Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Blood ; 119(3): 874-83, 2012 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-22117049

RESUMEN

The cytoprotective effects of activated protein C (aPC) are well established. In contrast, the receptors and signaling mechanism through which aPC conveys cytoprotection in various cell types remain incompletely defined. Thus, within the renal glomeruli, aPC preserves endothelial cells via a protease-activated receptor-1 (PAR-1) and endothelial protein C receptor-dependent mechanism. Conversely, the signaling mechanism through which aPC protects podocytes remains unknown. While exploring the latter, we identified a novel aPC/PAR-dependent cytoprotective signaling mechanism. In podocytes, aPC inhibits apoptosis through proteolytic activation of PAR-3 independent of endothelial protein C receptor. PAR-3 is not signaling competent itself as it requires aPC-induced heterodimerization with PAR-2 (human podocytes) or PAR-1 (mouse podocytes). This cytoprotective signaling mechanism depends on caveolin-1 dephosphorylation. In vivo aPC protects against lipopolysaccharide-induced podocyte injury and proteinuria. Genetic deletion of PAR-3 impairs the nephroprotective effect of aPC, demonstrating the crucial role of PAR-3 for aPC-dependent podocyte protection. This novel, aPC-mediated interaction of PARs demonstrates the plasticity and cell-specificity of cytoprotective aPC signaling. The evidence of specific, dynamic signaling complexes underlying aPC-mediated cytoprotection may allow the design of cell type specific targeted therapies.


Asunto(s)
Apoptosis , Citoprotección , Podocitos/metabolismo , Proteína C/metabolismo , Receptor PAR-1/metabolismo , Receptores de Trombina/metabolismo , Animales , Anticoagulantes/metabolismo , Comunicación Celular , Células Cultivadas , Humanos , Glomérulos Renales/citología , Glomérulos Renales/metabolismo , Lipopolisacáridos/farmacología , Microdominios de Membrana , Ratones , Podocitos/efectos de los fármacos , Podocitos/patología , Multimerización de Proteína , Transducción de Señal , Trombina
2.
Nat Med ; 13(11): 1349-58, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17982464

RESUMEN

Data providing direct evidence for a causative link between endothelial dysfunction, microvascular disease and diabetic end-organ damage are scarce. Here we show that activated protein C (APC) formation, which is regulated by endothelial thrombomodulin, is reduced in diabetic mice and causally linked to nephropathy. Thrombomodulin-dependent APC formation mediates cytoprotection in diabetic nephropathy by inhibiting glomerular apoptosis. APC prevents glucose-induced apoptosis in endothelial cells and podocytes, the cellular components of the glomerular filtration barrier. APC modulates the mitochondrial apoptosis pathway via the protease-activated receptor PAR-1 and the endothelial protein C receptor EPCR in glucose-stressed cells. These experiments establish a new pathway, in which hyperglycemia impairs endothelial thrombomodulin-dependent APC formation. Loss of thrombomodulin-dependent APC formation interrupts cross-talk between the vascular compartment and podocytes, causing glomerular apoptosis and diabetic nephropathy. Conversely, maintaining high APC levels during long-term diabetes protects against diabetic nephropathy.


Asunto(s)
Apoptosis , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Experimental/prevención & control , Nefropatías Diabéticas/patología , Nefropatías Diabéticas/prevención & control , Endotelio Vascular/patología , Podocitos/patología , Proteína C/fisiología , Sustitución de Aminoácidos/genética , Animales , Apoptosis/genética , Línea Celular Transformada , Células Cultivadas , Citoprotección/genética , Diabetes Mellitus Experimental/enzimología , Diabetes Mellitus Experimental/genética , Nefropatías Diabéticas/enzimología , Nefropatías Diabéticas/genética , Endotelio Vascular/enzimología , Activación Enzimática/genética , Humanos , Glomérulos Renales/irrigación sanguínea , Glomérulos Renales/enzimología , Glomérulos Renales/patología , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Ratones Transgénicos , Microcirculación/enzimología , Microcirculación/patología , Podocitos/enzimología , Proteína C/biosíntesis , Proteína C/genética , Transducción de Señal/genética , Trombomodulina/fisiología
3.
Nature ; 451(7182): 1076-81, 2008 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-18278032

RESUMEN

Understanding the neuropathology of multiple sclerosis (MS) is essential for improved therapies. Therefore, identification of targets specific to pathological types of MS may have therapeutic benefits. Here we identify, by laser-capture microdissection and proteomics, proteins unique to three major types of MS lesions: acute plaque, chronic active plaque and chronic plaque. Comparative proteomic profiles identified tissue factor and protein C inhibitor within chronic active plaque samples, suggesting dysregulation of molecules associated with coagulation. In vivo administration of hirudin or recombinant activated protein C reduced disease severity in experimental autoimmune encephalomyelitis and suppressed Th1 and Th17 cytokines in astrocytes and immune cells. Administration of mutant forms of recombinant activated protein C showed that both its anticoagulant and its signalling functions were essential for optimal amelioration of experimental autoimmune encephalomyelitis. A proteomic approach illuminated potential therapeutic targets selective for specific pathological stages of MS and implicated participation of the coagulation cascade.


Asunto(s)
Perfilación de la Expresión Génica , Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/patología , Proteómica , Adulto , Animales , Coagulación Sanguínea , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Femenino , Humanos , Inflamación/metabolismo , Inflamación/patología , Masculino , Ratones , Persona de Mediana Edad , Esclerosis Múltiple/clasificación , Esclerosis Múltiple/tratamiento farmacológico , Proteína C/genética , Proteína C/metabolismo , Proteína C/farmacología , Células TH1/inmunología , Células Th2/inmunología , Trombina/antagonistas & inhibidores , Trombina/metabolismo
4.
J Infect Dis ; 202(10): 1600-7, 2010 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-20925531

RESUMEN

BACKGROUND: Recombinant human activated protein C (APC) improves survival of patients with severe sepsis; this beneficial effect is especially apparent in patients with pneumococcal pneumonia. The aim of this study was to determine the effect of APC treatment initiated after induction of pneumococcal pneumonia on pulmonary coagulation, inflammation, and survival, with or without concurrent antibiotic therapy. METHODS: Mice were infected intranasally with viable Streptococcus pneumoniae and were treated intraperitoneally after 24 h of infection with vehicle, recombinant mouse (rm) APC (125 µg), ceftriaxone (500 µg), or rm-APC plus ceftriaxone. Treatment with rm-APC or vehicle was repeated every 8 h for a maximum of 96 h. Animals were either killed 48 h after infection or were monitored in a survival study (with an extra dose of ceftriaxone given after 72 h). RESULTS: Rm-APC treatment inhibited pulmonary activation of coagulation, as reflected by lower levels of thrombin-antithrombin complexes and D-dimer. Rm-APC did not affect the pulmonary levels of 55 inflammatory mediators in the context of antibiotic therapy. Rm-APC added to ceftriaxone markedly improved survival, compared with ceftriaxone treatment alone. CONCLUSIONS: Rm-APC inhibits pulmonary activation of coagulation and, when added to antibiotic therapy, improves survival in murine pneumococcal pneumonia.


Asunto(s)
Trastornos de las Proteínas de Coagulación/tratamiento farmacológico , Trastornos de las Proteínas de Coagulación/microbiología , Neumonía Neumocócica/tratamiento farmacológico , Proteína C/uso terapéutico , Animales , Antibacterianos/uso terapéutico , Factores de Coagulación Sanguínea/antagonistas & inhibidores , Ceftriaxona/administración & dosificación , Ceftriaxona/uso terapéutico , Evaluación Preclínica de Medicamentos , Quimioterapia Combinada , Humanos , Inyecciones Intraperitoneales , Masculino , Ratones , Ratones Endogámicos C57BL , Neumonía Neumocócica/complicaciones , Proteína C/administración & dosificación , Proteína C/farmacología , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/uso terapéutico
5.
J Clin Invest ; 117(7): 1951-60, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17557119

RESUMEN

Endothelial protein C receptor (EPCR) and thrombomodulin (TM) are expressed at high levels in the resting microvasculature and convert protein C (PC) into its activated form, which is a potent anticoagulant and antiinflammatory molecule. Here we provide evidence that in Crohn disease (CD) and ulcerative colitis (UC), the 2 major forms of inflammatory bowel disease (IBD), there was loss of expression of endothelial EPCR and TM, which in turns caused impairment of PC activation by the inflamed mucosal microvasculature. In isolated human intestinal endothelial cells, administration of recombinant activated PC had a potent antiinflammatory effect, as demonstrated by downregulated cytokine-dependent cell adhesion molecule expression and chemokine production as well as inhibited leukocyte adhesion. In vivo, administration of activated PC was therapeutically effective in ameliorating experimental colitis as evidenced by reduced weight loss, disease activity index, and histological colitis scores as well as inhibited leukocyte adhesion to the inflamed intestinal vessels. The results suggest that the PC pathway represents a new system crucially involved in governing intestinal homeostasis mediated by the mucosal microvasculature. Restoring the PC pathway may represent a new therapeutic approach to suppress intestinal inflammation in IBD.


Asunto(s)
Enfermedades Inflamatorias del Intestino/metabolismo , Enfermedades Inflamatorias del Intestino/patología , Microcirculación/patología , Proteína C/metabolismo , Transducción de Señal , Animales , Antígenos CD/metabolismo , Adhesión Celular/efectos de los fármacos , Células Cultivadas , Quimiocinas/biosíntesis , Regulación hacia Abajo , Receptor de Proteína C Endotelial , Endotelio/patología , Humanos , Inmunohistoquímica , Inflamación/metabolismo , Inflamación/patología , Molécula 1 de Adhesión Intercelular/metabolismo , Leucocitos/citología , Ratones , Microcirculación/metabolismo , Proteína C/farmacología , Receptores de Superficie Celular/metabolismo , Transducción de Señal/efectos de los fármacos , Trombomodulina/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Molécula 1 de Adhesión Celular Vascular/metabolismo
6.
Crit Care ; 14(2): R65, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20398279

RESUMEN

INTRODUCTION: Influenza accounts for 5 to 10% of community-acquired pneumonias and is a major cause of mortality. Sterile and bacterial lung injuries are associated with procoagulant and inflammatory derangements in the lungs. Activated protein C (APC) is an anticoagulant with anti-inflammatory properties that exert beneficial effects in models of lung injury. We determined the impact of lethal influenza A (H1N1) infection on systemic and pulmonary coagulation and inflammation, and the effect of recombinant mouse (rm-) APC here on. METHODS: Male C57BL/6 mice were intranasally infected with a lethal dose of a mouse adapted influenza A (H1N1) strain. Treatment with rm-APC (125 microg intraperitoneally every eight hours for a maximum of three days) or vehicle was initiated 24 hours after infection. Mice were euthanized 48 or 96 hours after infection, or observed for up to nine days. RESULTS: Lethal H1N1 influenza resulted in systemic and pulmonary activation of coagulation, as reflected by elevated plasma and lung levels of thrombin-antithrombin complexes and fibrin degradation products. These procoagulant changes were accompanied by inhibition of the fibrinolytic response due to enhanced release of plasminogen activator inhibitor type-1. Rm-APC strongly inhibited coagulation activation in both plasma and lungs, and partially reversed the inhibition of fibrinolysis. Rm-APC temporarily reduced pulmonary viral loads, but did not impact on lung inflammation or survival. CONCLUSIONS: Lethal influenza induces procoagulant and antifibrinolytic changes in the lung which can be partially prevented by rm-APC treatment.


Asunto(s)
Anticoagulantes/antagonistas & inhibidores , Anticoagulantes/uso terapéutico , Coagulación Sanguínea/efectos de los fármacos , Subtipo H1N1 del Virus de la Influenza A/patogenicidad , Infecciones por Orthomyxoviridae/sangre , Proteína C/antagonistas & inhibidores , Proteína C/uso terapéutico , Animales , Anticoagulantes/metabolismo , Regulación hacia Abajo/genética , Subtipo H1N1 del Virus de la Influenza A/genética , Pulmón/efectos de los fármacos , Pulmón/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Infecciones por Orthomyxoviridae/fisiopatología , Proteína C/genética , Proteína C/metabolismo , Proteínas Recombinantes , Carga Viral
7.
J Am Soc Nephrol ; 20(2): 267-77, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19092124

RESUMEN

Administration of activated protein C (APC) protects from renal dysfunction, but the underlying mechanism is unknown. APC exerts both antithrombotic and cytoprotective properties, the latter via modulation of protease-activated receptor-1 (PAR-1) signaling. We generated APC variants to study the relative importance of the two functions of APC in a model of LPS-induced renal microvascular dysfunction. Compared with wild-type APC, the K193E variant exhibited impaired anticoagulant activity but retained the ability to mediate PAR-1-dependent signaling. In contrast, the L8W variant retained anticoagulant activity but lost its ability to modulate PAR-1. By administering wild-type APC or these mutants in a rat model of LPS-induced injury, we found that the PAR-1 agonism, but not the anticoagulant function of APC, reversed LPS-induced systemic hypotension. In contrast, both functions of APC played a role in reversing LPS-induced decreases in renal blood flow and volume, although the effects on PAR-1-dependent signaling were more potent. Regarding potential mechanisms for these findings, APC-mediated PAR-1 agonism suppressed LPS-induced increases in the vasoactive peptide adrenomedullin and infiltration of iNOS-positive leukocytes into renal tissue. However, the anticoagulant function of APC was responsible for suppressing LPS-induced stimulation of the proinflammatory mediators ACE-1, IL-6, and IL-18, perhaps accounting for its ability to modulate renal hemodynamics. Both variants reduced active caspase-3 and abrogated LPS-induced renal dysfunction and pathology. We conclude that although PAR-1 agonism is solely responsible for APC-mediated improvement in systemic hemodynamics, both functions of APC play distinct roles in attenuating the response to injury in the kidney.


Asunto(s)
Enfermedades Renales/metabolismo , Riñón/lesiones , Proteína C/fisiología , Animales , Humanos , Inflamación , Interleucina-18/metabolismo , Interleucina-6/metabolismo , Riñón/metabolismo , Lipopolisacáridos/metabolismo , Masculino , Microcirculación , Proteína C/metabolismo , Ratas , Ratas Sprague-Dawley , Receptor PAR-1/metabolismo , Transducción de Señal
8.
J Pharmacol Exp Ther ; 325(1): 17-26, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18182560

RESUMEN

The protein C (PC) pathway plays an important role in vascular and immune function, and acquired deficiency during sepsis is associated with increased mortality in both animal models and in clinical studies. However, the association of acquired PC deficiency with the pathophysiology of lung injury is unclear. We hypothesized that low PC induced by sepsis would associate with increased pulmonary injury and that replacement with activated protein C (APC) would reverse the activation of pathways associated with injury. Using a cecal ligation and puncture (CLP) model of polymicrobial sepsis, we examined the role of acquired PC deficiency on acute lung injury assessed by analyzing changes in pulmonary pathology, chemokine response, inducible nitric-oxide synthase (iNOS), and the angiotensin pathway. Acquired PC deficiency was strongly associated with an increase in lung inflammation and drivers of pulmonary injury, including angiotensin (Ang) II, thymus and activation-regulated chemokine, plasminogen activator inhibitor (PAI)-1, and iNOS. In contrast, the protective factor angiotensin-converting enzyme (ACE)-2 was significantly suppressed in animals with acquired PC deficiency. The endothelial protein C receptor, required for the cytoprotective signaling of APC, was significantly increased post-CLP, suggesting a compensatory up-regulation of the signaling receptor. Treatment of septic animals with APC reduced pulmonary pathology, suppressed the macrophage inflammatory protein family chemokine response, iNOS expression, and PAI-1 activity and up-regulated ACE-2 expression with concomitant reduction in AngII peptide. These data demonstrate a clear link between acquired PC deficiency and pulmonary inflammatory response in the rat sepsis model and provide support for the concept of APC as a replacement therapy in acute lung injury associated with acquired PC deficiency.


Asunto(s)
Inhibidores de la Enzima Convertidora de Angiotensina/uso terapéutico , Peptidil-Dipeptidasa A/efectos de los fármacos , Deficiencia de Proteína C/tratamiento farmacológico , Síndrome de Dificultad Respiratoria/tratamiento farmacológico , Sepsis/complicaciones , Síndrome de Respuesta Inflamatoria Sistémica/tratamiento farmacológico , Enzima Convertidora de Angiotensina 2 , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Animales , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas Inflamatorias de Macrófagos/genética , Óxido Nítrico Sintasa de Tipo II/genética , Inhibidor 1 de Activador Plasminogénico/genética , Deficiencia de Proteína C/etiología , Ratas
9.
Arterioscler Thromb Vasc Biol ; 27(12): 2634-41, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17932312

RESUMEN

UNLABELLED: Background- APC is an antithrombotic and antiinflammatory serine protease that plays an important role in vascular function. We report that APC can suppress the proapoptotic mediator TRAIL in human umbilical vein endothelial cells, and we have investigated the signaling mechanism. METHODS AND RESULTS: APC inhibited endothelial TRAIL expression and secretion and its induction by cell activation. To explore the mechanism, we examined factors associated with TRAIL regulation and demonstrated that APC increased the level of EGR-1, a transcriptional factor known to suppress the TRAIL promoter. APC also induced a significant increase in phosphorylation of ERK-1/2, required to activate EGR-1 expression. Activation of ERK-1/2 was dependent on the protease activated receptor-1 (PAR-1), but independent of the endothelial protein C receptor (EPCR). Using siRNA, we found that the effect of APC on the EGR-1/ERK signaling required for TRAIL inhibition was dependent on the S1P1 receptor and S1P1 kinase. CONCLUSIONS: Our data suggest that APC may provide cytoprotective activity by activating the ERK pathway, which upregulates EGR-1 thereby suppressing the expression of TRAIL. Moreover, we provide evidence that APC can induce a cell signaling response through a PAR-1/S1P1-dependent but EPCR-independent mechanism.


Asunto(s)
Antígenos CD/metabolismo , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Células Endoteliales/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteína C/metabolismo , Receptores de Superficie Celular/metabolismo , Transducción de Señal , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Antígenos CD/genética , Células Cultivadas , Citoprotección , Células Endoteliales/enzimología , Receptor de Proteína C Endotelial , Activación Enzimática , Humanos , Fosforilación , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Interferencia de ARN , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Receptor PAR-1/metabolismo , Receptores de Superficie Celular/genética , Receptores de Lisoesfingolípidos/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/genética , Factores de Tiempo , Transcripción Genética , Factor de Necrosis Tumoral alfa/metabolismo
10.
Adv Exp Med Biol ; 614: 83-91, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18290317

RESUMEN

The protein C (PC) pathway plays an important role in vascular function, and acquired deficiency during sepsis is associated with increased mortality. We have explored the role of PC suppression in modulating early inflammatory events in a model of polymicrobial sepsis. We show that increased levels of organ damage and dysfunction are associated with decreased levels of endogenous PC. Notably, animals with low PC had correspondingly high levels of pulmonary iNOS expression, which correlated with chemokines KC/Gro and MIP2, previously shown to predict outcome in this model. Treatment with activated protein C (aPC) not only reduced the pathology score, leukocyte infiltration and markers of organ dysfunction, but also suppressed the induction of iNOS, and the chemokine response (including KC/Gro, MIP2, IP-10, RANTES, GCP-2 and lymphotactin), and increased apoA1. aPC treatment also suppressed the induction of VEGF, a marker recently suggested to play a pathophysiological role in sepsis. These data demonstrate a clear link between low protein C and degree of organ damage and dysfunction in sepsis, as well as the early reversal with aPC treatment. Moreover, our data show a direct role of aPC in broadly modulating monocyte and T-cell chemokines following systemic inflammatory response.


Asunto(s)
Anticoagulantes/uso terapéutico , Quimiocinas/metabolismo , Proteína C/fisiología , Proteína C/uso terapéutico , Sepsis/tratamiento farmacológico , Animales , Biomarcadores/sangre , Ciego/cirugía , Modelos Animales de Enfermedad , Inducción Enzimática/efectos de los fármacos , Ligadura , Óxido Nítrico Sintasa de Tipo II/metabolismo , Proteína C/genética , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/uso terapéutico , Sepsis/sangre , Sepsis/etiología , Sepsis/patología , Índice de Severidad de la Enfermedad , Estadística como Asunto , Resultado del Tratamiento
11.
Shock ; 28(4): 468-76, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17558353

RESUMEN

Activated protein C (APC) is an important modulator of vascular function that has antithrombotic and anti-inflammatory properties. Studies in humans have shown modulation of endotoxin-induced hypotension by recombinant human APC, drotrecogin alfa (activated), however, the mechanism for this effect is unclear. We have found that APC suppresses the induction of the potent vasoactive peptide adrenomedullin (ADM) and could downregulate lipopolysaccharide (LPS)-induced ADM messenger RNA (mRNA) and nitrite levels in cell culture. This effect was dependent on signaling through protease-activated receptor 1. Addition of 1400W, an irreversible inducible nitric oxide synthase (iNOS) inhibitor, inhibited LPS-induced ADM mRNA, suggesting that ADM induction is NO mediated. Furthermore, in a rat model of endotoxemia, APC (100 microg/kg, i.v.) prevented LPS (10 mg/kg, i.v.)-induced hypotension, and suppressed ADM mRNA and protein expression. APC also inhibited iNOS mRNA and protein levels along with reduction in NO by-products (NOx). We also observed a significant reduction in iNOS-positive leukocytes adhering to vascular endothelium after APC treatment. Moreover, we found that APC inhibited the expression of interferon-gamma (IFN-gamma), a potent activator of iNOS. In a human study of LPS-induced hypotension, APC reduced the upregulation of plasma ADM levels, coincident with protection against the hypotensive response. Overall, we demonstrate that APC blocks the induction of ADM, likely mediated by IFN-gamma and iNOS, and suggests a mechanism that may account for ameliorating LPS-induced hypotension. Furthermore, our data provide a new understanding for the role of APC in modulating vascular response to insult.


Asunto(s)
Adrenomedulina/metabolismo , Hipotensión/prevención & control , Lipopolisacáridos/toxicidad , Proteína C/farmacología , Adrenomedulina/sangre , Adrenomedulina/genética , Animales , Western Blotting , Línea Celular , Movimiento Celular/efectos de los fármacos , AMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Expresión Génica/efectos de los fármacos , Humanos , Hipotensión/inducido químicamente , Interferón gamma/genética , Interferón gamma/metabolismo , Leucocitos/citología , Leucocitos/efectos de los fármacos , Leucocitos/metabolismo , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Masculino , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Nitritos/metabolismo , Óxidos de Nitrógeno/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
12.
Biomaterials ; 28(28): 4047-55, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17597201

RESUMEN

Multi-functional bilayer polymeric coatings are prepared with both controlled nitric oxide (NO) release and surface-bound active thrombomodulin (TM) alone or in combination with immobilized heparin. The outer-layer is made of CarboSil, a commercially available copolymer of silicone rubber (SR) and polyurethane (PU). The CarboSil is either carboxylated or aminated via an allophanate reaction with a diisocyanate compound followed by a urea-forming reaction between the generated isocyanate group of the polymer and the amine group of an amino acid (glycine), an oligopeptide (triglycine) or a diamine. The carboxylated CarboSil can then be used to immobilize TM through the formation of an amide bond between the surface carboxylic acid groups and the lysine residues of TM. Aminated CarboSil can also be employed to initially couple heparin to the surface, and then the carboxylic acid groups on heparin can be further used to anchor TM. Both surface-bound TM and heparin's activity are evaluated by chromogenic assays and found to be at clinically significant levels. The underlying NO release layer is made with another commercial SR-PU copolymer (PurSil) mixed with a lipophilic NO donor (N-diazeniumdiolated dibutylhexanediamine (DBHD/N(2)O(2))). The NO release rate can be tuned by changing the thickness of top coatings, and the duration of NO release at physiologically relevant levels can be as long as 2 weeks. The combination of controlled NO release as well as immobilized active TM and heparin from/on the same polymeric surface mimics the highly thromboresistant endothelium layer. Hence, such multifunctional polymer coatings should provide more blood-compatible surfaces for biomedical devices.


Asunto(s)
Materiales Biocompatibles Revestidos , Endotelio/metabolismo , Heparina/metabolismo , Óxido Nítrico/metabolismo , Polímeros , Trombomodulina/metabolismo , Sangre/metabolismo , Materiales Biocompatibles Revestidos/química , Materiales Biocompatibles Revestidos/metabolismo , Humanos , Ensayo de Materiales , Estructura Molecular , Cemento de Policarboxilato/química , Polímeros/química , Polímeros/metabolismo , Poliuretanos/química , Unión Proteica , Siliconas/química , Propiedades de Superficie
13.
Eur J Pharmacol ; 575(1-3): 158-67, 2007 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-17719030

RESUMEN

The present investigation was designed to identify potential biomarker(s) and assess the involvement of inflammatory pathway in dimethylnitrosamine (DMN)-induced liver fibrosis in rats. Following DMN-treatment (10 mg/ml/kg, i.p., given three consecutive days each week for 4 weeks) body and liver weights were significantly decreased concurrent with increasing severity of liver damage assessed by bridging fibrosis, a histopathologic assessment and characteristic of human liver disease. Protein C along with albumin, C-reactive-protein (CRP), haptoglobin and total protein were significantly reduced and correlated with changes in liver histopathology. Biochemical markers of liver functions were significantly increased and correlated with changes in liver histopathology and plasma levels of protein C. Soluble intracellular-adhesion-molecule-1 (sICAM-1) levels were increased significantly but were poorly correlated with histopathology and protein C levels. Inflammatory chemokines and other analytes, monocyte-chemoattractant-protein-1 and 3 (MCP-1 and MCP-3), macrophage-colony-stimulating-factor (M-CSF) were significantly increased during the disease progression, whereas macrophage-derived-chemokine (MDC) and CRP were significantly suppressed. Circulating neutrophils and monocytes were also increased along with disease progression. The differential changes in sICAM-1, hyaluronic acid, gamma-glutamyltranspeptidase (GGT), neutrophil and other inflammatory chemokines suggest the involvement of inflammatory pathways in DMN-induced liver fibrosis. In conclusion, the progressive changes in protein C along with other noninvasive biochemical parameters whose levels were significantly correlated with disease progression may serve as biomarkers for pharmacological assessment of targeted therapy for liver fibrosis.


Asunto(s)
Fibrinolíticos/sangre , Inflamación/patología , Cirrosis Hepática/diagnóstico , Proteína C/metabolismo , Animales , Biomarcadores/sangre , Proteína C-Reactiva/metabolismo , Quimiocinas/sangre , Dimetilnitrosamina , Haptoglobinas/metabolismo , Humanos , Ácido Hialurónico/sangre , Inmunohistoquímica/métodos , Inflamación/metabolismo , Molécula 1 de Adhesión Intercelular/sangre , Cirrosis Hepática/sangre , Cirrosis Hepática/inducido químicamente , Cirrosis Hepática/patología , Factor Estimulante de Colonias de Macrófagos/sangre , Neutrófilos/metabolismo , Ratas , Ratas Sprague-Dawley , Albúmina Sérica/metabolismo , Factores de Tiempo , gamma-Glutamiltransferasa/sangre
14.
Biochim Biophys Acta ; 1649(1): 106-17, 2003 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-12818196

RESUMEN

The role of lysines 37-39 (chymotrypsin numbering) in the 37-loop of the serine protease activated protein C (APC) was studied by expressing acidic and neutral recombinant APC (rAPC) mutants. Activity of the APC mutants was assessed using human plasma and plasma-purified and recombinant derivatives of protein C inhibitor (PCI; also known as plasminogen activator inhibitor-3) and alpha(1)-antitrypsin, with and without heparin. The catalytic properties of the mutants to small peptidyl substrates were essentially the same as wild-type rAPC (wt-rAPC), yet their plasma anticoagulant activities were diminished. Analysis of the rAPC-protease inhibitor complexes formed after addition of wt-rAPC and mutants to plasma revealed no change in the inhibition pattern by alpha(1)-antitrypsin but a reduction in mutant complex formation by PCI in the presence of heparin. Using purified serpins, we found that inhibition rates of the mutants were the same as wt-rAPC with alpha(1)-antitrypsin; however, PCI (plasma-derived and recombinant forms) inhibition rates of the acidic mutants were slightly faster than that of wt-rAPC without heparin. By contrast, PCI-heparin inhibition rates of the mutants were not substantially accelerated compared to wt-rAPC. The mutants had reduced heparin-binding properties compared to wt-rAPC. Molecular modeling of the PCI-APC complex with heparin suggests that heparin may function not only to bridge PCI to APC, but also to alleviate putative non-optimal intermolecular interactions. Our results suggest that the basic residues of the 37-loop of APC are involved in macromolecular substrate interactions and in heparin binding, and they influence inhibition by PCI (with or without heparin) but not by alpha(1)-antitrypsin, two important blood plasma serpins.


Asunto(s)
Inhibidor de Proteína C/farmacología , Proteína C/antagonistas & inhibidores , Proteína C/química , Inhibidores de Serina Proteinasa/farmacología , alfa 1-Antitripsina/farmacología , Sitios de Unión , Heparina/metabolismo , Humanos , Lisina/metabolismo , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Mutación , Plasma/metabolismo , Proteína C/genética , Proteína C/metabolismo , Inhibidor de Proteína C/sangre , Conformación Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Relación Estructura-Actividad
15.
Thromb Haemost ; 106(6): 1189-96, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21901240

RESUMEN

Recombinant human activated protein C (APC), which has both anticoagulant and anti-inflammatory properties, improves survival of patients with severe sepsis. This beneficial effect is especially apparent in patients with pneumococcal pneumonia. Earlier treatment with APC in sepsis has been associated with a better therapeutic response as compared to later treatment. In a mouse model it was recently confirmed that recombinant murine (rm-)APC decreases coagulation activation and improves survival in pneumococcal pneumonia; however, APC did not impact on the inflammatory response. The aim of this study was to determine the effect of APC treatment instigated early in infection on activation of coagulation and inflammation after induction of pneumococcal pneumonia. Mice were infected intranasally with viable S. pneumoniae . Mice were treated with rm-APC (125 µg) or vehicle intraperitoneally 12 hours after infection and were sacrificed after 20 hours, after which blood and organs were harvested for determination of bacterial outgrowth, coagulation activation and inflammatory markers. In this early treatment model, rm-APC treatment inhibited pulmonary and systemic activation of coagulation as reflected by lower levels of thrombin-antithrombin complexes and D-dimer. Moreover, rm-APC reduced the levels of a large number of cytokines and chemokines in the lung. When administered early in pneumococcal pneumonia, rm-APC inhibits systemic and pulmonary activation of coagulation and moreover exerts various anti-inflammatory effects in the lung.


Asunto(s)
Pulmón/efectos de los fármacos , Neumonía Neumocócica/inmunología , Proteína C/administración & dosificación , Proteínas Recombinantes/administración & dosificación , Streptococcus pneumoniae/inmunología , Animales , Antiinflamatorios/administración & dosificación , Antitrombina III/metabolismo , Biomarcadores/metabolismo , Coagulación Sanguínea/efectos de los fármacos , Citocinas/metabolismo , Modelos Animales de Enfermedad , Regulación hacia Abajo , Productos de Degradación de Fibrina-Fibrinógeno/metabolismo , Humanos , Pulmón/metabolismo , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Péptido Hidrolasas/metabolismo , Neumonía Neumocócica/sangre , Streptococcus pneumoniae/crecimiento & desarrollo , Streptococcus pneumoniae/patogenicidad
16.
Am J Physiol Renal Physiol ; 293(1): F245-54, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17409278

RESUMEN

Endothelial dysfunction contributes significantly to acute renal failure (ARF) during inflammatory diseases including septic shock. Previous studies have shown that activated protein C (APC) exhibits anti-inflammatory properties and modulates endothelial function. Therefore, we investigated the effect of APC on ARF in a rat model of endotoxemia. Rats subjected to lipopolysaccharide (LPS) treatment exhibited ARF as illustrated by markedly reduced peritubular capillary flow and increased serum blood urea nitrogen (BUN) levels. Using quantitative two-photon intravital microscopy, we observed that at 3 h post-LPS treatment, rat APC (0.1 mg/kg iv bolus) significantly improved peritubular capillary flow [288 +/- 15 microm/s (LPS) vs. 734 +/- 59 microm/s (LPS+APC), P = 0.0009, n = 6], and reduced leukocyte adhesion (P = 0.003) and rolling (P = 0.01) compared with the LPS-treated group. Additional experiments demonstrated that APC treatment significantly improved renal blood flow and reduced serum BUN levels compared with 24-h post-LPS treatment. Biochemical analysis revealed that APC downregulated inducible nitric oxide synthase (iNOS) mRNA levels and NO by-products in the kidney. In addition, APC modulated the renin-angiotensin system by reducing mRNA expression levels of angiotensin-converting enzyme-1 (ACE1), angiotensinogen, and increasing ACE2 mRNA levels in the kidney. Furthermore, APC significantly reduced ANG II levels in the kidney compared with the LPS-treated group. Taken together, these data suggest that APC can suppress LPS-induced ARF by modulating factors involved in vascular inflammation, including downregulation of renal iNOS and ANG II systems. Furthermore, the data suggest a potential therapeutic role for APC in the treatment of ARF.


Asunto(s)
Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/prevención & control , Angiotensina II/biosíntesis , Lipopolisacáridos/antagonistas & inhibidores , Lipopolisacáridos/toxicidad , Óxido Nítrico Sintasa de Tipo II/biosíntesis , Proteína C/farmacología , Lesión Renal Aguda/patología , Angiotensina II/antagonistas & inhibidores , Animales , Nitrógeno de la Urea Sanguínea , Adhesión Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Endotoxemia/metabolismo , Endotoxemia/patología , Riñón/efectos de los fármacos , Riñón/metabolismo , Riñón/patología , Leucocitos/efectos de los fármacos , Activación de Macrófagos/efectos de los fármacos , Masculino , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/antagonistas & inhibidores , Ratas , Ratas Sprague-Dawley , Circulación Renal/efectos de los fármacos , Sistema Renina-Angiotensina/efectos de los fármacos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Urea/sangre , Vasoconstricción/efectos de los fármacos , Vasodilatación/efectos de los fármacos
17.
J Am Soc Nephrol ; 18(3): 860-7, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17301189

RESUMEN

Protein C (PC) plays an important role in vascular function, and acquired deficiency during sepsis is associated with increased mortality in both animal models and in clinical studies. This study explored the consequences of PC suppression on the kidney in a cecal ligation and puncture model of polymicrobial sepsis. This study shows that a rapid drop in PC after sepsis is strongly associated with an increase in blood urea nitrogen, renal pathology, and expression of known markers of renal injury, including neutrophil gelatinase-associated lipocalin, CXCL1, and CXCL2. The endothelial PC receptor, which is required for the anti-inflammatory and antiapoptotic activity of activated PC (APC), was significantly increased after cecal ligation and puncture as well as in the microvasculature of human kidneys after injury. Treatment of septic animals with APC reduced blood urea nitrogen, renal pathology, and chemokine expression and dramatically reduced the induction of inducible nitric oxide synthase and caspase-3 activation in the kidney. The data demonstrate a clear link between acquired PC deficiency and renal dysfunction in sepsis and suggest a compensatory upregulation of the signaling receptor. Moreover, these data suggest that APC treatment may be effective in reducing inflammatory and apoptotic insult during sepsis-induced acute renal failure.


Asunto(s)
Lesión Renal Aguda/etiología , Riñón/metabolismo , Proteína C/metabolismo , Sepsis/metabolismo , Proteínas de Fase Aguda/metabolismo , Animales , Apoptosis , Biomarcadores/metabolismo , Caspasa 3/metabolismo , Ciego/cirugía , Quimiocina CXCL1 , Quimiocina CXCL2 , Quimiocinas CXC/metabolismo , Modelos Animales de Enfermedad , Riñón/patología , Lipocalina 2 , Lipocalinas , Óxido Nítrico Sintasa/metabolismo , Deficiencia de Proteína C/complicaciones , Proteínas Proto-Oncogénicas/metabolismo , Ratas , Ratas Sprague-Dawley , Regulación hacia Arriba
18.
Clin Vaccine Immunol ; 13(3): 426-32, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16522789

RESUMEN

Low levels of protein C (PC) predict outcome as early as 10 h after insult in a rat polymicrobial sepsis model and were associated with suppression of PC mRNA, upstream transcription factor FoxA2, and cofactor hepatocyte nuclear factor 6 (HNF6). Small interfering RNA suppression of FoxA2 in isolated hepatocytes demonstrated regulation of both its cofactor HNF6 and PC. Our data suggest that reduced FoxA2 may be important in the suppression of PC and resulting poor outcome in sepsis.


Asunto(s)
Factor Nuclear 3-beta del Hepatocito/fisiología , Proteína C/antagonistas & inhibidores , Proteína C/biosíntesis , Sepsis/metabolismo , Enfermedad Aguda , Animales , Secuencia de Bases , Biomarcadores , Ciego , Modelos Animales de Enfermedad , Femenino , Factor Nuclear 3-beta del Hepatocito/antagonistas & inhibidores , Factor Nuclear 3-beta del Hepatocito/genética , Ligadura , Datos de Secuencia Molecular , Valor Predictivo de las Pruebas , Pronóstico , Proteína C/genética , Proteína C/fisiología , Punciones , ARN Mensajero/antagonistas & inhibidores , ARN Mensajero/biosíntesis , ARN Interferente Pequeño/farmacología , Ratas , Ratas Sprague-Dawley , Sepsis/diagnóstico , Sepsis/mortalidad
19.
Proc Natl Acad Sci U S A ; 100(8): 4423-8, 2003 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-12671072

RESUMEN

Human activated protein C (APC) is an antithrombotic, antiinflammatory serine protease that plays a central role in vascular homeostasis, and activated recombinant protein C, drotrecogin alfa (activated), has been shown to reduce mortality in patients with severe sepsis. Similar to other serine proteases, functional APC levels are regulated by the serine protease inhibitor family of proteins including alpha(1)-antitrypsin and protein C inhibitor. Using APC-substrate modeling, we designed and produced a number of derivatives with the goal of altering the proteolytic specificity of APC such that the variants exhibited resistance to inactivation by protein C inhibitor and alpha(1)-antitrypsin yet maintained their primary anticoagulant activity. Substitutions at Leu-194 were of particular interest, because they exhibited 4- to 6-fold reductions in the rate of inactivation in human plasma and substantially increased pharmacokinetic profiles compared with wild-type APC. This was achieved with minimal impairment of the anticoagulant/antithrombotic activity of APC. These data demonstrate the ability to selectively modulate substrate specificity and subsequently affect in vivo performance and suggest therapeutic opportunities for the use of protein C derivatives in disease states with elevated serine protease inhibitor levels.


Asunto(s)
Proteína C/química , Proteína C/metabolismo , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Anticoagulantes/química , Anticoagulantes/metabolismo , Anticoagulantes/farmacología , Variación Genética , Humanos , Técnicas In Vitro , Cinética , Macaca fascicularis , Modelos Moleculares , Proteína C/genética , Proteína C/farmacología , Inhibidor de Proteína C/farmacología , Ingeniería de Proteínas , Conejos , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología , Serpinas/farmacología , Especificidad por Sustrato , alfa 1-Antitripsina/farmacología
20.
Crit Care Med ; 32(7): 1570-8, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15241104

RESUMEN

OBJECTIVE: To evaluate protein C and other factors associated with the septic response as predictors of mortality in a clinically relevant animal model of sepsis. DESIGN: Laboratory investigation. SETTING: Eli Lilly and Company discovery research laboratory. SUBJECTS: Forty female Sprague Dawley rats weighing 245-265 g. INTERVENTIONS: Polyethylene catheters were surgically implanted into the femoral vein and sepsis was induced by cecal ligation and puncture (CLP). A solution of 5% dextrose in 0.9 % saline was continuously infused via femoral catheters immediately following surgery. Blood sampling was done before surgery and at 6 and 20 hrs after surgery. Rats were then monitored for survival out to 4 days. MEASUREMENTS AND MAIN RESULTS: Blood collections were used to measure blood glucose, bacteremia, plasma protein C, D-dimer, hormones, chemokines, cytokines, and myoglobin (as a marker of organ damage). Mortality was categorized into three groups: early death (before 30 hrs post-CLP), late death (after 30 hrs post-CLP), and survivors (96 hrs post-CLP). Compared with survivors, early death rats had statistically significant differences in 30 variables indicative of severe inflammation, coagulopathy, and muscle damage including less bacterial clearance, hypoglycemia, lower plasma protein C, higher plasma D dimer, higher plasma cytokine/ chemokines, and higher plasma myoglobin concentrations. Twenty variables had a moderate to strong correlation with time of death. Receiver operator characteristic curves generated from a simple logistic regression model indicated that KC and macrophage inflammatory protein-2, rodent homologues of the human growth related oncogene CXC chemokine family, and protein C were the best predictors of mortality in this model. CONCLUSIONS: The data from this study indicate that an early decrease in protein C concentration predicts poor outcome in a rat sepsis model. The data further indicate that increases in the CXC chemokines macrophage inflammatory protein-2 and KC precede poor outcome.


Asunto(s)
Proteína C/metabolismo , Sepsis/metabolismo , Animales , Biomarcadores , Glucemia , Femenino , Ligadura , Modelos Biológicos , Valor Predictivo de las Pruebas , Punciones , Curva ROC , Ratas , Ratas Sprague-Dawley , Sepsis/sangre , Sepsis/mortalidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA