LMWH may be protective from post-thrombotic vein wall injury in a PAI-1-dependent manner.

9.
Proc Natl Acad Sci U S A ; 110(51): E4941-9, 2013 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-24297881

RESUMEN

Plasminogen activator inhibitor type-1 (PAI-1) is a member of the serine protease inhibitor (serpin) family. Excessive PAI-1 activity is associated with human disease, making it an attractive pharmaceutical target. However, like other serpins, PAI-1 has a labile structure, making it a difficult target for the development of small molecule inhibitors, and to date, there are no US Food and Drug Administration-approved small molecule inactivators of any serpins. Here we describe the mechanistic and structural characterization of a high affinity inactivator of PAI-1. This molecule binds to PAI-1 reversibly and acts through an allosteric mechanism that inhibits PAI-1 binding to proteases and to its cofactor vitronectin. The binding site is identified by X-ray crystallography and mutagenesis as a pocket at the interface of ß-sheets B and C and α-helix H. A similar pocket is present on other serpins, suggesting that this site could be a common target in this structurally conserved protein family.


Asunto(s)
Inhibidor 1 de Activador Plasminogénico/química , Regulación Alostérica , Cristalografía por Rayos X , Humanos , Inhibidor 1 de Activador Plasminogénico/genética , Inhibidor 1 de Activador Plasminogénico/metabolismo , Unión Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Relación Estructura-Actividad , Vitronectina/química , Vitronectina/genética , Vitronectina/metabolismo
10.
PLoS One ; 7(2): e32204, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22363817

RESUMEN

The microenvironment of a tumor can influence both the morphology and the behavior of cancer cells which, in turn, can rapidly adapt to environmental changes. Increasing evidence points to the involvement of amoeboid cell migration and thus of cell blebbing in the metastatic process; however, the cues that promote amoeboid cell behavior in physiological and pathological conditions have not yet been clearly identified. Plasminogen Activator Inhibitor type-1 (PAI-1) is found in high amount in the microenvironment of aggressive tumors and is considered as an independent marker of bad prognosis. Here we show by immunoblotting, activity assay and immunofluorescence that, in SW620 human colorectal cancer cells, matrix-associated PAI-1 plays a role in the cell behavior needed for amoeboid migration by maintaining cell blebbing, localizing PDK1 and ROCK1 at the cell membrane and maintaining the RhoA/ROCK1/MLC-P pathway activation. The results obtained by modeling PAI-1 deposition around tumors indicate that matrix-bound PAI-1 is heterogeneously distributed at the tumor periphery and that, at certain spots, the elevated concentrations of matrix-bound PAI-1 needed for cancer cells to undergo the mesenchymal-amoeboid transition can be observed. Matrix-bound PAI-1, as a matricellular protein, could thus represent one of the physiopathological requirements to support metastatic formation.


Asunto(s)
Extensiones de la Superficie Celular/efectos de los fármacos , Extensiones de la Superficie Celular/metabolismo , Matriz Extracelular/metabolismo , Inhibidor 1 de Activador Plasminogénico/metabolismo , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Simulación por Computador , Activación Enzimática/efectos de los fármacos , Matriz Extracelular/efectos de los fármacos , Humanos , Proteínas Inmovilizadas/farmacología , Mesodermo/efectos de los fármacos , Mesodermo/patología , Modelos Biológicos , Inhibidor 1 de Activador Plasminogénico/farmacología , Unión Proteica/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Activador de Plasminógeno de Tipo Uroquinasa/farmacología
11.
J Pathol ; 228(2): 170-80, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22262246

RESUMEN

Fibrotic disorders of the lung are associated with perturbations in the plasminogen activation system. Specifically, plasminogen activator inhibitor-1 (PAI-1) expression is increased relative to the plasminogen activators. A direct role for this imbalance in modulating the severity of lung scarring following injury has been substantiated in the bleomycin model of pulmonary fibrosis. However, it remains unclear whether derangements in the plasminogen activation system contribute more generally to the pathogenesis of lung fibrosis beyond bleomycin injury. To answer this question, we employed an alternative model of lung scarring, in which type II alveolar epithelial cells (AECs) are specifically injured by administering diphtheria toxin (DT) to mice genetically engineered to express the human DT receptor (DTR) off the surfactant protein C promoter. This targeted AEC injury results in the diffuse accumulation of interstitial collagen. In the present study, we found that this targeted type II cell insult also increases PAI-1 expression in the alveolar compartment. We identified AECs and lung macrophages to be sources of PAI-1 production. To determine whether this elevated PAI-1 concentration was directly related to the severity of fibrosis, DTR(+) mice were crossed into a PAI-1-deficient background (DTR(+) : PAI-1(-/-) ). DT administration to DTR(+) : PAI-1(-/-) animals caused significantly less fibrosis than was measured in DTR(+) mice with intact PAI-1 production. PAI-1 deficiency also abrogated the accumulation of CD11b(+) exudate macrophages that were found to express PAI-1 and type-1 collagen. These observations substantiate the critical function of PAI-1 in pulmonary fibrosis pathogenesis and provide new insight into a potential mechanism by which this pro-fibrotic molecule influences collagen accumulation. Copyright © 2012 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Lesión Pulmonar Aguda/metabolismo , Células Epiteliales Alveolares/metabolismo , Macrófagos/metabolismo , Inhibidor 1 de Activador Plasminogénico/metabolismo , Fibrosis Pulmonar/metabolismo , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/patología , Células Epiteliales Alveolares/efectos de los fármacos , Células Epiteliales Alveolares/patología , Animales , Líquido del Lavado Bronquioalveolar/química , Colágeno Tipo I/metabolismo , Toxina Diftérica/toxicidad , Modelos Animales de Enfermedad , Exudados y Transudados/citología , Exudados y Transudados/efectos de los fármacos , Exudados y Transudados/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Inhibidor 1 de Activador Plasminogénico/análisis , Venenos/toxicidad , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/patología , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo
12.
J Vasc Surg ; 55(3): 815-22, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22119245

RESUMEN

BACKGROUND: Hyperlipidemia increases the level of blood plasminogen activator inhibitor-1 (PAI-1) that is responsible for regulating fibrinolysis by inhibiting both urokinase-type plasminogen activator (u-PA) and tissue-type plasminogen activator (t-PA). While this fibrinolytic pathway is well known, the role of PAI-1 in venous thrombosis (VT) under hyperlipidemic conditions has not been fully established. We sought to determine the effects of PAI-1 in an in vivo hyperlipidemic model of VT. METHODS: C57BL/6 wild-type (WT) mice, apolipoprotein E gene-deleted mice (ApoE-/-) having hyperlipidemia, and PAI-1 gene-deleted (PAI-1-/-) mice were used in this study. Inferior vena cava (IVC) ligation below the level of the renal veins was performed to create a stasis VT. Endpoints included measuring acute thrombosis (day 2) and chronic thrombosis (days 6 and 14). At euthanasia, blood samples were collected for plasmin and PAI-1 activity. In addition, the IVC and its thrombus were evaluated for thrombus weight (TW), u-PA activity, and differential leukocyte count while the vein wall only was analyzed for monocyte chemoattractant protein-1 (MCP-1), matrix metalloproteinase (MMP) 2, and MMP-9. RESULTS: Compared to WT at day 2, ApoE-/-mice demonstrated a statistically significant 14% increase in TW (P < .05) and a significant 41% increase in circulating PAI-1 activity (P < .05), while showing a trend of decreased plasmin activity. In addition, TW in ApoE-/-mice was 45% higher than PAI-1-/-mice at day 2 (P < .05), 33% at day 6 (P < .01), and 41% at day 14 (P < .01). ApoE-/-mice exhibited undetectable levels of u-PA in both vein wall and thrombus, compared to WT, at all time points. Also, vein wall MMP-2 was significantly decreased by 64% at day 6 (P < .01) and 58% at day 14 (P < .05). MMP-9 was significantly decreased by 71% at day 2 (P < .01) and 48% at day 6 (P < .01), in ApoE-/-mice compared to WT mice. In addition, in ApoE-/-mice, MCP-1 was significantly decreased by 38% at day 2 (P < .01) and 67% at day 6 (P < .01) vs WT mice. As expected in ApoE mice, following a decrease in MCP-1, monocyte recruitment was significantly decreased at days 6 (P < .01) and 14 (P < .05). CONCLUSIONS: A significant increase of circulating PAI-1 levels in hyperlipidemic mice correlated with an early increase in TW due to impaired fibrinolysis. The undetectable levels of u-PA in ApoE-/-mice correlated to a decrease in vein wall MMP-2, MMP-9, MCP-1, and a decrease in monocyte recruitment diminishing thrombus resolution.


Asunto(s)
Apolipoproteínas E/deficiencia , Fibrinólisis , Hiperlipidemias/complicaciones , Vena Cava Inferior/metabolismo , Trombosis de la Vena/etiología , Animales , Apolipoproteínas E/genética , Quimiocina CCL2/metabolismo , Modelos Animales de Enfermedad , Fibrinolisina/metabolismo , Fibrinólisis/genética , Hiperlipidemias/sangre , Hiperlipidemias/genética , Recuento de Leucocitos , Ligadura , Masculino , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Inhibidor 1 de Activador Plasminogénico/deficiencia , Inhibidor 1 de Activador Plasminogénico/genética , Factores de Tiempo , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Vena Cava Inferior/cirugía , Trombosis de la Vena/sangre
13.
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
14.
J Biol Chem ; 285(11): 7892-902, 2010 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-20061381

RESUMEN

Plasminogen activator inhibitor type 1, (PAI-1) the primary inhibitor of the tissue-type (tPA) and urokinase-type (uPA) plasminogen activators, has been implicated in a wide range of pathological processes, making it an attractive target for pharmacologic inhibition. Currently available small-molecule inhibitors of PAI-1 bind with relatively low affinity and do not inactivate PAI-1 in the presence of its cofactor, vitronectin. To search for novel PAI-1 inhibitors with improved potencies and new mechanisms of action, we screened a library selected to provide a range of biological activities and structural diversity. Five potential PAI-1 inhibitors were identified, and all were polyphenolic compounds including two related, naturally occurring plant polyphenols that were structurally similar to compounds previously shown to provide cardiovascular benefit in vivo. Unique second generation compounds were synthesized and characterized, and several showed IC(50) values for PAI-1 between 10 and 200 nm. This represents an enhanced potency of 10-1000-fold over previously reported PAI-1 inactivators. Inhibition of PAI-1 by these compounds was reversible, and their primary mechanism of action was to block the initial association of PAI-1 with a protease. Consistent with this mechanism and in contrast to previously described PAI-1 inactivators, these compounds inactivate PAI-1 in the presence of vitronectin. Two of the compounds showed efficacy in ex vivo plasma and one blocked PAI-1 activity in vivo in mice. These data describe a novel family of high affinity PAI-1-inactivating compounds with improved characteristics and in vivo efficacy, and suggest that the known cardiovascular benefits of dietary polyphenols may derive in part from their inactivation of PAI-1.


Asunto(s)
Diseño de Fármacos , Flavonoides/farmacología , Fenoles/farmacología , Inhibidor 1 de Activador Plasminogénico/metabolismo , Inhibidores de Proteasas/farmacología , Serpinas/metabolismo , Animales , Electroforesis en Gel de Poliacrilamida , Flavonoides/síntesis química , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fenoles/síntesis química , Polifenoles , Inhibidores de Proteasas/síntesis química , Proteínas Recombinantes/metabolismo , Serpina E2 , Resonancia por Plasmón de Superficie , Vitronectina/metabolismo
15.
J Clin Invest ; 119(5): 1136-49, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19381014

RESUMEN

Leukocyte and platelet accumulation at sites of cerebral ischemia exacerbate cerebral damage. The ectoenzyme CD39 on the plasmalemma of endothelial cells metabolizes ADP to suppress platelet accumulation in the ischemic brain. However, the role of leukocyte surface CD39 in regulating monocyte and neutrophil trafficking in this setting is not known. Here we have demonstrated in mice what we believe to be a novel mechanism by which CD39 on monocytes and neutrophils regulates their own sequestration into ischemic cerebral tissue, by catabolizing nucleotides released by injured cells, thereby inhibiting their chemotaxis, adhesion, and transmigration. Bone marrow reconstitution and provision of an apyrase, an enzyme that hydrolyzes nucleoside tri- and diphosphates, each normalized ischemic leukosequestration and cerebral infarction in CD39-deficient mice. Leukocytes purified from Cd39-/- mice had a markedly diminished capacity to phosphohydrolyze adenine nucleotides and regulate platelet reactivity, suggesting that leukocyte ectoapyrases modulate the ambient vascular nucleotide milieu. Dissipation of ATP by CD39 reduced P2X7 receptor stimulation and thereby suppressed baseline leukocyte alphaMbeta2-integrin expression. As alphaMbeta2-integrin blockade reversed the postischemic, inflammatory phenotype of Cd39-/- mice, these data suggest that phosphohydrolytic activity on the leukocyte surface suppresses cell-cell interactions that would otherwise promote thrombosis or inflammation. These studies indicate that CD39 on both endothelial cells and leukocytes reduces inflammatory cell trafficking and platelet reactivity, with a consequent reduction in tissue injury following cerebral ischemic challenge.


Asunto(s)
Antígenos CD/fisiología , Apirasa/fisiología , Movimiento Celular/fisiología , Leucocitos/citología , 5'-Nucleotidasa/antagonistas & inhibidores , Adenosina Difosfato/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Apirasa/farmacología , Trasplante de Médula Ósea , Isquemia Encefálica/genética , Isquemia Encefálica/patología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Células Endoteliales/metabolismo , Inhibidores Enzimáticos/farmacología , Leucocitos/efectos de los fármacos , Leucocitos/metabolismo , Antígeno de Macrófago-1/inmunología , Antígeno de Macrófago-1/metabolismo , Macrófagos/citología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Ratones Endogámicos , Ratones Noqueados , Monocitos/citología , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Neutrófilos/citología , Neutrófilos/metabolismo , Activación Plaquetaria/fisiología , Antagonistas del Receptor Purinérgico P2 , Interferencia de ARN , Receptores Purinérgicos P2/genética , Quimera por Trasplante/fisiología
16.
Oligonucleotides ; 19(2): 117-28, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19284310

RESUMEN

The serine protease inhibitor plasminogen activator inhibitor-1 (PAI-1) is increased in several cancers, including breast, where it is associated with a poor outcome. Metastatic breast cancer has a dismal prognosis, as evidenced by treatment goals that are no longer curative but are largely palliative in nature. PAI-1 competes with integrins and the urokinase plasminogen activator receptor on the surface of breast cancer cells for binding to vitronectin. This results in the detachment of tumor cells from the extracellular matrix, which is critical to the metastatic process. For this reason, we sought to isolate RNA aptamers that disrupt the interaction between PAI-1 and vitronectin. Through utilization of combinatorial chemistry techniques, aptamers have been selected that bind to PAI-1 with high affinity and specificity. We identified two aptamers, WT-15 and SM-20, that disrupt the interactions between PAI-1 and heparin, as well as PAI-1 and vitronectin, without affecting the antiprotease activity of PAI-1. Furthermore, SM-20 prevented the detachment of breast cancer cells (MDA-MB-231) from vitronectin in the presence of PAI-1, resulting in an increase in cellular adhesion. Therefore, the PAI-1 aptamer SM-20 demonstrates therapeutic potential as an antimetastatic agent and could possibly be used as an adjuvant to traditional chemotherapy for breast cancer.


Asunto(s)
Aptámeros de Nucleótidos/farmacología , Neoplasias de la Mama/terapia , Inhibidor 1 de Activador Plasminogénico/metabolismo , ARN/farmacología , Vitronectina/metabolismo , Aptámeros de Nucleótidos/química , Neoplasias de la Mama/patología , Adhesión Celular/efectos de los fármacos , Heparina/metabolismo , Humanos , Metástasis de la Neoplasia , ARN/química , Técnica SELEX de Producción de Aptámeros , Trombina/metabolismo
17.
Breast Cancer Res ; 10(6): R100, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-19055748

RESUMEN

INTRODUCTION: Snail, a family of transcriptional repressors implicated in cell movement, has been correlated with tumour invasion. The Plasminogen Activation (PA) system, including urokinase plasminogen activator (uPA), its receptor and its inhibitor, plasminogen activator inhibitor type 1(PAI-1), also plays a key role in cancer invasion and metastasis, either through proteolytic degradation or by non-proteolytic modulation of cell adhesion and migration. Thus, Snail and the PA system are both over-expressed in cancer and influence this process. In this study we aimed to determine if the activity of SNAI1 (a member of the Snail family) is correlated with expression of the PA system components and how this correlation can influence tumoural cell migration. METHODS: We compared the invasive breast cancer cell-line MDA-MB-231 expressing SNAI1 (MDA-mock) with its derived clone expressing a dominant-negative form of SNAI1 (SNAI1-DN). Expression of PA system mRNAs was analysed by cDNA microarrays and real-time quantitative RT-PCR. Wound healing assays were used to determine cell migration. PAI-1 distribution was assessed by immunostaining. RESULTS: We demonstrated by both cDNA microarrays and real-time quantitative RT-PCR that the functional blockade of SNAI1 induces a significant decrease of PAI-1 and uPA transcripts. After performing an in vitro wound-healing assay, we observed that SNAI1-DN cells migrate more slowly than MDA-mock cells and in a more collective manner. The blockade of SNAI1 activity resulted in the redistribution of PAI-1 in SNAI1-DN cells decorating large lamellipodia, which are commonly found structures in these cells. CONCLUSIONS: In the absence of functional SNAI1, the expression of PAI-1 transcripts is decreased, although the protein is redistributed at the leading edge of migrating cells in a manner comparable with that seen in normal epithelial cells.


Asunto(s)
Neoplasias de la Mama/metabolismo , Movimiento Celular/fisiología , Inhibidor 1 de Activador Plasminogénico/metabolismo , Factores de Transcripción/metabolismo , Neoplasias de la Mama/genética , Cadherinas/metabolismo , Femenino , Técnica del Anticuerpo Fluorescente , Perfilación de la Expresión Génica , Genes Dominantes , Humanos , Técnicas para Inmunoenzimas , Análisis de Secuencia por Matrices de Oligonucleótidos , Inhibidor 1 de Activador Plasminogénico/genética , Seudópodos/fisiología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción de la Familia Snail , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/genética , Células Tumorales Cultivadas , Activador de Plasminógeno de Tipo Uroquinasa/genética , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Cicatrización de Heridas
18.
J Biol Chem ; 283(26): 18147-57, 2008 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-18436534

RESUMEN

Plasminogen activator inhibitor type 1 (PAI-1) is a serine protease inhibitor (serpin) in which the reactive center loop (RCL) spontaneously inserts into a central beta-sheet, beta-sheet A, resulting in inactive inhibitor. Available x-ray crystallographic studies of PAI-1 in an active conformation relied on the use of stabilizing mutations. Recently it has become evident that these structural models do not adequately explain the behavior of wild-type PAI-1 (wtPAI-1) in solution. To probe the structure of native wtPAI-1, we used three conformationally sensitive ligands: the physiologic cofactor, vitronectin; a monoclonal antibody, 33B8, that binds preferentially to RCL-inserted forms of PAI-1; and RCL-mimicking peptides that insert into beta-sheet A. From patterns of interaction with wtPAI-1 and the stable mutant, 14-1B, we propose a model of the native conformation of wtPAI-1 in which the bottom of the central sheet is closed, whereas the top of the beta-sheet A is open to allow partial insertion of the RCL. Because the incorporation of RCL-mimicking peptides into wtPAI-1 is accelerated by vitronectin, we further propose that vitronectin alters the conformation of the RCL to allow increased accessibility to beta-sheet A, yielding a structural hypothesis that is contradictory to the current structural model of PAI-1 in solution and its interaction with vitronectin.


Asunto(s)
Inhibidor 1 de Activador Plasminogénico/química , Anticuerpos Monoclonales/química , Humanos , Cinética , Ligandos , Modelos Biológicos , Conformación Molecular , Mutación , Péptidos/química , Inhibidor 1 de Activador Plasminogénico/metabolismo , Conformación Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Resonancia por Plasmón de Superficie , Factores de Tiempo , Vitronectina/química
19.
Circulation ; 117(6): 798-805, 2008 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-18212290

RESUMEN

BACKGROUND: Fat inflammation may play an important role in comorbidities associated with obesity such as atherosclerosis. METHODS AND RESULTS: To first establish feasibility of fat transplantation, epididymal fat pads were harvested from wild-type C57BL/6J mice and transplanted into leptin-deficient (Lep(ob/ob)) mice. Fat transplantation produced physiological leptin levels and prevented obesity and infertility in Lep(ob/ob) mice. However, the transplanted fat depots were associated with chronically increased macrophage infiltration with characteristics identical to those observed in fat harvested from obese animals. The inflammation in transplanted adipose depots was regulated by the same factors that have been implicated in endogenous fat inflammation such as monocyte chemoattractant protein-1. To determine whether this inflamed adipose depot could affect vascular disease in mice, epididymal fat depots were transplanted into atherosclerosis-prone apolipoprotein E-deficient ApoE(-/-) mice. Plasma from ApoE(-/-) mice receiving fat transplants contained increased leptin, resistin, and monocyte chemoattractant protein-1 compared with plasma from sham-operated ApoE(-/-) mice. Furthermore, mice transplanted with visceral fat developed significantly more atherosclerosis compared with sham-operated animals, whereas transplants with subcutaneous fat did not affect atherosclerosis despite a similar degree of fat inflammation. Treatment of transplanted ApoE(-/-) mice with pioglitazone decreased macrophage content of the transplanted visceral fat pad and reduced plasma monocyte chemoattractant protein-1. Importantly, pioglitazone also reduced atherosclerosis triggered by inflammatory visceral fat but had no protective effect on atherosclerosis in the absence of the visceral fat transplantation. CONCLUSIONS: Our results indicate that visceral adipose-related inflammation accelerates atherosclerosis in mice. Drugs such as thiazolidinediones might be a useful strategy to specifically attenuate the vascular disease induced by visceral inflammatory fat.


Asunto(s)
Apolipoproteínas E/deficiencia , Aterosclerosis/etiología , Inflamación/fisiopatología , Grasa Intraabdominal/fisiología , Tiazolidinedionas/uso terapéutico , Adiponectina/sangre , Animales , Aterosclerosis/tratamiento farmacológico , Inflamación/complicaciones , Inflamación/patología , Grasa Intraabdominal/patología , Grasa Intraabdominal/trasplante , Leptina/deficiencia , Macrófagos/patología , Ratones , Ratones Endogámicos C57BL , Pioglitazona , Tiazolidinedionas/farmacología
20.
FASEB J ; 21(3): 935-49, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17197388

RESUMEN

Hypoxia, as occurs during tissue ischemia, tips the natural anticoagulant/procoagulant balance of the endovascular wall to favor activation of coagulation. Plasminogen activator inhibitor-1 (PAI-1) is an important factor suppressing fibrinolysis under conditions of low oxygen tension. We previously reported that hypoxia induced PAI-1 mRNA and antigen expression in murine macrophages secondary to increased de novo transcription as well as increased mRNA stability. We now show in RAW264.7 murine macrophages that the transcription factors early growth response gene-1 (Egr-1), hypoxia-inducible factor-1alpha (HIF-1alpha), and CCAAT/enhancer binding protein alpha (C/EBPalpha) are quickly activated in hypoxia and are responsible for transcription and expression of PAI-1. Murine PAI-1 promoter constructs, including Egr, HIF-1alpha, and/or C/EBPalpha binding sites, were transfected into RAW 264.7 murine macrophages. To identify the relative importance of each of these putative hypoxia-responsive elements, cells were exposed to normobaric hypoxia, and transcriptional activity was recorded. At 16 h of hypoxic exposure, murine PAI-1 promoter deletion constructs that included Egr, HIF-1alpha, and/or C/EBPalpha binding sites demonstrated increased transcriptional activity. Mutation of each of these three murine PAI-1 promoter sites (or a combination of them) resulted in a marked reduction in hypoxia sensitivity as detected by transcriptional analysis. Functional data obtained using 32P-labeled Egr, HIF-1alpha response element (HRE), and C/EBPalpha oligonucleotides revealed induction of DNA binding activity in nuclear extracts from hypoxic RAW cells, with supershift analysis confirming activation of Egr-1, HIF-1alpha, or C/EBPalpha. ChIP analysis confirmed the authenticity of these interactions as each of these transcription factors binds to chromatin under hypoxic conditions. Further, the induction of PAI-1 by Egr-1, HIF-1alpha, or C/EBPalpha was replicated in primary peritoneal macrophages. These data suggest that although HIF-1alpha appears to dominate the PAI-1 transcriptional response in hypoxia, Egr-1 and C/EBPalpha greatly augment this response and can do so independent of HIF-1alpha or each other. These studies are relevant to ischemic up-regulation of the PAI-1 gene and consequent accrual of microvascular thrombus under ischemic conditions.


Asunto(s)
Proteína alfa Potenciadora de Unión a CCAAT/fisiología , Hipoxia de la Célula/fisiología , Proteína 1 de la Respuesta de Crecimiento Precoz/fisiología , Subunidad alfa del Factor 1 Inducible por Hipoxia/fisiología , Oxígeno/metabolismo , Inhibidor 1 de Activador Plasminogénico/genética , Regiones Promotoras Genéticas/fisiología , Animales , Células Cultivadas , Regulación de la Expresión Génica , Humanos , Macrófagos/metabolismo , Ratones , Inhibidor 1 de Activador Plasminogénico/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA