Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
3.
Mol Cell Biol ; 33(15): 3050-61, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23732909

RESUMO

NF-κB transcription factors are crucial regulators of inflammation, immunity, stress responses, and cell differentiation. Many studies have demonstrated that ubiquitination of IκB kinase γ (IKKγ), a regulatory subunit of IKK, is instrumental in the activation of IKK and NF-κB. We and others previously identified EGLN3, a member of a family of prolyl hydroxylases, as a negative regulator of the NF-κB pathway. Here we report that EGLN3, but not EGLN1 or -2, interacts with and inhibits K63-linked ubiquitination of IKKγ. The effect appears to be related to inhibition of IKKγ ubiquitination mediated by cIAP1 rather than to stimulation of IKKγ deubiquitination by the deubiquitinases A20 and CYLD (cylindromatosis). EGLN3 does not affect the protein levels of cIAP1 or its E2 ubiquitin-conjugating enzymes UbcH5 and Ubc13. EGLN3 hydroxylase activity is not responsible for its effect on IKKγ ubiquitination and NF-κB signaling. Instead, interaction with IKKγ is required for the ability of EGLN3 to inhibit IKKγ ubiquitination and IKK-NF-κB signaling. EGLN3 competes with cIAP1 for IKKγ binding, leading to inhibition of cIAP1-IKKγ interaction, IKKγ ubiquitination, and IKK-NF-κB signaling. This study provides novel insights into EGLN3 function and sheds new light on the regulation of IKKγ ubiquitination and NF-κB.


Assuntos
Dioxigenases/metabolismo , Quinase I-kappa B/metabolismo , NF-kappa B/metabolismo , Pró-Colágeno-Prolina Dioxigenase/metabolismo , Ubiquitinação , Animais , Linhagem Celular , Humanos , Prolina Dioxigenases do Fator Induzível por Hipóxia , Proteínas Inibidoras de Apoptose/metabolismo , Mapas de Interação de Proteínas , Transdução de Sinais
4.
Arterioscler Thromb Vasc Biol ; 33(1): 4-12, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23139293

RESUMO

OBJECTIVE: Acyl-CoA:cholesterol acyltransferase (ACAT) converts cholesterol to cholesteryl esters in plaque foam cells. Complete deficiency of macrophage ACAT has been shown to increase atherosclerosis in hypercholesterolemic mice because of cytotoxicity from free cholesterol accumulation, whereas we previously showed that partial ACAT inhibition by Fujirebio compound F1394 decreased early atherosclerosis development. In this report, we tested F1394 effects on preestablished, advanced lesions of apolipoprotein-E-deficient mice. METHODS AND RESULTS: Apolipoprotein-E-deficient mice on Western diet for 14 weeks developed advanced plaques, and were either euthanized (Baseline), or continued on Western diet with or without F1394 and euthanized after 14 more weeks. F1394 was not associated with systemic toxicity. Compared with the baseline group, lesion size progressed in both groups; however, F1394 significantly retarded plaque progression and reduced plaque macrophage, free and esterified cholesterol, and tissue factor contents compared with the untreated group. Apoptosis of plaque cells was not increased, consistent with the decrease in lesional free cholesterol. There was no increase in plaque necrosis and unimpaired efferocytosis (phagocytic clearance of apoptotic cells). The effects of F1394 were independent of changes in plasma cholesterol levels. CONCLUSIONS: Partial ACAT inhibition by F1394 lowered plaque cholesterol content and had other antiatherogenic effects in advanced lesions in apolipoprotein-E-deficient mice without overt systemic or plaque toxicity, suggesting the continued potential of ACAT inhibition for the clinical treatment of atherosclerosis, in spite of recent trial data.


Assuntos
Acetil-CoA C-Aciltransferase/antagonistas & inibidores , Aorta/efeitos dos fármacos , Doenças da Aorta/prevenção & controle , Aterosclerose/tratamento farmacológico , Cicloexanos/farmacologia , Dioxanos/farmacologia , Inibidores Enzimáticos/farmacologia , Células Espumosas/efeitos dos fármacos , Acetil-CoA C-Aciltransferase/metabolismo , Animais , Aorta/enzimologia , Aorta/patologia , Doenças da Aorta/sangue , Doenças da Aorta/enzimologia , Doenças da Aorta/genética , Doenças da Aorta/patologia , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Apoptose/efeitos dos fármacos , Aterosclerose/sangue , Aterosclerose/enzimologia , Aterosclerose/genética , Aterosclerose/patologia , Colesterol/sangue , Dieta Aterogênica , Modelos Animais de Doenças , Progressão da Doença , Células Espumosas/enzimologia , Masculino , Camundongos , Camundongos Knockout , Necrose , Placa Aterosclerótica , Tromboplastina/metabolismo
5.
Mol Cell Biol ; 32(18): 3768-75, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22801372

RESUMO

Monocyte chemoattractant protein 1 (MCP-1) plays a pivotal role in many inflammatory processes, including the progression of atherosclerosis and the response of the arterial wall to injury. We previously demonstrated that dexamethasone (Dex) inhibits MCP-1 mRNA accumulation in smooth muscle cells by decreasing its half-life. The effect of Dex was dependent upon the glucocorticoid receptor (GR) and independent of new transcription. Using RNA affinity and column chromatography, we have identified two proteins involved in regulating MCP-1 mRNA stability: Y-box binding protein 1 (YB-1), a multifunctional DNA/RNA-binding protein, and endoribonuclease UK114 (UK). By immunoprecipitation, YB and GR formed a complex present in equal amounts in extracts from untreated and Dex-treated cells. YB-1, UK, and GR small interfering RNA (siRNA) substantially inhibited the effect of Dex on MCP-1 mRNA accumulation. In addition, YB-1 antibody blocked the degradation of MCP-1 mRNA by cytoplasmic extracts from the Dex-treated cells. The degradative activity of extracts immunoprecipitated with antibodies to either YB-1 or GR was blocked with UK antibody. UK did not degrade MCP-1 mRNA; however, upon addition to nondegrading control extracts, it rapidly degraded MCP-1 mRNA. These studies define new roles for GR, YB-1, and UK in the formation of a molecular complex that degrades MCP-1 mRNA.


Assuntos
Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Músculo Liso Vascular/metabolismo , Estabilidade de RNA , Receptores de Glucocorticoides/metabolismo , Ribonucleases/metabolismo , Proteína 1 de Ligação a Y-Box/metabolismo , Animais , Anticorpos , Dexametasona/farmacologia , Masculino , Interferência de RNA , RNA Mensageiro/metabolismo , RNA Interferente Pequeno , Ratos , Ratos Sprague-Dawley , Receptores de Glucocorticoides/genética , Ribonucleases/genética , Proteína 1 de Ligação a Y-Box/genética , Proteína 1 de Ligação a Y-Box/imunologia
6.
Environ Health Perspect ; 118(12): 1692-8, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20822968

RESUMO

BACKGROUND: Diabetes confers an increased risk for cardiovascular effects of airborne particles. OBJECTIVE: We hypothesized that inhalation of elemental carbon ultrafine particles (UFP) would activate blood platelets and vascular endothelium in people with type 2 diabetes. METHODS: In a randomized, double-blind, crossover trial, 19 subjects with type 2 diabetes inhaled filtered air or 50 µg/m³ elemental carbon UFP (count median diameter, 32 nm) by mouthpiece for 2 hr at rest. We repeatedly measured markers of vascular activation, coagulation, and systemic inflammation before and after exposure. RESULTS: Compared with air, particle exposure increased platelet expression of CD40 ligand (CD40L) and the number of platelet-leukocyte conjugates 3.5 hr after exposure. Soluble CD40L decreased with UFP exposure. Plasma von Willebrand factor increased immediately after exposure. There were no effects of particles on plasma tissue factor, coagulation factors VII or IX, or D-dimer. CONCLUSIONS: Inhalation of elemental carbon UFP for 2-hr transiently activated platelets, and possibly the vascular endothelium, in people with type 2 diabetes.


Assuntos
Poluentes Atmosféricos/toxicidade , Vasos Sanguíneos/efeitos dos fármacos , Carbono/toxicidade , Coagulantes/toxicidade , Diabetes Mellitus Tipo 2/fisiopatologia , Material Particulado/toxicidade , Adulto , Fatores Etários , Ligante de CD40/sangue , Estudos Cross-Over , Método Duplo-Cego , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Selectina-P/sangue , Fatores Sexuais , Vasculite Sistêmica/induzido quimicamente , Adulto Jovem , Fator de von Willebrand/metabolismo
7.
Mol Cell Biochem ; 344(1-2): 73-9, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20607592

RESUMO

Monocyte chemoattractant protein-1 (MCP-1) is an inflammatory chemokine that promotes atherosclerosis and is a mediator of the response to arterial injury. We previously demonstrated that platelet-derived growth factor (PDGF) and angiotensin II (Ang) induce the accumulation of MCP-1 mRNA in vascular smooth muscle cells mainly by increasing mRNA stability. In the present study, we have examined the signaling pathways involved in this stabilization of MCP-1 mRNA. The effect of PDGF (BB isoform) and Ang on MCP-1 mRNA stability was mediated by the PDGF ß and angiotensin II receptor AT1R, respectively, and did not involve transactivation between the two receptors. The effect of PDGF-BB was blocked by inhibitors of protein kinase C (PKC), but not by inhibitors of phosphoinositol 3-kinase (PI3K), Src, or NADPH oxidase (NADPHox). In contrast, the effect of Ang was blocked by inhibitors of Src, and PKC, but not by inhibitors of PI3 K, or NADPHox. The effect of PDGF BB on MCP-1 mRNA stability was blocked by siRNA directed against PKCδ and protein kinase D (PKD), whereas the effect of Ang was blocked only by siRNA directed against PKCδ. These results suggest that the enhancement of MCP-1 mRNA stability by PDGF-BB and Ang are mediated by distinct "proximal" signaling pathways that converge on activation of PKCδ. This study identifies a novel role for PKCδ in mediating mRNA stability in smooth muscle cells.


Assuntos
Quimiocina CCL2/genética , Músculo Liso Vascular/enzimologia , Proteína Quinase C-delta/metabolismo , RNA Mensageiro/genética , Animais , Western Blotting , Masculino , Músculo Liso Vascular/citologia , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Ativação Transcricional
8.
Arterioscler Thromb Vasc Biol ; 30(3): 591-8, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20042710

RESUMO

OBJECTIVE: To investigate the presence and role of NF-kappaB proteins in megakaryocytes and platelets. The nuclear factor-kappaB (NF-kappaB) transcription factor family is well known for its role in eliciting inflammation and promoting cell survival. We discovered that human megakaryocytes and platelets express the majority of NF-kappaB family members, including the regulatory inhibitor-kappaB (I-kappaB) and I-kappa kinase (IKK) molecules. METHODS AND RESULTS: Anucleate platelets exposed to NF-kappaB inhibitors demonstrated impaired fundamental functions involved in repairing vascular injury and thrombus formation. Specifically, NF-kappaB inhibition diminished lamellapodia formation, decreased clot retraction times, and reduced thrombus stability. Moreover, inhibition of I-kappaB-alpha phosphorylation (BAY-11-7082) reverted fully spread platelets back to a spheroid morphology. Addition of recombinant IKK-beta or I-kappaB-alpha protein to BAY inhibitor-treated platelets partially restored platelet spreading in I-kappaB-alpha inhibited platelets, and addition of active IKK-beta increased endogenous I-kappaB-alpha phosphorylation levels. CONCLUSIONS: These novel findings support a crucial and nonclassical role for the NF-kappaB family in modulating platelet function and reveal that platelets are sensitive to NF-kappaB inhibitors. As NF-kappaB inhibitors are being developed as antiinflammatory and anticancer agents, they may have unintended effects on platelets. On the basis of these data, NF-kappaB is also identified as a new target to dampen unwanted platelet activation.


Assuntos
Plaquetas/metabolismo , Megacariócitos/metabolismo , NF-kappa B/metabolismo , Adulto , Idoso , Plaquetas/efeitos dos fármacos , Plaquetas/patologia , Adesão Celular/efeitos dos fármacos , Diferenciação Celular , Linhagem Celular Tumoral , Forma Celular/efeitos dos fármacos , Feminino , Sangue Fetal/citologia , Sangue Fetal/metabolismo , Humanos , Leucemia Megacarioblástica Aguda/metabolismo , Leucemia Megacarioblástica Aguda/patologia , Masculino , Megacariócitos/efeitos dos fármacos , Megacariócitos/patologia , Pessoa de Meia-Idade , NF-kappa B/antagonistas & inibidores , Subunidade p50 de NF-kappa B/metabolismo , Nitrilas/farmacologia , Sulfonas/farmacologia , Fator de Transcrição RelA/metabolismo
9.
J Biol Chem ; 285(12): 8927-35, 2010 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-20089853

RESUMO

The egg-laying abnormal-9 (EGLN) prolyl hydroxylases have been shown to regulate the stability and thereby the activity of the alpha subunits of hypoxia-inducible factor (HIF) through its ability to catalyze their hydroxylation. We have previously shown that EGLN3 promotes differentiation of C2C12 skeletal myoblasts. However, the mechanism underlying this effect remains to be fully elucidated. Here, we report that exposure of C2C12 cells to dimethyl oxalylglycine (DMOG), desferrioxamine, and hypoxia, all inhibitors of prolyl hydroxylase activity, led to repression of C2C12 myogenic differentiation. Inactivation of HIF by expression of a HIF dominant-negative mutant or deletion of HIF-1alpha by RNA interference did not affect the inhibitory effect of DMOG, suggesting that the effect of DMOG is HIF-independent. Pharmacologic inactivation of EGLN3 hydroxylase resulted in activation of the canonical NF-kappaB pathway. The inhibitory effect of DMOG on myogenic differentiation was markedly impaired in C2C12 cells expressing a dominant-negative mutant of IkappaBalpha. Exogenous expression of wild-type EGLN3, but not its catalytically inactive mutant, significantly inhibited NF-kappaB activation induced by overexpressed TRAF2 or IkappaB kinase 2. In contrast, deletion of EGLN3 by small interfering RNAs led to activation of NF-kappaB. These data suggest that EGLN3 is a negative regulator of NF-kappaB, and its prolyl hydroxylase activity is required for this effect. Furthermore, wild-type EGLN3, but not its catalytically inactive mutant, potentiated myogenic differentiation. This study demonstrates a novel role for EGLN3 in the regulation of NF-kappaB and suggests that it is involved in mediating myogenic differentiation, which is HIF-independent.


Assuntos
Mioblastos Esqueléticos/citologia , NF-kappa B/metabolismo , Pró-Colágeno-Prolina Dioxigenase/metabolismo , Aminoácidos Dicarboxílicos/química , Animais , Diferenciação Celular , Deleção de Genes , Regulação Enzimológica da Expressão Gênica , Genes Dominantes , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Camundongos , Modelos Biológicos , Mutação , Mioblastos Esqueléticos/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo
10.
J Clin Oncol ; 27(29): 4834-8, 2009 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-19738116

RESUMO

Tissue factor (TF) is a transmembrane glycoprotein that localizes the coagulation serine protease factor VII/VIIa (FVII/VIIa) to the cell surface. The primary function of TF is to activate the clotting cascade. The TF:FVIIa complex also activates cells by cleavage of a G-protein coupled receptor called protease-activated receptor 2 (PAR2). TF is expressed by tumor cells and contributes to a variety of pathologic processes, such as thrombosis, metastasis, tumor growth, and tumor angiogenesis. For instance, tumor cells release TF-positive procoagulant microparticles into the circulation and these may trigger venous thromboembolism in patients with cancer. TF on circulating tumor cells also leads to the coating of the cells with fibrin that traps them within the microvasculature and facilitates hematogenous metastasis. In addition, TF:FVIIa-dependent activation of PAR2 on tumor cells increases tumor growth via an undefined mechanism. One possibility is that PAR2-dependent signaling increases the expression of proangiogenic proteins. Other studies have reported that endothelial cells in the tumor vasculature express TF and this may enhance angiogenesis. These results suggest that inhibition of TF should reduce several pathologic pathways that increase tumor growth and metastasis. This would represent a novel approach to anticancer therapy. Initial studies using inhibitors of the TF:FVIIa complex in mouse tumor models have produced encouraging results. Nevertheless, additional studies are needed to determine if this strategy can be successfully translated to the treatment of cancer patients.


Assuntos
Biomarcadores Tumorais/metabolismo , Neoplasias/metabolismo , Neovascularização Patológica/fisiopatologia , Tromboplastina/metabolismo , Trombose Venosa/metabolismo , Animais , Biomarcadores Tumorais/análise , Modelos Animais de Doenças , Progressão da Doença , Fator VIIa/metabolismo , Feminino , Humanos , Masculino , Camundongos , Invasividade Neoplásica/patologia , Neoplasias/mortalidade , Neovascularização Patológica/metabolismo , Prognóstico , Receptor PAR-2/metabolismo , Estudos de Amostragem , Índice de Gravidade de Doença , Análise de Sobrevida , Tromboplastina/fisiologia , Carga Tumoral , Trombose Venosa/mortalidade , Trombose Venosa/fisiopatologia
11.
Arterioscler Thromb Vasc Biol ; 29(11): 1874-82, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19661482

RESUMO

OBJECTIVE: Platelets are crucial for hemostasis and are vital regulators of inflammation. Foxp3 is a key transcription factor for T regulatory cell development. Humans with IPEX (immune dysregulation, polyendocrinopathy, enteropathy, x-linked) and the scurfy (Foxp3(sf)) mouse have mutations in the Foxp3 gene that lead to a host of pathologies including autoimmunity and skin diseases. Scurfy mice and some humans with IPEX are also thrombocytopenic. The purpose of this study was to determine whether the absence of functional Foxp3 leads to defects in megakaryocytes and platelets. METHODS AND RESULTS: We discovered that human and mouse megakaryocytes express Foxp3 mRNA and protein. Using shRNA and Foxp3(sf) mice, we demonstrated that Foxp3-deficient mouse and human megakaryocyte progenitors exhibited proliferation defects. Striking platelet abnormalities were observed in both an IPEX patient and Foxp3(sf) mice. Impaired platelet spreading and release of TGF-beta and CD40 ligand (CD40L), and abnormal levels of plasma CD40L were observed in a case of IPEX syndrome. Foxp3(sf) mice were thrombocytopenic and had increased platelet volume and altered serum levels of CD40L, TXB(2), and TGF-beta. CONCLUSIONS: These findings provide compelling new evidence that Foxp3 is needed for proper megakaryopoiesis and plays a role in regulating platelet function including spreading and release.


Assuntos
Plaquetas/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Megacariócitos/metabolismo , Trombopoese/fisiologia , Animais , Plaquetas/citologia , Western Blotting , Proliferação de Células , Células Cultivadas , Citometria de Fluxo , Fatores de Transcrição Forkhead/genética , Humanos , Masculino , Megacariócitos/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Probabilidade , RNA Mensageiro/análise , Especificidade da Espécie
12.
J Proteome Res ; 8(5): 2261-72, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19267493

RESUMO

Human platelets play a key role in hemostasis and thrombosis and have recently emerged as key regulators of inflammation. Platelets stored for transfusion produce pro-thrombotic and pro-inflammatory mediators implicated in adverse transfusion reactions. Correspondingly, these mediators are central players in pathological conditions including cardiovascular disease, the major cause of death in diabetics. In view of this, a mass spectrometry based proteomics study was performed on platelets collected from healthy and type-2 diabetics stored for transfusion. Strikingly, our innovative and sensitive proteomic approach identified 122 proteins that were either up- or down-regulated in type-2 diabetics relative to nondiabetic controls and 117 proteins whose abundances changed during a 5-day storage period. Notably, our studies are the first to characterize the proteome of platelets from diabetics before and after storage for transfusion. These identified differences allow us to formulate new hypotheses and experimentation to improve clinical outcomes by targeting "high risk platelets" that render platelet transfusion less effective or even unsafe.


Assuntos
Plaquetas/metabolismo , Preservação de Sangue/métodos , Diabetes Mellitus Tipo 2/sangue , Proteoma/análise , Proteômica/métodos , Adulto , Idoso , Bancos de Sangue , Cromatografia Líquida de Alta Pressão , Feminino , Humanos , Integrina alfa2beta1/sangue , Masculino , Espectrometria de Massas , Pessoa de Meia-Idade , Transfusão de Plaquetas , Proteoma/classificação , Fatores de Tempo , Adulto Jovem
13.
Cancer Invest ; 27(4): 430-4, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19219655

RESUMO

Tissue factor (TF) is the principal physiologic initiator of coagulation. It also plays an important role in tumor growth and metastasis possibly by contributing to angiogenesis. We evaluated the expression of TF in benign and malignant prostate tissue and correlated it with the expression of the pro-angiogenic protein, vascular endothelial growth factor (VEGF). We used a tissue microarray (TMA) constructed from 80 archival prostatectomy specimens. Core samples were collected from benign prostate tissue (BP) (n= 77), high-grade prostatic intraepithelial neoplasia (PIN) (n= 26), and carcinoma (PCa) (n= 93). TMA sections were stained with an immunopurified polyclonal TF antibody and a rabbit polyclonal VEGF. Two pathologists manually scored staining in epithelial cells using the German Immunoreactive Score. Positive staining for TF was seen predominantly in PCa with rare positive glands in BP and PIN. TF expression was significantly lower in BP versus PCa specimens (p< .001) and in PIN versus PCa specimens (p< .001). Positive staining for VEGF was seen in PCa, BP, and PIN. Rates of VEGF expression were also significantly lower in BP versus PCa specimens (p= .003) but not in PCa versus PIN (p= .430). The majority of PCa samples positive for TF were also positive for VEGF (p< .001). Our findings reinforce the link between angiogenesis and TF expression in PCa. We suggest further exploration of TF-mediated pathways leading to increased tumor aggressiveness in PCa, and the possible use of anti-TF agents in PCa.


Assuntos
Carcinoma/química , Neoplasia Prostática Intraepitelial/química , Neoplasias da Próstata/química , Tromboplastina/análise , Análise Serial de Tecidos , Fator A de Crescimento do Endotélio Vascular/análise , Carcinoma/irrigação sanguínea , Carcinoma/patologia , Carcinoma/cirurgia , Humanos , Imuno-Histoquímica , Masculino , Neovascularização Patológica/metabolismo , Prostatectomia , Neoplasia Prostática Intraepitelial/irrigação sanguínea , Neoplasia Prostática Intraepitelial/patologia , Neoplasia Prostática Intraepitelial/cirurgia , Neoplasias da Próstata/irrigação sanguínea , Neoplasias da Próstata/patologia , Neoplasias da Próstata/cirurgia
14.
Immunol Res ; 45(2-3): 251-60, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19184537

RESUMO

The discovery that platelets possess cell membrane, cytoplasmic, and secreted forms of the co-stimulatory molecule CD40 ligand (CD40L, also known as CD154) has led to a revolution in the view of this anucleate, differentiated cell fragment, previously thought only to be involved in blood clotting (hemostasis). During the last decade, it has become clear that platelets function in innate and adaptive immunity and possess pro-inflammatory, as well as pro-thrombotic properties. They interact not only with other platelets and endothelial cells, but also with lymphocytes, dendritic cells, and structural cells such as fibroblasts. Soluble forms of CD40L (sCD40L) in the human circulation are almost entirely derived from platelets. Elevated levels of CD40L are associated with clinically important conditions, such as vascular disease, abnormal clotting (thrombosis), lung injury, and autoimmune disease. Each year millions of platelet transfusions are given to patients that contain large amounts of sCD40L. sCD40L in the supernatant of stored platelets can induce cytokines, chemokines, and lipid mediators by activating CD40 bearing cells. Increased levels of sCD40L in transfused blood are associated with transfusion-related acute lung injury, a potentially fatal complication, as well as more common, milder transfusion reactions such as fever and rigors. These effects come under the rubric of transfusion immunomodulation, which postulates that transfusion recipient biology, particularly immune function, is dramatically altered by transfusion of stored allogeneic blood.


Assuntos
Plaquetas/imunologia , Ligante de CD40/imunologia , Imunomodulação/imunologia , Transfusão de Plaquetas , Imunidade Adaptativa/imunologia , Plaquetas/metabolismo , Antígenos CD40/imunologia , Antígenos CD40/metabolismo , Ligante de CD40/metabolismo , Citocinas/imunologia , Citocinas/metabolismo , Humanos , Imunidade Inata/imunologia
15.
Blood ; 113(3): 705-13, 2009 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-18931346

RESUMO

Tissue factor (TF) initiates coagulation, regulates hemostasis, and plays a critical role in mediating arterial thrombosis. TF is up-regulated in vascular smooth muscle cells (VSMCs) in atherosclerosis and arterial injury. To examine the biologic role of VSMC-derived TF, we crossed TF(flox/flox) mice with SM22alphaCre(+/-) mice. TF mRNA and activity were decreased in the aortic media of TF-deficient mice by 96% and 94.8%, respectively. There were no differences in TF activity measured in plasma or concentrated microparticles. TF-deficient mice were generated with the expected frequency, showed no evidence of bleeding or increased mortality, and had similar activated partial thromboplastin and tail vein bleeding times. Thrombus-mediated flow reduction in response to ferric chloride injury of the carotid arteries was significantly attenuated in VSMC-specific TF-deficient. Stable occlusion was seen in 11 of 12 wild-type mice, but in only 6 of 16 VSMC-specific TF-deficient mice (P = .001). These data suggest that VSMC-derived TF is critical in a macrovascular model of arterial thrombosis. This mouse model should be valuable in determining the contribution of VSMC-derived TF in other TF-mediated phenomena, such as restenosis.


Assuntos
Trombose das Artérias Carótidas/metabolismo , Compostos Férricos/toxicidade , Músculo Liso Vascular/metabolismo , Noxas/toxicidade , Tromboplastina/metabolismo , Animais , Aorta/patologia , Trombose das Artérias Carótidas/patologia , Cloretos , Ensaio de Imunoadsorção Enzimática , Feminino , Fibrose , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Mutantes , Miocárdio/patologia , Miócitos de Músculo Liso/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Tromboplastina/genética , Trombose/patologia
16.
Blood ; 112(10): 4051-60, 2008 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-18755987

RESUMO

Thrombocytopenia is a critical problem that occurs in many hematologic diseases, as well as after cancer therapy and radiation exposure. Platelet transfusion is the most commonly used therapy but has limitations of alloimmunization, availability, and expense. Thus, the development of safe, small, molecules to enhance platelet production would be advantageous for the treatment of thrombocytopenia. Herein, we report that an important lipid mediator and a peroxisome proliferator-activated receptor gamma (PPARgamma) ligand called 15-deoxy-Delta(12,14) prostaglandin J(2) (15d-PGJ(2)), increases Meg-01 maturation and platelet production. 15d-PGJ(2) also promotes platelet formation from culture-derived mouse and human megakaryocytes and accelerates platelet recovery after in vivo radiation-induced bone marrow injury. Interestingly, the platelet-enhancing effects of 15d-PGJ(2) in Meg-01 cells are independent of PPARgamma, but dependent on reactive oxygen species (ROS) accumulation; treatment with antioxidants such as glutathione ethyl ester (GSH-EE); or N-acetylcysteine (NAC) attenuate 15d-PGJ(2)-induced platelet production. Collectively, these data support the concept that megakaryocyte redox status plays an important role in platelet generation and that small electrophilic molecules may have clinical efficacy for improving platelet numbers in thrombocytopenic patients.


Assuntos
Antineoplásicos/farmacologia , Plaquetas/metabolismo , Megacariócitos/metabolismo , Prostaglandina D2/análogos & derivados , Trombocitopenia/metabolismo , Acetilcisteína/farmacologia , Animais , Antineoplásicos/uso terapêutico , Antioxidantes/farmacologia , Medula Óssea/metabolismo , Linhagem Celular , Feminino , Glutationa/análogos & derivados , Glutationa/farmacologia , Humanos , Masculino , Camundongos , PPAR gama/metabolismo , Transfusão de Plaquetas , Prostaglandina D2/farmacologia , Prostaglandina D2/uso terapêutico , Lesões Experimentais por Radiação/metabolismo , Lesões Experimentais por Radiação/terapia , Espécies Reativas de Oxigênio/metabolismo , Trombocitopenia/terapia
17.
Thromb Res ; 122 Suppl 1: S78-81, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18691507

RESUMO

Tissue factor (TF) is a glycoprotein that initiates coagulation, regulates hemostasis and plays a critical role in arterial thrombosis. Vascular smooth muscle cells (SMC) are the major cellular component of the arterial wall. Under normal conditions, SMC express minimal levels of TF; however TF is rapidly induced in SMC by growth factors and cytokines and is expressed in abundance in arterial SMC in response to injury and during atherogenesis. Recent studies have suggested that SMC-derived TF plays an important role in promoting arterial thrombosis and in mediating intimal hyperplasia in response to arterial injury. This latter role may be related to non-procoagulant properties of TF.


Assuntos
Artérias/lesões , Músculo Liso Vascular/fisiologia , Miócitos de Músculo Liso/fisiologia , Tromboplastina/fisiologia , Trombose/fisiopatologia , Animais , Camundongos , Camundongos Knockout , Tromboplastina/deficiência , Trombose/genética
18.
J Vasc Res ; 45(6): 538-46, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18463419

RESUMO

BACKGROUND: Transplant arteriopathy is the leading cause of long term morbidity and mortality following heart transplantation. Animal models have demonstrated that monocyte chemoattractant protein (MCP)-1 is induced early after transplant in cardiac and aortic allografts. We have previously reported that deficiency of MCP-1 or its receptor, CC chemokine receptor 2 (CCR2), is associated with a reduction in intimal proliferation in a mouse femoral artery injury model. Using knockout mice, we have now examined the role of MCP-1 and CCR2 in the development of the intimal proliferation of transplant arteriopathy. METHODS: C57Bl/6 CCR2 and MCP-1 wild-type and knockout mice were used in the studies and aortic transplants were performed between Balb/c mice and C57Bl/6 mice. Aortas from recipient animals were harvested 8 weeks after transplant. RESULTS: Unlike arterial injury, in an aortic transplant model inhibition of MCP-1/CCR2 signaling did not result in reduced intimal proliferation. CONCLUSIONS: Despite a pathology that appears similar, the inflammatory mediators that regulate transplant arteriopathy differ from those regulating intimal proliferation secondary to wire injury. Our results suggest that targeting MCP-1/CCR2 signaling is not sufficient to block transplant arteriopathy across a complete MHC-mismatch barrier.


Assuntos
Aorta/patologia , Aorta/transplante , Doenças da Aorta/patologia , Proliferação de Células , Quimiocina CCL2/metabolismo , Receptores CCR2/metabolismo , Transdução de Sinais , Túnica Íntima/patologia , Animais , Aorta/imunologia , Doenças da Aorta/etiologia , Doenças da Aorta/imunologia , Quimiocina CCL2/deficiência , Quimiocina CCL2/genética , Modelos Animais de Doenças , Hiperplasia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transplante de Órgãos/efeitos adversos , Receptores CCR2/deficiência , Receptores CCR2/genética , Transdução de Sinais/imunologia , Túnica Íntima/imunologia
20.
Thromb Haemost ; 99(1): 86-95, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18217139

RESUMO

Peroxisome proliferator-activated receptor gamma (PPARgamma) and its ligands are important regulators of lipid metabolism, inflammation, and diabetes. We previously demonstrated that anucleate human platelets express the transcription factor PPARgamma and that PPARgamma ligands blunt platelet activation. To further understand the nature of PPARgamma in platelets, we determined the platelet PPARgamma isoform(s) and investigated the fate of PPARgamma following platelet activation. Our studies demonstrated that human platelets contain only the PPARgamma1 isoform and after activation with thrombin, TRAP, ADP or collagen PPARgamma is released from internal stores. PPARgamma release was blocked by a cytoskeleton inhibitor, Latrunculin A. Platelet-released PPARgamma was complexed with the retinoid X receptor (RXR) and retained its ability to bind DNA. Interestingly, the released PPARgamma and RXR were microparticle associated and the released PPARgamma/RXR complex retained DNA-binding ability. Additionally, a monocytic cell line, THP-1, is capable of internalizing PMPs. Further investigation following treatment of these cells with the PPARgamma agonist, rosiglitazone and PMPs revealed a possible transcellular mechanism to attenuate THP-1 activation. These new findings are the first to demonstrate transcription factor release from platelets, revealing the complex spectrum of proteins expressed and expelled from platelets, and suggests that platelet PPARgamma has an undiscovered role in human biology.


Assuntos
Plaquetas/metabolismo , PPAR gama/metabolismo , Ativação Plaquetária , Receptores X de Retinoides/metabolismo , Vesículas Transportadoras/metabolismo , Adulto , Plaquetas/efeitos dos fármacos , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Linhagem Celular Tumoral , Colágeno/metabolismo , DNA/metabolismo , Dimerização , Feminino , Humanos , Masculino , Megacariócitos/metabolismo , Pessoa de Meia-Idade , Monócitos/metabolismo , PPAR gama/agonistas , Fragmentos de Peptídeos/metabolismo , Ligação Proteica , Isoformas de Proteínas/metabolismo , RNA Mensageiro/metabolismo , Receptor X Retinoide alfa/metabolismo , Receptor X Retinoide beta/metabolismo , Receptores X de Retinoides/genética , Rosiglitazona , Tiazolidinedionas/farmacologia , Tiazolidinas/farmacologia , Trombina/metabolismo , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...