Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Shock ; 54(2): 245-255, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-31490354

RESUMO

Mortalin/GRP75 (glucose regulated protein 75), a member of heat shock protein 70 family of chaperones, is involved in several cellular processes including proliferation and signaling, and plays a pivotal role in cancer and neurodegenerative disorders. In this study, we sought to determine the role of mortalin/GRP75 in mediating vascular inflammation and permeability linked to the pathogenesis of acute lung injury (ALI). In an aerosolized bacterial lipopolysaccharide inhalation mouse model of ALI, we found that administration of mortalin/GRP75 inhibitor mean kinetic temperature-077, both prophylactically and therapeutically, protected against polymorphonuclear leukocytes influx into alveolar airspaces, microvascular leakage, and expression of pro-inflammatory mediators such as interleukin-1ß, E-selectin, and tumor necrosis factor TNFα. Consistent with this, thrombin-induced inflammation in cultured human endothelial cells (EC) was also protected upon before and after treatment with mean kinetic temperature-077. Similar to pharmacological inhibition of mortalin/GRP75, siRNA-mediated depletion of mortalin/GRP75 also blocked thrombin-induced expression of proinflammatory mediators such as intercellular adhesion molecule-1 and vascular adhesion molecule-1. Mechanistic analysis in EC revealed that inactivation of mortalin/GRP75 interfered with the binding of the liberated NF-κB to the DNA, thereby leading to inhibition of downstream expression of adhesion molecules, cytokines, and chemokines. Importantly, thrombin-induced Ca signaling and EC permeability were also prevented upon mortalin/GRP75 inactivation/depletion. Thus, this study provides evidence for a novel role of mortalin/GRP75 in mediating EC inflammation and permeability associated with ALI.


Assuntos
Lesão Pulmonar Aguda/tratamento farmacológico , Lesão Pulmonar Aguda/metabolismo , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Proteínas de Choque Térmico HSP70/metabolismo , Proteínas de Membrana/metabolismo , Animais , Ensaio de Imunoadsorção Enzimática , Imunofluorescência , Proteínas de Choque Térmico HSP70/antagonistas & inibidores , Humanos , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Masculino , Proteínas de Membrana/antagonistas & inibidores , Camundongos , Camundongos Endogâmicos C57BL , Ligação Proteica/efeitos dos fármacos , Piridinas/uso terapêutico , Transdução de Sinais/efeitos dos fármacos , Tiazóis/uso terapêutico
2.
Cell Signal ; 61: 120-129, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31054328

RESUMO

Recent studies have implicated autophagy in several inflammatory diseases involving aberrant endothelial cell (EC) responses, such as acute lung injury (ALI). However, the mechanistic basis for a role of autophagy in EC inflammation and permeability remain poorly understood. In this study, we impaired autophagy by silencing the essential Beclin1 autophagy gene in human pulmonary artery EC. This resulted in reduced expression of proinflammatory genes in response to thrombin, a procoagulant and proinflammatory mediator whose concentration is elevated in many diseases including sepsis and ALI. These (Beclin1-depleted) cells also displayed a marked decrease in NF-κB activity secondary to impaired DNA binding of RelA/p65 in the nucleus, but exhibited normal IκBα degradation in the cytosol. Further analysis showed that Beclin1 knockdown was associated with impaired RelA/p65 translocation to the nucleus. Additionally, Beclin1 knockdown attenuated thrombin-induced phosphorylation of RelA/p65 at Ser536, a critical event necessary for the transcriptional activity of RelA/p65. Beclin1 silencing also protected against thrombin-induced EC barrier disruption by preventing the loss of VE-cadherin at adherens junctions. Moreover, Beclin1 knockdown reduced thrombin-induced phosphorylation/inactivation of actin depolymerizing protein Cofilin1 and thereby actin stress fiber formation required for EC permeability as well as RelA/p65 nuclear translocation. Together, these data identify Beclin1 as a novel mechanistic link between autophagy and EC dysfunction (inflammation and permeability).


Assuntos
Junções Aderentes/metabolismo , Autofagia/genética , Proteína Beclina-1/metabolismo , Células Endoteliais/metabolismo , Fator de Transcrição RelA/metabolismo , Autofagia/efeitos dos fármacos , Proteína Beclina-1/genética , Núcleo Celular/metabolismo , Células Cultivadas , Cofilina 1/metabolismo , DNA/metabolismo , Técnicas de Silenciamento de Genes , Humanos , Inflamação/metabolismo , Inibidor de NF-kappaB alfa/metabolismo , Fosforilação/efeitos dos fármacos , Fosforilação/genética , Artéria Pulmonar/citologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Trombina/farmacologia , Transfecção
3.
Cell Signal ; 44: 103-117, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29331583

RESUMO

Nucleocytoplasmic shuttling via importins is central to the function of eukaryotic cells and an integral part of the processes that lead to many human diseases. In this study, we addressed the role of α and ß importins in the mechanism of endothelial cell (EC) inflammation and permeability, important pathogenic features of many inflammatory diseases such as acute lung injury and atherosclerosis. RNAi-mediated knockdown of importin α4 or α3 each inhibited NF-κB activation, proinflammatory gene (ICAM-1, VCAM-1, and IL-6) expression, and thereby endothelial adhesivity towards HL-60 cells, upon thrombin challenge. The inhibitory effect of α4 and α3 knockdown was associated with impaired nuclear import and consequently, DNA binding of RelA/p65 subunit of NF-κB and occurred independently of IκBα degradation. Intriguingly, knockdown of importins α4 and α3 also inhibited thrombin-induced RelA/p65 phosphorylation at Ser536, showing a novel role of α importins in regulating transcriptional activity of RelA/p65. Similarly, knockdown of importin ß1, but not ß2, blocked thrombin-induced activation of RelA/p65 and its target genes. In parallel studies, TNFα-mediated inflammatory responses in EC were refractory to knockdown of importins α4, α3 or ß1, indicating a stimulus-specific regulation of RelA/p65 and EC inflammation by these importins. Importantly, α4, α3, or ß1 knockdown also protected against thrombin-induced EC barrier disruption by inhibiting the loss of VE-cadherin at adherens junctions and by regulating actin cytoskeletal rearrangement. These results identify α4, α3 and ß1 as critical mediators of EC inflammation and permeability associated with intravascular coagulation.


Assuntos
Inflamação/metabolismo , Subunidade p50 de NF-kappa B/metabolismo , alfa Carioferinas/fisiologia , beta Carioferinas/fisiologia , Antígenos CD/metabolismo , Caderinas/metabolismo , Permeabilidade da Membrana Celular , Células Cultivadas , Células Endoteliais/metabolismo , Regulação da Expressão Gênica , Humanos , Molécula 1 de Adesão Intercelular/metabolismo , Interleucina-6/metabolismo , Fosforilação , Transdução de Sinais , Fator de Transcrição RelA/metabolismo , Molécula 1 de Adesão de Célula Vascular/metabolismo , alfa Carioferinas/genética , beta Carioferinas/genética
4.
Am J Respir Cell Mol Biol ; 55(5): 722-735, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27374190

RESUMO

Administration of supplemental oxygen remains a critical clinical intervention for survival of preterm infants with respiratory failure. However, prolonged exposure to hyperoxia can augment pulmonary damage, resulting in developmental lung diseases embodied as hyperoxia-induced acute lung injury and bronchopulmonary dysplasia (BPD). We sought to investigate the role of autophagy in hyperoxia-induced apoptotic cell death in developing lungs. We identified increased autophagy signaling in hyperoxia-exposed mouse lung epithelial-12 cells, freshly isolated fetal type II alveolar epithelial cells, lungs of newborn wild-type mice, and human newborns with respiratory distress syndrome and evolving and established BPD. We found that hyperoxia exposure induces autophagy in a Trp53-dependent manner in mouse lung epithelial-12 cells and in neonatal mouse lungs. Using pharmacological inhibitors and gene silencing techniques, we found that the activation of autophagy, upon hyperoxia exposure, demonstrated a protective role with an antiapoptotic response. Specifically, inhibiting regulatory-associated protein of mechanistic target of rapamycin (RPTOR) in hyperoxia settings, as evidenced by wild-type mice treated with torin2 or mice administered (Rptor) silencing RNA via intranasal delivery or Rptor+/-, limited lung injury by increased autophagy, decreased apoptosis, improved lung architecture, and increased survival. Furthermore, we identified increased protein expression of phospho-beclin1, light chain-3-II and lysosomal-associated membrane protein 1, suggesting altered autophagic flux in the lungs of human neonates with established BPD. Collectively, our study unveils a novel demonstration of enhancing autophagy and antiapoptotic effects, specifically through the inhibition of RPTOR as a potentially useful therapeutic target for the treatment of hyperoxia-induced acute lung injury and BPD in developing lungs.


Assuntos
Lesão Pulmonar Aguda/etiologia , Lesão Pulmonar Aguda/patologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Apoptose , Autofagia , Hiperóxia/complicações , Hiperóxia/patologia , Lesão Pulmonar Aguda/metabolismo , Células Epiteliais Alveolares/metabolismo , Animais , Animais Recém-Nascidos , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Displasia Broncopulmonar/complicações , Displasia Broncopulmonar/metabolismo , Displasia Broncopulmonar/patologia , Linhagem Celular , Feminino , Humanos , Hiperóxia/metabolismo , Hipertensão Pulmonar/complicações , Hipertensão Pulmonar/patologia , Hipertrofia Ventricular Direita/complicações , Hipertrofia Ventricular Direita/patologia , Recém-Nascido , Pulmão/metabolismo , Pulmão/patologia , Camundongos , Proteínas Associadas aos Microtúbulos/metabolismo , Naftiridinas/farmacologia , Fenótipo , Proteína Regulatória Associada a mTOR , Fatores de Tempo , Proteína Supressora de Tumor p53/metabolismo
5.
Am J Physiol Lung Cell Mol Physiol ; 311(2): L517-24, 2016 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-27371732

RESUMO

Phospholipase C-ε (PLC-ε) is a unique PLC isoform that can be regulated by multiple signaling inputs from both Ras family GTPases and heterotrimeric G proteins and has primary sites of expression in the heart and lung. Whereas the role of PLC-ε in cardiac function and pathology has been documented, its relevance in acute lung injury (ALI) is unclear. We used PLC-ε(-/-) mice to address the role of PLC-ε in regulating lung vascular inflammation and injury in an aerosolized bacterial LPS inhalation mouse model of ALI. PLC-ε(-/-) mice showed a marked decrease in LPS-induced proinflammatory mediators (ICAM-1, VCAM-1, TNF-α, IL-1ß, IL-6, macrophage inflammatory protein 2, keratinocyte-derived cytokine, monocyte chemoattractant protein 1, and granulocyte-macrophage colony-stimulating factor), lung neutrophil infiltration and microvascular leakage, and loss of VE-cadherin compared with PLC-ε(+/+) mice. These data identify PLC-ε as a critical determinant of proinflammatory and leaky phenotype of the lung. To test the possibility that PLC-ε activity in endothelial cells (EC) could contribute to ALI, we determined its role in EC inflammation and barrier disruption. RNAi knockdown of PLC-ε inhibited NF-κB activity in response to diverse proinflammatory stimuli, thrombin, LPS, TNF-α, and the nonreceptor agonist phorbol 13-myristate 12-acetate (phorbol esters) in EC. Depletion of PLC-ε also inhibited thrombin-induced expression of NF-κB target gene, VCAM-1. Importantly, PLC-ε knockdown also protected against thrombin-induced EC barrier disruption by inhibiting the loss of VE-cadherin at adherens junctions and formation of actin stress fibers. These data identify PLC-ε as a novel regulator of EC inflammation and permeability and show a hitherto unknown role of PLC-ε in the pathogenesis of ALI.


Assuntos
Lesão Pulmonar Aguda/enzimologia , Fosfoinositídeo Fosfolipase C/fisiologia , Lesão Pulmonar Aguda/imunologia , Lesão Pulmonar Aguda/patologia , Animais , Antígenos CD/metabolismo , Caderinas/metabolismo , Permeabilidade Capilar , Células Cultivadas , Endotélio Vascular/enzimologia , Endotélio Vascular/patologia , Humanos , Pulmão/irrigação sanguínea , Pulmão/patologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , NF-kappa B/metabolismo , Transdução de Sinais , Fibras de Estresse/metabolismo , Vasculite/enzimologia
6.
Am J Physiol Lung Cell Mol Physiol ; 309(3): L293-304, 2015 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-26024894

RESUMO

Chronic obstructive pulmonary disease (COPD) is a highly prevalent, chronic inflammatory lung disease with limited existing therapeutic options. While modulation of peroxisome proliferator-activating receptor (PPAR)-γ activity can modify inflammatory responses in several models of lung injury, the relevance of the PPARG pathway in COPD pathogenesis has not been previously explored. Mice lacking Pparg specifically in airway epithelial cells displayed increased susceptibility to chronic cigarette smoke (CS)-induced emphysema, with excessive macrophage accumulation associated with increased expression of chemokines, Ccl5, Cxcl10, and Cxcl15. Conversely, treatment of mice with a pharmacological PPARγ activator attenuated Cxcl10 and Cxcl15 expression and macrophage accumulation in response to CS. In vitro, CS increased lung epithelial cell chemokine expression in a PPARγ activation-dependent fashion. The ability of PPARγ to regulate CS-induced chemokine expression in vitro was not specifically associated with peroxisome proliferator response element (PPRE)-mediated transactivation activity but was correlated with PPARγ-mediated transrepression of NF-κB activity. Pharmacological or genetic activation of PPARγ activity abrogated CS-dependent induction of NF-κB activity. Regulation of NF-κB activity involved direct PPARγ-NF-κB interaction and PPARγ-mediated effects on IKK activation, IκBα degradation, and nuclear translocation of p65. Our data indicate that PPARG represents a disease-relevant pathophysiological and pharmacological target in COPD. Its activation state likely contributes to NF-κB-dependent, CS-induced chemokine-mediated regulation of inflammatory cell accumulation.


Assuntos
Quimiocinas/metabolismo , PPAR gama/fisiologia , Enfisema Pulmonar/metabolismo , Fumar/efeitos adversos , Animais , Linhagem Celular , Suscetibilidade a Doenças , Feminino , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Enfisema Pulmonar/etiologia , Enfisema Pulmonar/imunologia , Transdução de Sinais , Fumar/imunologia , Fumar/metabolismo , Ativação Transcricional
7.
Shock ; 42(6): 562-9, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25057925

RESUMO

We addressed the role of transglutaminase 2 (TG2), a calcium-dependent enzyme that catalyzes cross-linking of proteins, in the mechanism of endothelial cell (EC) inflammation and lung polymorphonuclear lymphocyte (PMN) infiltration. Exposure of EC to thrombin, a procoagulant and proinflammatory mediator, resulted in activation of the transcription factor nuclear factor κB (NF-κB) and its target genes, vascular cell adhesion molecule 1, monocyte chemotactic protein 1, and interleukin 6. RNAi knockdown of TG2 inhibited these responses. Analysis of NF-κB activation pathway showed that TG2 knockdown was associated with inhibition of thrombin-induced DNA binding as well as serine phosphorylation of RelA/p65, a crucial event that controls transcriptional capacity of the DNA-bound RelA/p65. These results implicate an important role for TG2 in mediating EC inflammation by promoting DNA-binding and transcriptional activity of RelA/p65. Because thrombin is released in high amounts during sepsis, and its concentration is elevated in plasma and lavage fluids of patients with acute respiratory distress syndrome, we determined the in vivo relevance of TG2 in a mouse model of sepsis-induced lung PMN recruitment. A marked reduction in NF-κB activation, adhesion molecule expression, and lung PMN sequestration was observed in TG2 knockout mice compared with wild-type mice exposed to endotoxemia. Together, these results identify TG2 as an important mediator of EC inflammation and lung PMN sequestration associated with intravascular coagulation and sepsis.


Assuntos
Proteínas de Ligação ao GTP/metabolismo , Regulação Enzimológica da Expressão Gênica , Inflamação/metabolismo , Neutrófilos/citologia , Transglutaminases/metabolismo , Animais , Adesão Celular , Células Cultivadas , Células Endoteliais/enzimologia , Endotoxemia/metabolismo , Fibrose/fisiopatologia , Humanos , Pulmão/metabolismo , Linfócitos/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , NF-kappa B/metabolismo , Fosforilação , Proteína 2 Glutamina gama-Glutamiltransferase , Artéria Pulmonar/metabolismo , Trombina/metabolismo , Fator de Transcrição RelA/metabolismo
8.
Am J Physiol Lung Cell Mol Physiol ; 305(9): L651-64, 2013 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-24039253

RESUMO

Endothelial cell (EC) inflammation is a central event in the pathogenesis of many pulmonary diseases such as acute lung injury and its more severe form acute respiratory distress syndrome. Alterations in actin cytoskeleton are shown to be crucial for NF-κB regulation and EC inflammation. Previously, we have described a role of actin binding protein cofilin in mediating cytoskeletal alterations essential for NF-κB activation and EC inflammation. The present study describes a dynamic mechanism in which LIM kinase 1 (LIMK1), a cofilin kinase, and slingshot-1Long (SSH-1L), a cofilin phosphatase, are engaged by procoagulant and proinflammatory mediator thrombin to regulate these responses. Our data show that knockdown of LIMK1 destabilizes whereas knockdown of SSH-1L stabilizes the actin filaments through modulation of cofilin phosphorylation; however, in either case thrombin-induced NF-κB activity and expression of its target genes (ICAM-1 and VCAM-1) is inhibited. Further mechanistic analyses reveal that knockdown of LIMK1 or SSH-1L each attenuates nuclear translocation and thereby DNA binding of RelA/p65. In addition, LIMK1 or SSH-1L depletion inhibited RelA/p65 phosphorylation at Ser(536), a critical event conferring transcriptional competency to the bound NF-κB. However, unlike SSH-1L, LIMK1 knockdown also impairs the release of RelA/p65 by blocking IKKß-dependent phosphorylation/degradation of IκBα. Interestingly, LIMK1 or SSH-1L depletion failed to inhibit TNF-α-induced RelA/p65 nuclear translocation and proinflammatory gene expression. Thus this study provides evidence for a novel role of LIMK1 and SSH-1L in selectively regulating EC inflammation associated with intravascular coagulation.


Assuntos
Células Endoteliais/metabolismo , Quinases Lim/metabolismo , Fosfoproteínas Fosfatases/metabolismo , Pneumonia/metabolismo , Trombina/metabolismo , Fator de Transcrição RelA/metabolismo , Linhagem Celular , Cofilina 1/metabolismo , Coagulação Intravascular Disseminada/imunologia , Coagulação Intravascular Disseminada/metabolismo , Células Endoteliais/citologia , Células Endoteliais/imunologia , Técnicas de Silenciamento de Genes , Humanos , Quinase I-kappa B/metabolismo , Quinases Lim/genética , NF-kappa B/metabolismo , Fosfoproteínas Fosfatases/genética , Fosforilação/fisiologia , Pneumonia/imunologia , Artéria Pulmonar/citologia , Artéria Pulmonar/imunologia , Vasculite/imunologia , Vasculite/metabolismo
9.
PLoS One ; 8(3): e59965, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23555849

RESUMO

The pathogenesis of acute lung injury (ALI) involves bidirectional cooperation and close interaction between inflammatory and coagulation pathways. A key molecule linking coagulation and inflammation is the procoagulant thrombin, a serine protease whose concentration is elevated in plasma and lavage fluids of patients with ALI and acute respiratory distress syndrome (ARDS). However, little is known about the mechanism by which thrombin contributes to lung inflammatory response. In this study, we developed a new mouse model that permits investigation of lung inflammation associated with intravascular coagulation. Using this mouse model and in vitro approaches, we addressed the role of non-muscle myosin light chain kinase (nmMLCK) in thrombin-induced endothelial cell (EC) inflammation and lung neutrophil (PMN) infiltration. Our in vitro experiments revealed a key role of nmMLCK in ICAM-1 expression by its ability to control nuclear translocation and transcriptional capacity of RelA/p65 in EC. When subjected to intraperitoneal thrombin challenge, wild type mice showed a marked increase in lung PMN infiltration via expression of ICAM-1. However, these responses were markedly attenuated in mice deficient in nmMLCK. These results provide mechanistic insight into lung inflammatory response associated with intravascular coagulation and identify nmMLCK as a critical target for modulation of lung inflammation.


Assuntos
Células Endoteliais/citologia , Regulação da Expressão Gênica , Leucócitos Mononucleares/citologia , Pulmão/metabolismo , Quinase de Cadeia Leve de Miosina/fisiologia , Trombina/metabolismo , Animais , Coagulação Sanguínea , Núcleo Celular/metabolismo , Células Endoteliais/metabolismo , Feminino , Células Endoteliais da Veia Umbilical Humana , Humanos , Inflamação , Molécula 1 de Adesão Intercelular/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neutrófilos/metabolismo , Peroxidase/metabolismo , Fator de Transcrição RelA/metabolismo
10.
Am J Respir Cell Mol Biol ; 47(5): 660-8, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22842493

RESUMO

We investigated the role of proline-rich tyrosine kinase 2 (Pyk2) in the mechanism of NF-κB activation and endothelial cell (EC) inflammation induced by thrombin, a procoagulant serine protease released in high amounts during sepsis and other inflammatory conditions. Stimulation of ECs with thrombin resulted in a time-dependent activation of Pyk2. RNA interference knockdown of Pyk2 attenuated thrombin-induced activity of NF-κB and expression of its target genes, vascular cell adhesion molecule-1 and monocyte chemoattractant protein-1. Pyk2 knockdown impaired thrombin-induced activation of IκB kinase (IKK) and phosphorylation (Ser32 and Ser36) of IkappaBα, but, surprisingly, failed to prevent IκBα degradation. However, depletion of IKKα or IKKß was effective in inhibiting IκBα phosphorylation/degradation, as expected. Intriguingly, Pyk2 knockdown impaired nuclear translocation and DNA binding of RelA/p65, despite the inability to prevent IκBα degradation. In addition, Pyk2 knockdown was associated with inhibition of RelA/p65 phosphorylation at Ser536, which is important for transcriptional activity of RelA/p65. Depletion of IKKα or IKKß each impaired RelA/p65 phosphorylation. Taken together, these data identify Pyk2 as a critical regulator of EC inflammation by virtue of engaging IKK to promote the release and the transcriptional capacity of RelA/p65, and, additionally, by its ability to facilitate the nuclear translocation of the released RelA/p65. Thus, specific targeting of Pyk2 may be an effective anti-inflammatory strategy in vascular diseases associated with EC inflammation and intravascular coagulation.


Assuntos
Células Endoteliais/enzimologia , Quinase 2 de Adesão Focal/metabolismo , Fator de Transcrição RelA/metabolismo , Transporte Ativo do Núcleo Celular , Células Cultivadas , Células Endoteliais/imunologia , Endotélio Vascular/patologia , Ativação Enzimática , Quinase 2 de Adesão Focal/genética , Técnicas de Silenciamento de Genes , Humanos , Quinase I-kappa B/genética , Quinase I-kappa B/metabolismo , Mediadores da Inflamação/metabolismo , Fosforilação , Ligação Proteica , Processamento de Proteína Pós-Traducional , Proteólise , Artéria Pulmonar/patologia , Interferência de RNA , Trombina/fisiologia , Ativação Transcricional , Molécula 1 de Adesão de Célula Vascular/metabolismo , Vasculite/genética , Vasculite/metabolismo
11.
Cancer Res ; 71(16): 5558-68, 2011 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-21724588

RESUMO

Angiogenesis is a critical step during cancer progression. The VEGF is a major stimulator for angiogenesis and is predominantly contributed by cancer cells in tumors. Inhibition of the VEGF signaling pathway has shown promising therapeutic benefits for cancer patients, but adaptive tumor responses are often observed, indicating the need for further understanding of VEGF regulation. We report that a novel G protein-coupled receptor, GPR56, inhibits VEGF production from the melanoma cell lines and impedes melanoma angiogenesis and growth, through the serine threonine proline-rich segment in its N-terminus and a signaling pathway involving protein kinase Cα. We also present evidence that the two fragments of GPR56, which are generated by autocatalyzed cleavage, played distinct roles in regulating VEGF production and melanoma progression. Finally, consistent with its suppressive roles in melanoma progression, the expression levels of GPR56 are inversely correlated with the malignancy of melanomas in human subjects. We propose that components of the GPR56-mediated signaling pathway may serve as new targets for antiangiogenic treatment of melanoma.


Assuntos
Melanoma/patologia , Neovascularização Patológica/fisiopatologia , Receptores Acoplados a Proteínas G/fisiologia , Fator A de Crescimento do Endotélio Vascular/biossíntese , Animais , Linhagem Celular Tumoral , Ensaio de Imunoadsorção Enzimática , Humanos , Melanoma/irrigação sanguínea , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID
12.
Antioxid Redox Signal ; 12(12): 1355-69, 2010 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-19929443

RESUMO

VEGF receptor 2 (VEGFR2), a tyrosine kinase receptor, is activated by VEGF and fluid shear stress (FSS), and its downstream signaling is important in the regulation of endothelial functions, such as cell migration, endothelium-dependent relaxation, and angiogenesis. Cigarette smoke (CS) is known to cause oxidative/nitrosative stress, leading to modifications of tyrosine kinase receptors and impaired downstream signaling. We hypothesized that CS-induced oxidative/nitrosative stress impairs VEGF- and FSS-mediated VEGFR2 activation, leading to endothelial dysfunction. Human lung microvascular endothelial cells and human umbilical vein endothelial cells were treated with different concentrations of cigarette smoke extract (CSE) to investigate the VEGF- or FSS-mediated VEGFR2 phosphorylation and its downstream signaling involved in endothelial function. CSE treatment impaired both VEGF- and FSS-mediated VEGFR2 phosphorylation, resulting in impaired endothelial nitric oxide synthase (eNOS) phosphorylation by Akt. CS-derived reactive oxygen/nitrogen species react with VEGFR2, rendering VEGFR2 inactive for its downstream signaling. Pretreatment with nitric oxide scavenger (PTIO), reactive oxygen species scavengers (combination of SOD with catalase), and N-acetyl-L-cysteine, significantly attenuated the CSE-induced impairment of VEGF-mediated Akt and eNOS phosphorylation. These findings suggest that CSE-induced oxidative/nitrosative stress impairs VEGF- and FSS-mediated endothelial cell function and has important implications in the pathogenesis of CS-induced pulmonary and cardiovascular diseases associated with endothelial dysfunction.


Assuntos
Células Endoteliais/efeitos dos fármacos , Sequestradores de Radicais Livres/farmacologia , Nicotiana , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Espécies Reativas de Nitrogênio/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Reologia , Transdução de Sinais/efeitos dos fármacos , Fumaça/efeitos adversos , Estresse Mecânico , Fator A de Crescimento do Endotélio Vascular/fisiologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/fisiologia , Acetilcisteína/farmacologia , Catalase/farmacologia , Células Cultivadas/efeitos dos fármacos , Células Cultivadas/metabolismo , Óxidos N-Cíclicos/farmacologia , Células Endoteliais/metabolismo , Humanos , Imidazóis/farmacologia , Pulmão/irrigação sanguínea , Oxirredução , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/fisiologia , Transdução de Sinais/fisiologia , Superóxido Dismutase/farmacologia , Veias Umbilicais , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/efeitos dos fármacos
13.
J Biol Chem ; 284(31): 21047-56, 2009 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-19483084

RESUMO

Activation of RhoA/Rho-associated kinase (ROCK) pathway and the associated changes in actin cytoskeleton induced by thrombin are crucial for activation of NF-kappaB and expression of its target gene ICAM-1 in endothelial cells. However, the events acting downstream of RhoA/ROCK to mediate these responses remain unclear. Here, we show a central role of cofilin-1, an actin-binding protein that promotes actin depolymerization, in linking RhoA/ROCK pathway to dynamic alterations in actin cytoskeleton that are necessary for activation of NF-kappaB and thereby expression of ICAM-1 in these cells. Stimulation of human umbilical vein endothelial cells with thrombin resulted in Ser(3) phosphorylation/inactivation of cofilin and formation of actin stress fibers in a ROCK-dependent manner. RNA interference knockdown of cofilin-1 stabilized the actin filaments and inhibited thrombin- and RhoA-induced NF-kappaB activity. Similarly, constitutively inactive mutant of cofilin-1 (Cof1-S3D), known to stabilize the actin cytoskeleton, inhibited NF-kappaB activity by thrombin. Overexpression of wild type cofilin-1 or constitutively active cofilin-1 mutant (Cof1-S3A), known to destabilize the actin cytoskeleton, also impaired thrombin-induced NF-kappaB activity. Additionally, depletion of cofilin-1 was associated with a marked reduction in ICAM-1 expression induced by thrombin. The effect of cofilin-1 depletion on NF-kappaB activity and ICAM-1 expression occurred downstream of IkappaBalpha degradation and was a result of impaired RelA/p65 nuclear translocation and consequently, RelA/p65 binding to DNA. Together, these data show that cofilin-1 occupies a central position in RhoA-actin pathway mediating nuclear translocation of RelA/p65 and expression of ICAM-1 in endothelial cells.


Assuntos
Núcleo Celular/metabolismo , Cofilina 1/metabolismo , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Molécula 1 de Adesão Intercelular/metabolismo , Trombina/farmacologia , Fator de Transcrição RelA/metabolismo , Núcleo Celular/efeitos dos fármacos , Células Endoteliais/citologia , Células Endoteliais/enzimologia , Técnicas de Silenciamento de Genes , Humanos , Proteínas I-kappa B/metabolismo , Molécula 1 de Adesão Intercelular/genética , Modelos Biológicos , Inibidor de NF-kappaB alfa , Fosforilação/efeitos dos fármacos , Regiões Promotoras Genéticas/genética , Inibidores de Proteínas Quinases/farmacologia , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Interferência de RNA/efeitos dos fármacos , Fibras de Estresse/efeitos dos fármacos , Fibras de Estresse/metabolismo , Quinases Associadas a rho/antagonistas & inibidores , Proteína rhoA de Ligação ao GTP/metabolismo
14.
Antioxid Redox Signal ; 11(4): 823-39, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18808323

RESUMO

Stable adhesion of leukocytes to endothelium is crucial for transendothelial migration (TEM) of leukocytes evoked during inflammatory responses, immune surveillance, and homing and mobilization of hematopoietic progenitor cells. The basis of stable adhesion involves expression of intercellular adhesion molecule-1 (ICAM-1), an inducible endothelial adhesive protein that serves as a counter-receptor for beta(2)-integrins on leukocytes. Interaction of ICAM-1 with beta(2)-integrins enables leukocytes to adhere firmly to the vascular endothelium and subsequently, to migrate across the endothelial barrier. The emerging paradigm is that ICAM-1, in addition to firmly capturing leukocytes, triggers intracellular signaling events that may contribute to active participation of the endothelium in facilitating the TEM of adherent leukocytes. The nature, duration, and intensity of ICAM-1-dependent signaling events may contribute to the determination of the route (paracellular vs. transcellular) of leukocyte passage; these aspects of ICAM-1 signaling may in turn be influenced by density and distribution of ICAM-1 on the endothelial cell surface, the source of endothelial cells it is present on, and the type of leukocytes with which it is engaged. This review summarizes our current understanding of the "ICAM-1 paradigm" of TEM with an emphasis on the signaling events mediating ICAM-1 expression and activated by ICAM-1 engagement in endothelial cells.


Assuntos
Movimento Celular/fisiologia , Molécula 1 de Adesão Intercelular/fisiologia , Leucócitos/citologia , Animais , GTP Fosfo-Hidrolases/metabolismo , Humanos , Molécula 1 de Adesão Intercelular/metabolismo , Leucócitos/metabolismo , Oxidantes/metabolismo , Proteínas Quinases/metabolismo , Transdução de Sinais
15.
J Biol Chem ; 284(7): 4052-61, 2009 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-19074768

RESUMO

We have shown that the mammalian target of rapamycin (mTOR) down-regulates thrombin-induced ICAM-1 expression in endothelial cells by suppressing the activation of NF-kappaB. However, the mechanisms by which mTOR is activated to modulate these responses remain to be addressed. Here, we show that thrombin engages protein kinase C (PKC)-delta and phosphattidylinositol 3-kinase (PI3K)/Akt pathways to activate mTOR and thereby dampens NF-kappaB activation and intercellular adhesion molecule 1 (ICAM-1) expression. Stimulation of human vascular endothelial cells with thrombin induced the phosphorylation of mTOR and its downstream target p70 S6 kinase in a PKC-delta- and PI3K/Akt-dependent manner. Consistent with this, thrombin-induced phosphorylation of p70 S6 kinase was defective in embryonic fibroblasts from mice with targeted disruption of PKC-delta (Pkc-delta(-)(/)(-)), p85alpha and p85beta subunits of the PI3K (p85alpha(-)(/)(-)beta(-)(/)(-)), or Akt1 and Akt2 (Akt1(-)(/)(-)2(-)(/)(-)). Furthermore, we observed that expression of the constitutively active form of PKC-delta or Akt was sufficient to induce NF-kappaB activation and ICAM-1 expression, and that co-expression of mTOR suppressed these responses. In reciprocal experiments, inhibition/depletion of mTOR augmented NF-kappaB activation and ICAM-1 expression induced by PKC-delta or Akt. In control experiments, increasing or impairing mTOR signaling by the above approaches produced similar effects on NF-kappaB activation and ICAM-1 expression induced by thrombin. Thus, these data reveal an important role of PKC-delta and PI3K/Akt pathways in activating mTOR as an endogenous modulator to ensure a tight regulation of NF-kappaB signaling of ICAM-1 expression in endothelial cells.


Assuntos
Proteínas de Transporte/metabolismo , Células Endoteliais/metabolismo , Regulação da Expressão Gênica/fisiologia , Molécula 1 de Adesão Intercelular/biossíntese , NF-kappa B/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Proteína Quinase C-delta/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Proteínas de Transporte/genética , Linhagem Celular , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Células Endoteliais/citologia , Fibroblastos/citologia , Fibroblastos/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Hemostáticos/farmacologia , Humanos , Molécula 1 de Adesão Intercelular/genética , Camundongos , Camundongos Knockout , NF-kappa B/genética , Fosfatidilinositol 3-Quinases/genética , Fosforilação/efeitos dos fármacos , Fosforilação/fisiologia , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Proteína Quinase C-delta/genética , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Quinases S6 Ribossômicas 70-kDa/genética , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Serina-Treonina Quinases TOR , Trombina/farmacologia
16.
J Biol Chem ; 283(21): 14674-84, 2008 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-18362147

RESUMO

Protein kinase C-delta (PKC-delta) plays a pivotal role in mediating thrombin-induced NF-kappaB activation and ICAM-1 expression in endothelial cells. However, the downstream mechanisms mediating its function are unclear. In this study, we show that PKC-delta-mediated activation of protein-tyrosine kinase Syk plays an important role in thrombin signaling of NF-kappaB activation and intercellular adhesion molecule-1 (ICAM-1) expression in endothelial cells. Stimulation of human vascular endothelial cells with thrombin resulted in a time-dependent phosphorylation of Syk on tyrosine 525 and 526, an indication of Syk activation. Inhibition of PKC-delta by pharmacological and genetic approaches prevented Syk activation by thrombin. These results place Syk downstream of PKC-delta in transmitting thrombin-activated signaling in endothelial cells. Consistent with this, thrombin-induced NF-kappaB activity and ICAM-1 expression were prevented by the expression of a kinase-defective mutant or RNA interference knockdown of Syk. Similarly, inhibiting Syk also impaired NF-kappaB activity and ICAM-1 expression induced by a constitutively active mutant of PKC-delta. Analysis of the NF-kappaB pathway showed that Syk contributes to thrombin-induced NF-kappaB activation by controlling its transactivation potential and that this response is associated with tyrosine phosphorylation of RelA/p65. Thus, these data unveil a novel pathway in which Syk signals downstream of PKC-delta to mediate thrombin induced ICAM-1 expression in endothelial cells by increasing transcriptional capacity of NF-kappaB via a mechanism that relies on tyrosine phosphorylation of RelA/p65.


Assuntos
Células Endoteliais/metabolismo , Molécula 1 de Adesão Intercelular/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Fosfotirosina/metabolismo , Proteína Quinase C-delta/metabolismo , Proteínas Tirosina Quinases/metabolismo , Trombina/metabolismo , Fator de Transcrição RelA/metabolismo , Transporte Ativo do Núcleo Celular , Células Cultivadas , DNA/metabolismo , Ativação Enzimática , Regulação da Expressão Gênica , Humanos , Quinase I-kappa B/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , NF-kappa B/metabolismo , Ligação Proteica , Proteínas Tirosina Quinases/genética , Transdução de Sinais , Quinase Syk , Fator de Transcrição RelA/genética , Ativação Transcricional/genética
17.
FASEB J ; 22(7): 2297-310, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18263699

RESUMO

Vascular endothelial growth factor (VEGF) induces phosphorylation of VEGF receptor-2 (VEGFR-2) and activates the downstream signaling pathway resulting in endothelial cell migration, proliferation, and survival. Cigarette smoking is associated with abnormal vascular and endothelial function, leading to airspace enlargement. Herein, we investigated the mechanism of cigarette smoke (CS) -induced endothelial dysfunction by studying the VEGF-VEGFR-2 signaling in mouse lung and human endothelial cells. CS exposure caused oxidative stress, as shown by increased levels of 4-hydroxy-2-nonenal-adducts in mouse lung and reactive oxygen species generation in human lung microvascular endothelial cells (HMVEC-Ls). Inhibition of VEGFR-2 by a specific kinase inhibitor (NVP-AAD777) enhanced the CS-induced oxidative stress, causing augmented inflammatory cell influx and proinflammatory mediators release in mouse lung. The levels of endothelial nitric oxide synthase (eNOS) and phosphorylated (p) -eNOS in the lungs of mice exposed to CS and/or treated with VEGFR-2 inhibitor were decreased. CS down-regulated VEGFR-2 expression, eNOS levels, and VEGF-induced VEGFR-2 phosphorylation in HMVEC-Ls, resulting in impaired VEGF-induced endothelial cell migration and angiogenesis. Overall, these data show that inhibition of VEGFR-2 augmented CS-induced oxidative stress and inflammatory responses leading to endothelial dysfunction. This explains the mechanism of endothelial dysfunction in smokers and has implications in understanding the pathogenesis of pulmonary and cardiovascular diseases.


Assuntos
Endotélio Vascular/fisiopatologia , Inflamação/fisiopatologia , Estresse Oxidativo/fisiologia , Fumar/fisiopatologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Animais , Líquido da Lavagem Broncoalveolar , Modelos Animais de Doenças , Endotélio Vascular/patologia , Imuno-Histoquímica , Inflamação/patologia , Pulmão/patologia , Pulmão/fisiopatologia , Camundongos , Óxido Nítrico Sintase Tipo III/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
18.
Am J Physiol Lung Cell Mol Physiol ; 292(2): L396-404, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17012367

RESUMO

The procoagulant thrombin promotes polymorphonuclear leukocyte (PMN) adhesion to endothelial cells by a mechanism involving expression of intercellular adhesion molecule-1 (ICAM-1) via an NF-kappaB-dependent pathway. We now provide evidence that activation of c-Src is crucial in signaling thrombin-induced ICAM-1 expression via tyrosine phosphorylation of RelA/p65. Stimulation of human umbilical vein endothelial cells with thrombin resulted in a time-dependent activation of c-Src, with maximal activation occurring at 30 min after thrombin challenge. Inhibition of c-Src by pharmacological and genetic approaches impaired thrombin-induced NF-kappaB-dependent reporter activity and ICAM-1 expression. Analysis of the NF-kappaB pathway revealed that the effect of c-Src inhibition occurred independently of IkappaBalpha degradation and NF-kappaB DNA binding function and was not associated with exchange of NF-kappaB dimers. Phosphorylation of RelA/p65 at Ser(536), an event mediating the transcriptional activity of DNA-bound RelA/p65, was also insensitive to c-Src inhibition. Interestingly, thrombin induced association of c-Src with RelA/p65, and inhibition of c-Src prevented this response, indicating that this interaction is contingent on activation of c-Src. We also observed that thrombin induced tyrosine phosphorylation of RelA/p65, and this phosphorylation was lost upon inhibition of c-Src, consistent with the requirement of activated c-Src for interaction with RelA/p65. These data implicate an important role of c-Src in phosphorylating RelA/p65 to promote the transcriptional activity of NF-kappaB and thereby ICAM-1 expression in endothelial cells.


Assuntos
Células Endoteliais/enzimologia , Molécula 1 de Adesão Intercelular/metabolismo , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Trombina/farmacologia , Fator de Transcrição RelA/metabolismo , Proteína Tirosina Quinase CSK , DNA/metabolismo , Dimerização , Células Endoteliais/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Proteínas I-kappa B/metabolismo , Molécula 1 de Adesão Intercelular/genética , Inibidor de NF-kappaB alfa , Fosforilação/efeitos dos fármacos , Fosfosserina/metabolismo , Fosfotirosina/metabolismo , Ligação Proteica/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/deficiência , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas/deficiência , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Quinases da Família src
19.
J Biol Chem ; 282(6): 3940-50, 2007 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-17158457

RESUMO

Activation of the transcription factor NF-kappaB involves its release from the inhibitory protein IkappaBalpha in the cytoplasm and subsequently, its translocation to the nucleus. Whereas the events responsible for its release have been elucidated, mechanisms regulating the nuclear transport of NF-kappaB remain elusive. We now provide evidence for actin cytoskeleton-dependent and -independent mechanisms of RelA/p65 nuclear transport using the proinflammatory mediators, thrombin and tumor necrosis factor alpha, respectively. We demonstrate that thrombin alters the actin cytoskeleton in endothelial cells and interfering with these alterations, whether by stabilizing or destabilizing the actin filaments, prevents thrombin-induced NF-kappaB activation and consequently, expression of its target gene, ICAM-1. The blockade of NF-kappaB activation occurs downstream of IkappaBalpha degradation and is associated with impaired RelA/p65 nuclear translocation. Importantly, thrombin induces association of RelA/p65 with actin and this interaction is sensitive to stabilization/destabilization of the actin filaments. In parallel studies, stabilizing or destabilizing the actin filaments fails to inhibit RelA/p65 nuclear accumulation and ICAM-1 expression by tumor necrosis factor alpha, consistent with its inability to induce actin filament formation comparable with thrombin. Thus, these studies reveal the existence of actin cytoskeleton-dependent and -independent pathways that may be engaged in a stimulus-specific manner to facilitate RelA/p65 nuclear import and thereby ICAM-1 expression in endothelial cells.


Assuntos
Actinas/fisiologia , Núcleo Celular/metabolismo , Citoesqueleto/fisiologia , Endotélio Vascular/citologia , Endotélio Vascular/fisiologia , Transdução de Sinais/fisiologia , Fator de Transcrição RelA/metabolismo , Actinas/antagonistas & inibidores , Actinas/metabolismo , Transporte Ativo do Núcleo Celular/genética , Transporte Ativo do Núcleo Celular/fisiologia , Núcleo Celular/genética , Células Cultivadas , Citoesqueleto/metabolismo , Endotélio Vascular/metabolismo , Humanos , Mediadores da Inflamação/antagonistas & inibidores , Mediadores da Inflamação/metabolismo , Mediadores da Inflamação/fisiologia , Molécula 1 de Adesão Intercelular/biossíntese , Molécula 1 de Adesão Intercelular/genética , Molécula 1 de Adesão Intercelular/metabolismo , NF-kappa B/antagonistas & inibidores , NF-kappa B/metabolismo , Estabilidade de RNA/genética , RNA Mensageiro/antagonistas & inibidores , RNA Mensageiro/metabolismo , Transdução de Sinais/genética , Trombina/antagonistas & inibidores , Trombina/fisiologia , Fator de Transcrição RelA/antagonistas & inibidores , Fator de Necrose Tumoral alfa/fisiologia
20.
Free Radic Biol Med ; 41(4): 601-9, 2006 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-16863993

RESUMO

p21(Cip1/WAF1/Sdi1) is a major transcriptional target of p53 that promotes survival of cells exposed to continuous oxidative stress caused by hyperoxia. Because p21 can protect against genotoxic stress by reducing p53-dependent transcription of the proapoptotic proteins PUMA and Bax, the current study uses genetically modified lines of HCT116 colon carcinoma cells to investigate whether p21-mediated protection against hyperoxia involves attenuation of the p53 apoptotic pathway. Hyperoxia stimulated p53-dependent expression of p21 and Bax. Genetic ablation of p21 increased cell death, and loss of Bax or PUMA increased cell survival. Unlike damage caused by adriamycin, whereby p21 sensitivity could be rescued by removal of p53, PUMA, or Bax, increased sensitivity of p21-deficient cells to hyperoxia could not be rescued by additional loss of these genes. Instead, expression of the antiapoptotic protein Bcl-X(L) declined in p21-deficient cells exposed to hyperoxia, but when genetically restored, increased their survival. Conversely, siRNA knockdown of Bcl-X(L) in parental HCT116 cells increased hyperoxia-induced cell death. These findings reveal that p21-mediated protection against hyperoxia does not involve attenuation of p53-dependent apoptosis, but rather functions to maintain Bcl-X(L) expression during periods of persistent oxidative stress.


Assuntos
Neoplasias do Colo/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/fisiologia , Hiperóxia/fisiopatologia , Proteína bcl-X/metabolismo , Apoptose , Sequência de Bases , Western Blotting , Linhagem Celular Tumoral , Neoplasias do Colo/patologia , Primers do DNA , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA