Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
Brain Res Mol Brain Res ; 136(1-2): 177-88, 2005 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-15893602

RESUMO

The apolipoprotein E gene (APOE) plays an important role in the pathogenesis of Alzheimer's disease (AD), and amyloid plaque comprised mostly of the amyloid-beta peptide (A(beta)) is one of the major hallmarks of AD. However, the relationship between these two important molecules is poorly understood. We examined how A(beta) treatment affects APOE expression in cultured cells and tested the role of the transcription factor NF-(kappa)B in APOE gene regulation. To delineate NF-(kappa)B's role, we have characterized a 1098 nucleotide (nt) segment containing the 5'-flanking region of the human APOE gene (-1054/+44, +1 transcription start site). Sequence analysis of this region suggests the presence of two potential NF-(kappa)B elements. To demonstrate promoter activity, the region was cloned upstream of a promoterless luciferase (reporter) gene. This segment was able to drive expression of luciferase in transient transfections of human fetal glial cells. Promoter activity was stimulated twofold by A(beta)(1-40) (25 microM, 24 h) treatment. Pretreatment with double-stranded DNA decoy oligonucleotides against NF-(kappa)B (2 microM) reduced A(beta) stimulation. Deletion and mutagenetic analyses demonstrated that the distal NF-(kappa)B element was functional and showed a strong DNA-protein complex band in gel shift analysis, similar to that from control NF-(kappa)B consensus element. An anti-inflammatory and anti-NF-(kappa)B drug, sodium salicylate, significantly blocked A(beta)-induced APOE promoter function. Our data provide evidence that upregulation of APOE by A(beta) in astroglial cells is mediated by an NF-(kappa)B-element present in the 5'-flanking region of the APOE gene.


Assuntos
Peptídeos beta-Amiloides/farmacologia , Apolipoproteínas E/genética , Astrócitos/efeitos dos fármacos , NF-kappa B/metabolismo , Fragmentos de Peptídeos/farmacologia , Regiões Promotoras Genéticas/efeitos dos fármacos , Apolipoproteínas E/metabolismo , Astrócitos/metabolismo , Linhagem Celular , Clonagem Molecular/métodos , Inibidores de Ciclo-Oxigenase/farmacologia , DNA/metabolismo , Relação Dose-Resposta a Droga , Interações Medicamentosas , Ensaio de Desvio de Mobilidade Eletroforética/métodos , Genes Reporter/fisiologia , Humanos , Luciferases/metabolismo , Mutagênese/fisiologia , NF-kappa B/genética , Regiões Promotoras Genéticas/fisiologia , Salicilatos/farmacologia , Ativação Transcricional/efeitos dos fármacos , Ativação Transcricional/fisiologia , Transfecção/métodos
10.
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
11.
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
12.
J Biol Chem ; 282(51): 36837-44, 2007 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-17928287

RESUMO

Inducible nitric-oxide synthase (iNOS) plays a central role in the regulation of vascular function and response to injury. A central mediator controlling iNOS expression is transforming growth factor-beta (TGF-beta), which represses its expression through a mechanism that is poorly understood. We have identified a binding site in the iNOS promoter that interacts with the nuclear heterodimer TCF11/MafG using chromatin immunoprecipitation and mutation analyses. We demonstrate that binding at this site acts to repress the induction of iNOS gene expression by cytokines. We show that this repressor is induced by TGF-beta1 and by Smad6-short, which enhances TGF-beta signaling. In contrast, the up-regulation of TCF11/MafG binding could be suppressed by overexpression of the TGF-beta inhibitor Smad7, and a small interfering RNA to TCF11 blocked the suppression of iNOS by TGF-beta. The binding of TCF11/MafG to the iNOS promoter could be enhanced by phorbol 12-myristate 13-acetate and suppressed by the protein kinase C inhibitor staurosporine. Moreover, the induction of TCF11/MafG binding by TGF-beta and Smad6-short could be blocked by staurosporine, and the effect of TGF-beta was blocked by the selective protein kinase C inhibitor calphostin C. Consistent with the in vitro data, we found suppression of TCF11 coincident with iNOS up-regulation in a rat model of endotoxemia, and we observed a highly significant negative correlation between TCF11 and nitric oxide production. Furthermore, treatment with activated protein C, a serine protease effective in septic shock, blocked the down-regulation of TCF11 and suppressed endotoxin-induced iNOS. Overall, our results demonstrate a novel mechanism by which iNOS expression is regulated in the context of inflammatory activation.


Assuntos
Regulação Enzimológica da Expressão Gênica , Fator 1-beta Nuclear de Hepatócito/metabolismo , Óxido Nítrico Sintase Tipo II/biossíntese , Elementos de Resposta , Fator de Crescimento Transformador beta1/metabolismo , Animais , Carcinógenos/farmacologia , Células Cultivadas , Dimerização , Modelos Animais de Doenças , Endotoxemia/enzimologia , Endotoxemia/genética , Inibidores Enzimáticos/farmacologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica/genética , Fator 1-beta Nuclear de Hepatócito/antagonistas & inibidores , Fator 1-beta Nuclear de Hepatócito/genética , Humanos , Inflamação/enzimologia , Inflamação/genética , Fator de Transcrição MafG/genética , Fator de Transcrição MafG/metabolismo , Masculino , Mutação , Naftalenos/farmacologia , Óxido Nítrico Sintase Tipo II/genética , Proteína Quinase C/antagonistas & inibidores , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/farmacologia , Ratos , Ratos Sprague-Dawley , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Elementos de Resposta/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Proteína Smad6/genética , Proteína Smad6/metabolismo , Proteína Smad7/genética , Proteína Smad7/metabolismo , Estaurosporina/farmacologia , Acetato de Tetradecanoilforbol/farmacologia , Fator de Crescimento Transformador beta1/farmacologia
13.
J Am Soc Nephrol ; 18(3): 860-7, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17301189

RESUMO

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.


Assuntos
Injúria Renal Aguda/etiologia , Rim/metabolismo , Proteína C/metabolismo , Sepse/metabolismo , Proteínas de Fase Aguda/metabolismo , Animais , Apoptose , Biomarcadores/metabolismo , Caspase 3/metabolismo , Ceco/cirurgia , Quimiocina CXCL1 , Quimiocina CXCL2 , Quimiocinas CXC/metabolismo , Modelos Animais de Doenças , Rim/patologia , Lipocalina-2 , Lipocalinas , Óxido Nítrico Sintase/metabolismo , Deficiência de Proteína C/complicações , Proteínas Proto-Oncogênicas/metabolismo , Ratos , Ratos Sprague-Dawley , Regulação para Cima
14.
Clin Vaccine Immunol ; 13(3): 426-32, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16522789

RESUMO

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.


Assuntos
Fator 3-beta Nuclear de Hepatócito/fisiologia , Proteína C/antagonistas & inibidores , Proteína C/biossíntese , Sepse/metabolismo , Doença Aguda , Animais , Sequência de Bases , Biomarcadores , Ceco , Modelos Animais de Doenças , Feminino , Fator 3-beta Nuclear de Hepatócito/antagonistas & inibidores , Fator 3-beta Nuclear de Hepatócito/genética , Ligadura , Dados de Sequência Molecular , Valor Preditivo dos Testes , Prognóstico , Proteína C/genética , Proteína C/fisiologia , Punções , RNA Mensageiro/antagonistas & inibidores , RNA Mensageiro/biossíntese , RNA Interferente Pequeno/farmacologia , Ratos , Ratos Sprague-Dawley , Sepse/diagnóstico , Sepse/mortalidade
15.
J Biol Chem ; 280(15): 14943-7, 2005 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-15716278

RESUMO

Plasminogen activator inhibitor-1 (PAI-1) is a serpin class protease inhibitor that plays a central role in the regulation of vascular function and tissue remodeling by modulating thrombosis, inflammation, and the extracellular matrix. A central mediator controlling PAI-1 is transforming growth factor-beta (TGF-beta), which induces its expression and promotes fibrosis. We have found that a unique member of the Smad family of signal transduction molecules, Smad6s, modulates the expression of PAI-1. Overexpression of Smad6s in endothelial cells increases promoter activity and PAI-1 secretion, and an antisense to Smad6s suppresses the induction of PAI-1 by TGF-beta. The effect of Smad6s on the PAI-1 promoter appeared to be the result of increase binding of the forkhead winged helix factor FoxD1 to a TGF-beta-responsive element. Furthermore, the effect of Smad6s on PAI-1 up-regulation and on FoxD1 binding was found to result from up-regulation of TGF-beta and could be inhibited by the blocking TGF-beta signaling with Smad7. The ability of Smad6s to regulate the TGF-beta promoter and subsequent PAI-1 induction was suppressed by a selective protein kinase C-beta (PKC-beta) inhibitor. Consistent with the in vitro data, we found that increased Smad6s in diseased vessels correlated with increased TGF-beta and PAI-1 levels. Overall, our results demonstrate that the level of Smad6s can alter the level of TGF-beta and the subsequent induction of PAI-1 via a FoxD1 transcription site. Furthermore, our data suggest that this process, which is up-regulated in diseased vessels, can be modulated by the inhibition of PKC-beta.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Proteína Quinase C/metabolismo , Transativadores/fisiologia , Fatores de Transcrição/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Antígenos CD34/biossíntese , Núcleo Celular/metabolismo , Cloranfenicol O-Acetiltransferase/metabolismo , Biologia Computacional , DNA/metabolismo , Proteínas de Ligação a DNA/metabolismo , Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Ensaio de Imunoadsorção Enzimática , Fatores de Transcrição Forkhead , Humanos , Luciferases/metabolismo , Oligonucleotídeos Antissenso/química , Regiões Promotoras Genéticas , Ligação Proteica , Proteína Quinase C beta , Transdução de Sinais , Proteína Smad6 , Transativadores/metabolismo , Transcrição Gênica , Regulação para Cima
16.
J Biol Chem ; 277(51): 49815-9, 2002 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-12407115

RESUMO

Protein C is a plasma protease that in its active form plays a central role in the regulation of vascular function by modulating thrombosis, inflammation, and apoptosis. A central player in this pathway is the cytokine-regulated receptor thrombomodulin (TM), which functions as a co-factor for the thrombin-dependent generation of activated protein C. We have found that tumor necrosis factor-beta (TGF-beta)-dependent suppression of TM on endothelial cells is differentially regulated by endothelial Smad6s and Smad7. Overexpression of Smad6s resulted in activation of a TGF-beta reporter alone and enhanced TGF-beta response. Moreover, Smad6s overexpression suppressed TM and subsequently reduced activated protein C generation. Antisense inhibition of Smad6s expression enhanced the TM-dependent activation of protein C, whereas blocking the inhibitory Smad7 by antisense resulted in reduced TM-dependent activation of protein C. The effect of Smad6s appeared to be due, at least in part, to up-regulation of TGF-beta itself. Immunohistochemistry studies in normal versus atherosclerotic vessels showed that TM levels were suppressed in the endothelium over plaque. Consistent with the in vitro data, we found differential expression of Smad6s and Smad7 in normal versus atherosclerotic vessels, with Smad6s expression low in normal vessels but elevated in atherosclerotic vessels. In contrast, the opposite expression pattern was observed for Smad7. Overall, our results suggest that the relative balance of these intracellular Smads modulate the balance of endothelial function with regard to protein C activation.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Endotélio Vascular/metabolismo , Linfotoxina-alfa/metabolismo , Proteína C/metabolismo , Trombomodulina/metabolismo , Transativadores/metabolismo , Animais , Arteriosclerose/patologia , Sobrevivência Celular , Cloranfenicol O-Acetiltransferase/metabolismo , Relação Dose-Resposta a Droga , Regulação para Baixo , Endotélio Vascular/citologia , Ensaio de Imunoadsorção Enzimática , Genes Reporter , Humanos , Imuno-Histoquímica , Oligonucleotídeos Antissenso/farmacologia , Transdução de Sinais , Proteína Smad6 , Proteína Smad7 , Fatores de Tempo , Ativação Transcricional , Transfecção , Fator de Crescimento Transformador beta/metabolismo , Regulação para Cima , Xenopus , Proteínas de Xenopus
17.
Proc Natl Acad Sci U S A ; 100(8): 4423-8, 2003 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-12671072

RESUMO

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.


Assuntos
Proteína C/química , Proteína C/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Anticoagulantes/química , Anticoagulantes/metabolismo , Anticoagulantes/farmacologia , Variação Genética , Humanos , Técnicas In Vitro , Cinética , Macaca fascicularis , Modelos Moleculares , Proteína C/genética , Proteína C/farmacologia , Inibidor da Proteína C/farmacologia , Engenharia de Proteínas , Coelhos , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Serpinas/farmacologia , Especificidade por Substrato , alfa 1-Antitripsina/farmacologia
18.
Crit Care Med ; 32(7): 1570-8, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15241104

RESUMO

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.


Assuntos
Proteína C/metabolismo , Sepse/metabolismo , Animais , Biomarcadores , Glicemia , Feminino , Ligadura , Modelos Biológicos , Valor Preditivo dos Testes , Punções , Curva ROC , Ratos , Ratos Sprague-Dawley , Sepse/sangue , Sepse/mortalidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA