Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Blood ; 119(3): 874-83, 2012 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-22117049

RESUMO

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.


Assuntos
Apoptose , Citoproteção , Podócitos/metabolismo , Proteína C/metabolismo , Receptor PAR-1/metabolismo , Receptores de Trombina/metabolismo , Animais , Anticoagulantes/metabolismo , Comunicação Celular , Células Cultivadas , Humanos , Glomérulos Renais/citologia , Glomérulos Renais/metabolismo , Lipopolissacarídeos/farmacologia , Microdomínios da Membrana , Camundongos , Podócitos/efeitos dos fármacos , Podócitos/patologia , Multimerização Proteica , Transdução de Sinais , Trombina
2.
Nat Med ; 13(11): 1349-58, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17982464

RESUMO

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.


Assuntos
Apoptose , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Experimental/prevenção & controle , Nefropatias Diabéticas/patologia , Nefropatias Diabéticas/prevenção & controle , Endotélio Vascular/patologia , Podócitos/patologia , Proteína C/fisiologia , Substituição de Aminoácidos/genética , Animais , Apoptose/genética , Linhagem Celular Transformada , Células Cultivadas , Citoproteção/genética , Diabetes Mellitus Experimental/enzimologia , Diabetes Mellitus Experimental/genética , Nefropatias Diabéticas/enzimologia , Nefropatias Diabéticas/genética , Endotélio Vascular/enzimologia , Ativação Enzimática/genética , Humanos , Glomérulos Renais/irrigação sanguínea , Glomérulos Renais/enzimologia , Glomérulos Renais/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Camundongos Transgênicos , Microcirculação/enzimologia , Microcirculação/patologia , Podócitos/enzimologia , Proteína C/biossíntese , Proteína C/genética , Transdução de Sinais/genética , Trombomodulina/fisiologia
3.
Nature ; 451(7182): 1076-81, 2008 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-18278032

RESUMO

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.


Assuntos
Perfilação da Expressão Gênica , Esclerose Múltipla/metabolismo , Esclerose Múltipla/patologia , Proteômica , Adulto , Animais , Coagulação Sanguínea , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/metabolismo , Encefalomielite Autoimune Experimental/patologia , Feminino , Humanos , Inflamação/metabolismo , Inflamação/patologia , Masculino , Camundongos , Pessoa de Meia-Idade , Esclerose Múltipla/classificação , Esclerose Múltipla/tratamento farmacológico , Proteína C/genética , Proteína C/metabolismo , Proteína C/farmacologia , Células Th1/imunologia , Células Th2/imunologia , Trombina/antagonistas & inibidores , Trombina/metabolismo
4.
Am J Physiol Renal Physiol ; 302(6): F703-12, 2012 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-22129968

RESUMO

Chronic kidney disease pathogenesis involves both tubular and vascular injuries. Despite abundant investigations to identify the risk factors, the involvement of chronic endothelial dysfunction in developing nephropathies is insufficiently explored. Previously, soluble thrombomodulin (sTM), a cofactor in the activation of protein C, has been shown to protect endothelial function in models of acute kidney injury. In this study, the role for sTM in treating chronic kidney disease was explored by employing a mouse model of chronic vascular activation using endothelial-specific TNF-α-expressing (tie2-TNF) mice. Analysis of kidneys from these mice after 3 mo showed no apparent phenotype, whereas 6-mo-old mice demonstrated infiltration of CD45-positive leukocytes accompanied by upregulated gene expression of inflammatory chemokines, markers of kidney injury, and albuminuria. Intervention with murine sTM with biweekly subcutaneous injections during this window of disease development between months 3 and 6 prevented the development of kidney pathology. To better understand the mechanisms of these findings, we determined whether sTM could also prevent chronic endothelial cell activation in vitro. Indeed, treatment with sTM normalized increased chemokines, adhesion molecule expression, and reduced transmigration of monocytes in continuously activated TNF-expressing endothelial cells. Our results suggest that vascular inflammation associated with vulnerable endothelium can contribute to loss in renal function as suggested by the tie2-TNF mice, a unique model for studying the role of vascular activation and inflammation in chronic kidney disease. Furthermore, the ability to restore the endothelial balance by exogenous administration of sTM via downregulation of specific adhesion molecules and chemokines suggests a potential for therapeutic intervention in kidney disease associated with chronic inflammation.


Assuntos
Albuminúria/prevenção & controle , Inflamação/tratamento farmacológico , Falência Renal Crônica/metabolismo , Trombomodulina/uso terapêutico , Albuminúria/tratamento farmacológico , Albuminúria/genética , Albuminúria/metabolismo , Animais , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Regulação da Expressão Gênica/fisiologia , Inflamação/genética , Inflamação/metabolismo , Falência Renal Crônica/genética , Camundongos , Camundongos Transgênicos , Receptores Proteína Tirosina Quinases/genética , Receptores Proteína Tirosina Quinases/metabolismo , Receptor TIE-2 , Técnicas de Cultura de Tecidos , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
5.
J Infect Dis ; 202(10): 1600-7, 2010 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-20925531

RESUMO

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.


Assuntos
Transtornos de Proteínas de Coagulação/tratamento farmacológico , Transtornos de Proteínas de Coagulação/microbiologia , Pneumonia Pneumocócica/tratamento farmacológico , Proteína C/uso terapêutico , Animais , Antibacterianos/uso terapêutico , Fatores de Coagulação Sanguínea/antagonistas & inibidores , Ceftriaxona/administração & dosagem , Ceftriaxona/uso terapêutico , Avaliação Pré-Clínica de Medicamentos , Quimioterapia Combinada , Humanos , Injeções Intraperitoneais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pneumonia Pneumocócica/complicações , Proteína C/administração & dosagem , Proteína C/farmacologia , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/uso terapêutico
6.
J Clin Invest ; 117(7): 1951-60, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17557119

RESUMO

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.


Assuntos
Doenças Inflamatórias Intestinais/metabolismo , Doenças Inflamatórias Intestinais/patologia , Microcirculação/patologia , Proteína C/metabolismo , Transdução de Sinais , Animais , Antígenos CD/metabolismo , Adesão Celular/efeitos dos fármacos , Células Cultivadas , Quimiocinas/biossíntese , Regulação para Baixo , Receptor de Proteína C Endotelial , Endotélio/patologia , Humanos , Imuno-Histoquímica , Inflamação/metabolismo , Inflamação/patologia , Molécula 1 de Adesão Intercelular/metabolismo , Leucócitos/citologia , Camundongos , Microcirculação/metabolismo , Proteína C/farmacologia , Receptores de Superfície Celular/metabolismo , Transdução de Sinais/efeitos dos fármacos , Trombomodulina/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Molécula 1 de Adesão de Célula Vascular/metabolismo
7.
Crit Care ; 14(2): R65, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20398279

RESUMO

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.


Assuntos
Anticoagulantes/antagonistas & inibidores , Anticoagulantes/uso terapêutico , Coagulação Sanguínea/efeitos dos fármacos , Vírus da Influenza A Subtipo H1N1/patogenicidade , Infecções por Orthomyxoviridae/sangue , Proteína C/antagonistas & inibidores , Proteína C/uso terapêutico , Animais , Anticoagulantes/metabolismo , Regulação para Baixo/genética , Vírus da Influenza A Subtipo H1N1/genética , Pulmão/efeitos dos fármacos , Pulmão/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais , Infecções por Orthomyxoviridae/fisiopatologia , Proteína C/genética , Proteína C/metabolismo , Proteínas Recombinantes , Carga Viral
8.
J Am Soc Nephrol ; 20(3): 524-34, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19176699

RESUMO

Altered coagulation and inflammation contribute to the pathogenesis of ischemic renal injury. Thrombomodulin is a necessary factor in the anticoagulant protein C pathway and has inherent anti-inflammatory properties. We studied the effect of soluble thrombomodulin (sTM) in a hypoperfusion model of ischemic kidney injury. To markedly reduce infrarenal aortic blood flow and femoral arterial pressures, we clamped the suprarenal aorta of rats, occluding them 90%, for 60 min. Reversible acute kidney injury (AKI) occurred at 24 h in rats subjected to hypoperfusion. Histologic analysis at 24 h revealed acute tubular necrosis (ATN), and intravital two-photon microscopy showed flow abnormalities in the microvasculature and defects of endothelial permeability. Pretreatment with rat sTM markedly reduced both I-R-induced renal dysfunction and tubular histologic injury scores. sTM also significantly improved microvascular erythrocyte flow rates, reduced microvascular endothelial leukocyte rolling and attachment, and minimized endothelial permeability to infused fluorescence dextrans, assessed by intravital quantitative multiphoton microscopy. Furthermore, sTM administered 2 h after reperfusion protected against ischemia-induced renal dysfunction at 24 h and improved survival. By using an sTM variant, we also determined that the protective effects of sTM were independent of its ability to generate activated protein C. These data suggest that sTM may have therapeutic potential for ischemic AKI.


Assuntos
Isquemia/prevenção & controle , Rim/irrigação sanguínea , Rim/efeitos dos fármacos , Trombomodulina/administração & dosagem , Animais , Sequência de Bases , Permeabilidade Capilar/efeitos dos fármacos , Adesão Celular/efeitos dos fármacos , Primers do DNA/genética , Fibrinolíticos/administração & dosagem , Variação Genética , Rim/lesões , Necrose Tubular Aguda/patologia , Necrose Tubular Aguda/fisiopatologia , Necrose Tubular Aguda/prevenção & controle , Leucócitos/efeitos dos fármacos , Leucócitos/fisiologia , Masculino , Proteína C/metabolismo , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/genética , Circulação Renal/efeitos dos fármacos , Solubilidade , Trombomodulina/genética , Trombomodulina/fisiologia
9.
J Am Soc Nephrol ; 20(2): 267-77, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19092124

RESUMO

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.


Assuntos
Nefropatias/metabolismo , Rim/lesões , Proteína C/fisiologia , Animais , Humanos , Inflamação , Interleucina-18/metabolismo , Interleucina-6/metabolismo , Rim/metabolismo , Lipopolissacarídeos/metabolismo , Masculino , Microcirculação , Proteína C/metabolismo , Ratos , Ratos Sprague-Dawley , Receptor PAR-1/metabolismo , Transdução de Sinais
10.
J Pharmacol Exp Ther ; 325(1): 17-26, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18182560

RESUMO

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.


Assuntos
Inibidores da Enzima Conversora de Angiotensina/uso terapêutico , Peptidil Dipeptidase A/efeitos dos fármacos , Deficiência de Proteína C/tratamento farmacológico , Síndrome do Desconforto Respiratório/tratamento farmacológico , Sepse/complicações , Síndrome de Resposta Inflamatória Sistêmica/tratamento farmacológico , Enzima de Conversão de Angiotensina 2 , Inibidores da Enzima Conversora de Angiotensina/farmacologia , Animais , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas Inflamatórias de Macrófagos/genética , Óxido Nítrico Sintase Tipo II/genética , Inibidor 1 de Ativador de Plasminogênio/genética , Deficiência de Proteína C/etiologia , Ratos
11.
Arterioscler Thromb Vasc Biol ; 27(12): 2634-41, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17932312

RESUMO

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.


Assuntos
Antígenos CD/metabolismo , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Células Endoteliais/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Proteína C/metabolismo , Receptores de Superfície Celular/metabolismo , Transdução de Sinais , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Antígenos CD/genética , Células Cultivadas , Citoproteção , Células Endoteliais/enzimologia , Receptor de Proteína C Endotelial , Ativação Enzimática , Humanos , Fosforilação , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Interferência de RNA , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , Receptor PAR-1/metabolismo , Receptores de Superfície Celular/genética , Receptores de Lisoesfingolipídeo/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/genética , Fatores de Tempo , Transcrição Gênica , Fator de Necrose Tumoral alfa/metabolismo
12.
Adv Exp Med Biol ; 614: 83-91, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18290317

RESUMO

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.


Assuntos
Anticoagulantes/uso terapêutico , Quimiocinas/metabolismo , Proteína C/fisiologia , Proteína C/uso terapêutico , Sepse/tratamento farmacológico , Animais , Biomarcadores/sangue , Ceco/cirurgia , Modelos Animais de Doenças , Indução Enzimática/efeitos dos fármacos , Ligadura , Óxido Nítrico Sintase Tipo II/metabolismo , Proteína C/genética , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/uso terapêutico , Sepse/sangue , Sepse/etiologia , Sepse/patologia , Índice de Gravidade de Doença , Estatística como Assunto , Resultado do Tratamento
13.
Shock ; 28(4): 468-76, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17558353

RESUMO

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.


Assuntos
Adrenomedulina/metabolismo , Hipotensão/prevenção & controle , Lipopolissacarídeos/toxicidade , Proteína C/farmacologia , Adrenomedulina/sangue , Adrenomedulina/genética , Animais , Western Blotting , Linhagem Celular , Movimento Celular/efeitos dos fármacos , AMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Expressão Gênica/efeitos dos fármacos , Humanos , Hipotensão/induzido quimicamente , Interferon gama/genética , Interferon gama/metabolismo , Leucócitos/citologia , Leucócitos/efeitos dos fármacos , Leucócitos/metabolismo , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Masculino , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/metabolismo , Nitritos/metabolismo , Óxidos de Nitrogênio/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa
14.
Biomaterials ; 28(28): 4047-55, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17597201

RESUMO

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.


Assuntos
Materiais Revestidos Biocompatíveis , Endotélio/metabolismo , Heparina/metabolismo , Óxido Nítrico/metabolismo , Polímeros , Trombomodulina/metabolismo , Sangue/metabolismo , Materiais Revestidos Biocompatíveis/química , Materiais Revestidos Biocompatíveis/metabolismo , Humanos , Teste de Materiais , Estrutura Molecular , Cimento de Policarboxilato/química , Polímeros/química , Polímeros/metabolismo , Poliuretanos/química , Ligação Proteica , Silicones/química , Propriedades de Superfície
15.
Front Biosci ; 11: 676-98, 2006 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-16146761

RESUMO

Protein C is a plasma protease that when activated plays a central role in modulating the function of the vascular endothelium and its interface with the innate immune system. A recombinant form of human activated protein C (APC), drotrecogin alfa (activated), has shown efficacy in a number of preclinical models of thrombosis and ischemia and reduces mortality in patients that have a high risk of dying from severe sepsis. Studies have begun to elucidate the mechanism for the multifunctional role of APC in modulating not only coagulation, but also inflammation and apoptotic processes. From gene profiling to pharmacology studies, drotrecogin alfa (activated) appears to directly modulate endothelial dysfunction by blocking cytokine signaling, functional cell adhesion expression, vascular permeability and preventing the induction of apoptosis. Moreover, APC, via endothelial protein C receptor/protease activated receptor-1 mediated mechanisms, also appears to directly modulate leukocyte migration and adhesion. The ability of APC to suppress pro-inflammatory pathways and enhance cellular survival suggests that APC has a role in the adaptive response at the vessel wall, in which it protects the wall from vascular insult and prolongs endothelial, cellular, and organ survival. The emerging data further suggest that APC effectively modulates the complex changes that occur during multi-system activation and dysfunction in sepsis.


Assuntos
Proteína C/fisiologia , Sepse/sangue , Anti-Inflamatórios/farmacologia , Coagulação Sanguínea , Fatores de Coagulação Sanguínea/metabolismo , Adesão Celular , Movimento Celular , Citocinas/metabolismo , Endotélio Vascular/embriologia , Fibrinólise , Fibrinolíticos/farmacologia , Humanos , Inflamação , Isquemia/patologia , Leucócitos/metabolismo , Pulmão/patologia , Proteína C/metabolismo , Proteína C/uso terapêutico , Receptores de Superfície Celular/metabolismo , Receptores Ativados por Proteinase/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/uso terapêutico , Sepse/metabolismo , Transdução de Sinais
16.
Biochim Biophys Acta ; 1649(1): 106-17, 2003 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-12818196

RESUMO

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.


Assuntos
Inibidor da Proteína C/farmacologia , Proteína C/antagonistas & inibidores , Proteína C/química , Inibidores de Serina Proteinase/farmacologia , alfa 1-Antitripsina/farmacologia , Sítios de Ligação , Heparina/metabolismo , Humanos , Lisina/metabolismo , Modelos Moleculares , Mutagênese Sítio-Dirigida , Mutação , Plasma/metabolismo , Proteína C/genética , Proteína C/metabolismo , Inibidor da Proteína C/sangue , Conformação Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Relação Estrutura-Atividade
17.
PLoS One ; 10(3): e0120770, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25790110

RESUMO

Endothelial dysfunction contributes to the development of acute kidney injury (AKI) in animal models of ischemia reperfusion injury and sepsis. There are limited data on markers of endothelial dysfunction in human AKI. We hypothesized that Protein C (PC) and soluble thrombomodulin (sTM) levels could predict AKI. We conducted a multicenter prospective study in 80 patients to assess the relationship of PC and sTM levels to AKI, defined by the AKIN creatinine (AKI Scr) and urine output criteria (AKI UO). We measured marker levels for up to 10 days from intensive care unit admission. We used area under the curve (AUC) and time-dependent multivariable Cox proportional hazard model to predict AKI and logistic regression to predict mortality/non-renal recovery. Protein C and sTM were not different in patients with AKI UO only versus no AKI. On intensive care unit admission, as PC levels are usually lower with AKI Scr, the AUC to predict the absence of AKI was 0.63 (95%CI 0.44-0.78). The AUC using log10 sTM levels to predict AKI was 0.77 (95%CI 0.62-0.89), which predicted AKI Scr better than serum and urine neutrophil gelatinase-associated lipocalin (NGAL) and cystatin C, urine kidney injury molecule-1 and liver-fatty acid-binding protein. In multivariable models, PC and urine NGAL levels independently predicted AKI (p=0.04 and 0.02) and PC levels independently predicted mortality/non-renal recovery (p=0.04). In our study, PC and sTM levels can predict AKI Scr but are not modified during AKI UO alone. PC levels could independently predict mortality/non-renal recovery. Additional larger studies are needed to define the relationship between markers of endothelial dysfunction and AKI.


Assuntos
Injúria Renal Aguda/sangue , Proteína C/metabolismo , Trombomodulina/sangue , Trombomodulina/química , Injúria Renal Aguda/diagnóstico , Injúria Renal Aguda/mortalidade , Biomarcadores/sangue , Estado Terminal , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Prognóstico , Estudos Prospectivos , Solubilidade
18.
Thromb Haemost ; 106(6): 1189-96, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21901240

RESUMO

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.


Assuntos
Pulmão/efeitos dos fármacos , Pneumonia Pneumocócica/imunologia , Proteína C/administração & dosagem , Proteínas Recombinantes/administração & dosagem , Streptococcus pneumoniae/imunologia , Animais , Anti-Inflamatórios/administração & dosagem , Antitrombina III/metabolismo , Biomarcadores/metabolismo , Coagulação Sanguínea/efeitos dos fármacos , Citocinas/metabolismo , Modelos Animais de Doenças , Regulação para Baixo , Produtos de Degradação da Fibrina e do Fibrinogênio/metabolismo , Humanos , Pulmão/metabolismo , Pulmão/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Peptídeo Hidrolases/metabolismo , Pneumonia Pneumocócica/sangue , Streptococcus pneumoniae/crescimento & desenvolvimento , Streptococcus pneumoniae/patogenicidade
19.
Curr Drug Targets ; 10(12): 1212-26, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19715537

RESUMO

Protein C is a plasma serine protease that when activated plays a central role in modulating the function of the vascular endothelium and its interface with the innate immune system. Activated protein C (APC) has a dual mechanism of action via the feedback inhibition of thrombin generation, and as an agonist of protease activated receptor-1 (PAR-1). Through different cofactor interactions, this dual mechanism of antithrombotic and cytoprotective activity results in the ability of APC to modulate endothelial dysfunction by blocking cytokine signaling, functional cell adhesion expression, vascular permeability, apoptosis, and modulating leukocyte migration and adhesion. Deficiency in protein C, which occurs during systemic inflammatory activation, is highly associated with organ dysfunction. APC has shown efficacy in a number of preclinical models of thrombosis and ischemia, and the recombinant human APC drotrecogin alfa (activated), reduces mortality in patients with high-risk severe sepsis. The ability of APC to suppress pro-inflammatory pathways and enhance cellular survival suggests that APC plays a key role in the adaptive response to protect the vessel wall from insult and to enhance endothelial, cellular, and organ survival. The focus of this review will be to summarize the emerging data suggesting the potential therapeutic benefit of APC and related members of the pathway in the prevention and treatment of acute kidney injury.


Assuntos
Injúria Renal Aguda/tratamento farmacológico , Injúria Renal Aguda/metabolismo , Proteína C/fisiologia , Receptor PAR-1/efeitos dos fármacos , Resistência à Proteína C Ativada/patologia , Injúria Renal Aguda/prevenção & controle , Animais , Humanos , Rim/fisiologia , Substâncias Protetoras/uso terapêutico , Proteína C/uso terapêutico , Proteínas Recombinantes/uso terapêutico , Transdução de Sinais/efeitos dos fármacos
20.
Am J Physiol Renal Physiol ; 293(1): F245-54, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17409278

RESUMO

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.


Assuntos
Injúria Renal Aguda/induzido quimicamente , Injúria Renal Aguda/prevenção & controle , Angiotensina II/biossíntese , Lipopolissacarídeos/antagonistas & inibidores , Lipopolissacarídeos/toxicidade , Óxido Nítrico Sintase Tipo II/biossíntese , Proteína C/farmacologia , Injúria Renal Aguda/patologia , Angiotensina II/antagonistas & inibidores , Animais , Nitrogênio da Ureia Sanguínea , Adesão Celular/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Endotoxemia/metabolismo , Endotoxemia/patologia , Rim/efeitos dos fármacos , Rim/metabolismo , Rim/patologia , Leucócitos/efeitos dos fármacos , Ativação de Macrófagos/efeitos dos fármacos , Masculino , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo II/antagonistas & inibidores , Ratos , Ratos Sprague-Dawley , Circulação Renal/efeitos dos fármacos , Sistema Renina-Angiotensina/efeitos dos fármacos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ureia/sangue , Vasoconstrição/efeitos dos fármacos , Vasodilatação/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA