Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Curr Oncol ; 25(2): 163-169, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29719432

RESUMO

Epidermal growth factor receptor (egfr) tyrosine kinase inhibitors (tkis) are recommended as first-line systemic therapy for patients with non-small-cell lung cancer (nsclc) having mutations in the EGFR gene. Resistance to tkis eventually occurs in all nsclc patients treated with such drugs. In patients with resistance to tkis caused by the EGFR T790M mutation, the third-generation tki osimertinib is now the standard of care. For optimal patient management, accurate EGFR T790M testing is required. A multidisciplinary working group of pathologists, laboratory medicine specialists, medical oncologists, a respirologist, and a thoracic radiologist from across Canada was convened to discuss best practices for EGFR T790M mutation testing in Canada. The group made recommendations in the areas of the testing algorithm and the pre-analytic, analytic, and post-analytic aspects of clinical testing for both tissue testing and liquid biopsy circulating tumour dna testing. The recommendations aim to improve EGFR T790M testing in Canada and to thereby improve patient care.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/genética , Testes Genéticos/métodos , Neoplasias Pulmonares/genética , Mutação , Algoritmos , Canadá , Análise Mutacional de DNA/métodos , Análise Mutacional de DNA/normas , DNA de Neoplasias/genética , Receptores ErbB/genética , Medicina Baseada em Evidências/métodos , Testes Genéticos/normas , Humanos
2.
Blood Cancer J ; 7(9): e613, 2017 09 29.
Artigo em Inglês | MEDLINE | ID: mdl-28960191

RESUMO

Meningioma 1 (MN1) is an independent prognostic marker for normal karyotype acute myeloid leukemia (AML), with high expression linked to all-trans retinoic acid resistance and poor survival. MN1 is also a potent and sufficient oncogene in murine leukemia models, strongly dependent on the MEIS1/AbdB-like HOX protein complex to transform common myeloid progenitors, block myeloid differentiation, and promote leukemic stem cell self-renewal. To identify key genes and pathways underlying leukemic activity, we functionally assessed MN1 cell phenotypic heterogeneity, revealing leukemic and non-leukemic subsets. Using gene expression profiling of these subsets combined with previously published comparisons of full-length MN1 and mutants with varying leukemogenic activity, we identified candidate genes critical to leukemia. Functional analysis identified Hlf and Hoxa9 as critical to MN1 in vitro proliferation, self-renewal and impaired myeloid differentiation. Although critical to transformation, Meis1 knockdown had little impact on these properties in vitro. However, we identified Meis2 as critical to MN1-induced leukemia, with essential roles in proliferation, self-renewal, impairment of differentiation and disease progression in vitro and in vivo. Here, we provide evidence of phenotypic and functional hierarchy in MN1-induced leukemic cells, characterise contributions of Hlf, Hoxa9 and Meis1 to in vitro leukemic properties, and reveal Meis2 as a novel player in MN1-induced leukemogenesis.


Assuntos
Regulação Leucêmica da Expressão Gênica , Proteínas de Homeodomínio/metabolismo , Leucemia/metabolismo , Proteínas Oncogênicas/metabolismo , Animais , Fatores de Transcrição de Zíper de Leucina Básica/genética , Fatores de Transcrição de Zíper de Leucina Básica/metabolismo , Técnicas de Silenciamento de Genes , Proteínas de Homeodomínio/genética , Leucemia/genética , Leucemia/patologia , Camundongos , Proteína Meis1/genética , Proteína Meis1/metabolismo , Proteínas Oncogênicas/genética , Transativadores , Proteínas Supressoras de Tumor
3.
Curr Oncol ; 24(2): 111-119, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28490925

RESUMO

Starting in the early 2000s, non-small-cell lung cancer (nsclc) subtypes have evolved from being histologically described to molecularly defined. Management of lung adenocarcinomas now generally requires multiple molecular tests at baseline to define the optimal treatment strategy. More recently, second biopsies performed at progression in patients treated with tyrosine kinase inhibitors (tkis) have further defined the continued use of molecularly targeted therapy. In the present article, we focus on one molecular subtype: EGFR-mutated nsclc. For that patient population, multiple lines of tki therapy are now available either clinically or in clinical trials. Each line of treatment is guided by the specific mutations (for example, L858R, T790M, C797S) identified in EGFR. We first describe the various mechanisms of acquired resistance to EGFR tki treatment. We then focus on strategies that clinicians and pathologists can both use during tissue acquisition and handling to optimize patient results. We also discuss future directions for the molecular characterization of lung cancers with driver mutations, including liquid biopsies. Finally, we provide an algorithm to guide treating physicians managing patients with EGFR-mutated nsclc. The same framework can also be applied to other molecularly defined nsclc subgroups as resistance patterns are elucidated and additional lines of treatment are developed.

5.
Blood Cancer J ; 5: e291, 2015 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-25768405

RESUMO

Nonsynonymous TP53 exon 4 single-nucleotide polymorphism (SNP), R72P, is linked to cancer and mutagen susceptibility. R72P associations with specific cancer risk, particularly hematological malignancies, have been conflicting. Myelodysplastic syndrome (MDS) with chromosome 5q deletion is characterized by erythroid hypoplasia arising from lineage-specific p53 accumulation resulting from ribosomal insufficiency. We hypothesized that apoptotically diminished R72P C-allele may influence predisposition to del(5q) MDS. Bone marrow and blood DNA was sequenced from 705 MDS cases (333 del(5q), 372 non-del(5q)) and 157 controls. Genotype distribution did not significantly differ between del(5q) cases (12.6% CC, 38.1% CG, 49.2% GG), non-del(5q) cases (9.7% CC, 44.6% CG, 45.7% GG) and controls (7.6% CC, 37.6% CG, 54.8% GG) (P=0.13). Allele frequency did not differ between non-del(5q) and del(5q) cases (P=0.91) but trended towards increased C-allele frequency comparing non-del(5q) (P=0.08) and del(5q) (P=0.10) cases with controls. Median lenalidomide response duration increased proportionate to C-allele dosage in del(5q) patients (2.2 (CC), 1.3 (CG) and 0.89 years (GG)). Furthermore, C-allele homozygosity in del(5q) was associated with prolonged overall and progression-free survival and non-terminal interstitial deletions that excluded 5q34, whereas G-allele homozygozity was associated with inferior outcome and terminal deletions involving 5q34 (P=0.05). These findings comprise the largest MDS R72P SNP analysis.


Assuntos
Deleção Cromossômica , Síndromes Mielodisplásicas/genética , Proteína Supressora de Tumor p53/genética , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Frequência do Gene , Humanos , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Mutação , Síndromes Mielodisplásicas/tratamento farmacológico , Síndromes Mielodisplásicas/patologia , Polimorfismo de Nucleotídeo Único , Resultado do Tratamento
6.
Curr Oncol ; 21(6): 305-8, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25489257

RESUMO

BACKGROUND: Activating mutations of the epidermal growth factor receptor (EGFR) gene are known to drive a proportion of non-small-cell lung cancers. Identification of lung cancers harbouring such mutations can lead to effective treatment using one of the agents that targets and blocks egfr-mediated signalling. METHODS: All specimens received at the BC Cancer Agency (Vancouver) for EGFR testing were prospectively identified and catalogued, together with clinical information and EGFR status, over a 14-month period. RESULTS: Specimens from 586 patients were received for EGFR testing, and EGFR status was reported for 509 patients. No relationship between specimen type or site of origin and EGFR test failure rate was identified. Concurrent immunohistochemical (ihc) status for thyroid transcription factor 1 (ttf1) was available for 309 patients. The negative predictive value of ttf1-negative status by ihc was 94.2% for predicting negative EGFR status. CONCLUSIONS: In patients with limited tissue available for testing, a surrogate for EGFR status would aid in timely management. Immunohistochemistry for ttf1 is readily available and correlates highly with EGFR status. In conjunction with genetic assays, ttf1 could be used to optimize an EGFR testing strategy.

7.
Oncogene ; 33(36): 4464-73, 2014 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-24096489

RESUMO

In an effort to identify novel biallelically inactivated tumor suppressor genes (TSGs) in sporadic invasive and preinvasive non-small-cell lung cancer (NSCLC) genomes, we applied a comprehensive integrated multiple 'omics' approach to investigate patient-matched, paired NSCLC tumor and non-malignant parenchymal tissues. By surveying lung tumor genomes for genes concomitantly inactivated within individual tumors by multiple mechanisms, and by the frequency of disruption in tumors across multiple cohorts, we have identified a putative lung cancer TSG, Eyes Absent 4 (EYA4). EYA4 is frequently and concomitantly deleted, hypermethylated and underexpressed in multiple independent lung tumor data sets, in both major NSCLC subtypes and in the earliest stages of lung cancer. We found that decreased EYA4 expression is not only associated with poor survival in sporadic lung cancers but also that EYA4 single-nucleotide polymorphisms are associated with increased familial cancer risk, consistent with EYA4s proximity to the previously reported lung cancer susceptibility locus on 6q. Functionally, we found that EYA4 displays TSG-like properties with a role in modulating apoptosis and DNA repair. Cross-examination of EYA4 expression across multiple tumor types suggests a cell-type-specific tumorigenic role for EYA4, consistent with a tumor suppressor function in cancers of epithelial origin. This work shows a clear role for EYA4 as a putative TSG in NSCLC.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/epidemiologia , Inativação Gênica , Neoplasias Pulmonares/patologia , Transativadores/genética , Carcinoma Pulmonar de Células não Pequenas/genética , Cromossomos Humanos Par 6 , Metilação de DNA , Epigênese Genética , Deleção de Genes , Regulação Neoplásica da Expressão Gênica , Frequência do Gene , Genes Supressores de Tumor , Estudos de Associação Genética , Variação Genética , Genoma Humano , Humanos , Neoplasias Pulmonares/genética , Polimorfismo de Nucleotídeo Único , Transativadores/metabolismo , Células Tumorais Cultivadas
8.
Handb Exp Pharmacol ; (180): 89-114, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17554506

RESUMO

Until recently, the adult neovasculature was thought to arise only through angiogenesis, the mechanism by which new blood vessels form from preexisting vessels through endothelial cell migration and proliferation. However, recent studies have provided evidence that postnatal neovasculature can also arise though vasculogenesis, a process by which endothelial progenitor cells are recruited and differentiate into mature endothelial cells to form new blood vessels. Evidence for the existence of endothelial progenitors has come from studies demonstrating the ability of bone marrow-derived cells to incorporate into adult vasculature. However, the exact nature of endothelial progenitor cells remains controversial. Because of the lack of definitive markers of endothelial progenitors, the in vivo contribution of progenitor cells to physiological and pathological neovascularization remains unclear. Early studies reported that endothelial progenitor cells actively integrate into the adult vasculature and are critical in the development of many types of vascular-dependent disorders such as neoplastic progression. Moreover, it has been suggested that endothelial progenitor cells can be used as a therapeutic strategy aimed at promoting vascular growth in a variety of ischemic diseases. However, increasing numbers of studies have reported no clear contribution of endothelial progenitors in physiological or pathological angiogenesis. In this chapter, we discuss the origin of the endothelial progenitor cell in the embryo and adult, and we discuss the cell's link to the primitive hematopoietic stem cell. We also review the potential significance of endothelial progenitor cells in the formation of a postnatal vascular network and discuss the factors that may account for the current lack of consensus of the scientific community on this important issue.


Assuntos
Vasos Sanguíneos/crescimento & desenvolvimento , Células da Medula Óssea/fisiologia , Células Endoteliais/fisiologia , Neovascularização Fisiológica , Células-Tronco/fisiologia , Animais , Hematopoese , Humanos , Neoplasias/irrigação sanguínea , Transplante de Células-Tronco
9.
Cell Death Differ ; 9(12): 1360-7, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12478473

RESUMO

Anchorage of cells to the extracellular matrix and integrin-mediated signals play crucial roles in cell survival. We have previously shown that during growth factor deprivation-induced apoptosis in human umbilical vein endothelial cells (HUVECs), key molecules in focal adhesions and adherens junctions are cleaved by caspases. In this study we provide evidence for a selective upregulation of cell-associated matrix metalloproteinases (MMPs). We observe a physical association of MMP2 with beta1 and alphav integrins, which increased three- to fourfold during apoptosis and is dependent upon integrin beta1 levels and activation state. Both enforced activation of beta1 integrin by a specific antibody and inhibition of MMPs protect HUVECs from apoptosis. We hypothesize that, prior to the commitment to apoptosis, 'inside-out' signals initiated by the apoptotic stimulus alter cell shape together with the activation states and/or the availability of integrins, which promote matrix-degrading activity around dying cells. This 'auxiliary' apoptotic pathway may interrupt ECM-mediated survival signaling, and thus accelerate the efficient execution of the cell death program.


Assuntos
Apoptose/fisiologia , Adesão Celular/fisiologia , Sobrevivência Celular/fisiologia , Endotélio Vascular/enzimologia , Matriz Extracelular/enzimologia , Integrinas/metabolismo , Metaloendopeptidases/metabolismo , Apoptose/efeitos dos fármacos , Adesão Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Proteínas do Citoesqueleto/metabolismo , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Humanos , Integrina alfaV/efeitos dos fármacos , Integrina alfaV/metabolismo , Integrina beta1/efeitos dos fármacos , Integrina beta1/metabolismo , Integrinas/efeitos dos fármacos , Metaloproteinase 2 da Matriz/efeitos dos fármacos , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinases da Matriz Associadas à Membrana , Metaloendopeptidases/antagonistas & inibidores , Paxilina , Fosfoproteínas/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
10.
Cancer Res ; 61(18): 6788-94, 2001 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-11559552

RESUMO

Tissue invasion is an important determinant of angiogenesis and metastasis and constitutes an attractive target for cancer therapy. We have developed an assay to identify agents that inhibit invasion by mechanisms other than inhibition of cell attachment or cytotoxicity. A screen of marine sponge extracts identified motuporamines as micromolar inhibitors of invasion of basement membrane gels by MDA-231 breast carcinoma, PC-3 prostate carcinoma, and U-87 and U-251 glioma cells. Motuporamine C inhibits cell migration in monolayer cultures and impairs actin-mediated membrane ruffling at the leading edge of lamellae. Motuporamine C also reduces beta1-integrin activation, raising the possibility that it interferes with "inside-out" signaling to integrins. In addition, motuporamine C inhibits angiogenesis in an in vitro sprouting assay with human endothelial cells and an in vivo chick chorioallantoic membrane assay. The motuporamines show little or no toxicity or inhibition of cell proliferation, and they are structurally simple and easy to synthesize, making them attractive drug candidates.


Assuntos
Alcaloides/farmacologia , Antineoplásicos/farmacologia , Neoplasias/tratamento farmacológico , Neovascularização Patológica/prevenção & controle , Alcaloides/isolamento & purificação , Animais , Neoplasias da Mama/irrigação sanguínea , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Movimento Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Embrião de Galinha , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Glioma/irrigação sanguínea , Glioma/tratamento farmacológico , Glioma/patologia , Humanos , Integrina beta1/metabolismo , Masculino , Invasividade Neoplásica , Neoplasias/irrigação sanguínea , Neoplasias/patologia , Neovascularização Patológica/tratamento farmacológico , Neovascularização Fisiológica/efeitos dos fármacos , Poríferos/química , Neoplasias da Próstata/irrigação sanguínea , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/patologia , Relação Estrutura-Atividade
11.
Lab Invest ; 81(8): 1119-31, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11502863

RESUMO

There is increasing evidence that antiproteases are able to affect the inflammatory response. To further examine this question, we administered human alpha-1-antitrypsin (alpha1AT) or a synthetic metalloprotease inhibitor (RS113456) to C57 mice followed by a single intratracheal dose of quartz, a dust that evokes a marked, lasting, polymorphonuclear leukocyte (PMN) infiltrate. At 2 hours after dust administration, both antiproteases completely suppressed silica-induced PMN influx into the lung and macrophage inflammatory protein-2 (MIP-2)/monocyte chemotactic protein-1 (MCP-1) (neutrophil/macrophage chemoattractant) gene expression, partially suppressed nuclear transcription factor kappaB (NF-kappaB) translocation, and increased inhibitor of NF-kappaB (IkappaB) levels. By 24 hours, PMN influx and connective tissue breakdown measured as lavage desmosine or hydroxyproline were still at, or close to, control levels after antiprotease treatment, and increases in NF-kappaB translocation and MIP-2/MCP-1 gene expression were variably suppressed. At both time points, neither agent prevented silica-induced increases in amount of whole lung MIP-2 or MCP-1 protein, but both did prevent increases in whole lung intercellular adhesion molecule-1 (ICAM-1) at 24 hours. Inactivating the alpha1AT by oxidation to the point that it no longer possessed antiproteolytic properties did not affect its ability to suppress inflammation. Both antiproteases also prevented the silica-induced acute inflammatory response in mice with knocked out genes for macrophage metalloelastase (MME -/-), mice that develop inflammation, but not connective tissue breakdown, and the pattern of alpha1AT breakdown fragments was identical in control and MME -/- animals. These findings suggest that, in this model of acute PMN mediated inflammation, a serine protease inhibitor and a metalloprotease inhibitor have similar anti-inflammatory properties, that inflammation is not mediated by proteolysis with generation of chemotactic matrix fragments, and that classic antiproteolysis (complexing of protease to antiprotease) probably does not play a role in suppression of inflammation. The antiproteolytic effects of these agents do not seem to be mediated by protection of endogenous alpha1AT.


Assuntos
Anti-Inflamatórios/farmacologia , Metaloendopeptidases/antagonistas & inibidores , Piranos/farmacologia , Inibidores de Serina Proteinase/farmacologia , alfa 1-Antitripsina/farmacologia , Animais , Quimiocina CCL2/biossíntese , Quimiocina CCL2/genética , Quimiocina CXCL2 , Quinase I-kappa B , Inflamação/induzido quimicamente , Inflamação/imunologia , Molécula 1 de Adesão Intercelular/metabolismo , Pulmão/efeitos dos fármacos , Pulmão/imunologia , Metaloproteinase 12 da Matriz , Metaloendopeptidases/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monocinas/biossíntese , Monocinas/genética , NF-kappa B/metabolismo , Infiltração de Neutrófilos , Proteínas Serina-Treonina Quinases/metabolismo , RNA Mensageiro/biossíntese , Dióxido de Silício
12.
Jpn Circ J ; 65(6): 556-60, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11407740

RESUMO

Although Fas-mediated cell death may play a role in atherogenesis, causal data in support of this hypothesis are lacking. The present study investigated the possibility that endothelial cells are involved in vascular smooth muscle cell (VSMC) apoptosis via the Fas-FasL pathway, and hence in atherogenesis. FACS analysis detected FasL on the surface of human umbilical vein endothelial cells (HUVECs) and immunofluorescence staining of the HUVECs demonstrated high levels of FasL in the intracellular compartment. FasL was down-regulated 4 h after tumor necrosis factor (TNFalpha) treatment, coinciding with maximal surface expression of the adhesion molecules vascular cell adhesion molecule-1 and E-selectin. However, the down-regulation of FasL expression was transient, as surface expression returned within 24 h of TNFalpha treatment. When cocultured with VSMCs, the FasL-expressing EC could kill the VSMCs in a manner that could be blocked by recombinant Fas-Fc, deployed as a soluble receptor for Fas. Moreover, when human coronary arteries were studied with immunohistochemistry using G247-4 monoclonal antibody for the detection of FasL, few FasL positive EC were observed in diffuse intimal thickening. In contrast, endothelium overlying the plaque showed prominent and uniform expression of FasL. These findings suggest that the Fas/FasL pathway can be used by EC to induce VSMC apoptosis in the atherosclerotic lesion.


Assuntos
Apoptose/efeitos dos fármacos , Endotélio Vascular/química , Glicoproteínas de Membrana/farmacologia , Músculo Liso Vascular/efeitos dos fármacos , Adulto , Idoso , Técnicas de Cocultura , Doença da Artéria Coronariana/etiologia , Doença da Artéria Coronariana/patologia , Regulação para Baixo/efeitos dos fármacos , Endotélio Vascular/patologia , Proteína Ligante Fas , Feminino , Humanos , Imuno-Histoquímica , Masculino , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/fisiologia , Pessoa de Meia-Idade , Músculo Liso Vascular/citologia , Fator de Necrose Tumoral alfa/farmacologia
13.
Histol Histopathol ; 15(4): 1303-25, 2000 10.
Artigo em Inglês | MEDLINE | ID: mdl-11005254

RESUMO

Tumor necrosis factor alpha (TNFalpha) has been shown to trigger many signaling pathways. Following oligomerization by TNFalpha, the receptors TNF-RI and TNF-RII associate with adapter molecules via specific protein-protein interactions. The subsequent recruitment of downstream molecules to the receptor complex enables propagation of the TNFalpha signal. Two cellular responses to TNFalpha have been well documented, the induction of cell death and the activation of gene transcription for cell survival. TNFalpha-induced apoptosis involves the activation of caspase cascades, which culminate in the cleavage of specific cellular substrates to effect cell death. TNFalpha has also been implicated in various caspase-independent cell death processes. Two transcription factors activated by TNFalpha are nuclear factor kappaB (NFkappaB) and activating protein 1 (AP-1). Pathways that promote the activation of these transcription factors involve signaling molecules such as kinases, phospholipases, and sphingomyelinases. In addition to increased survival (anti-apoptotic) gene expression, NFkappaB and AP-1 also induce the expression of genes involved in inflammation, cell growth, and signal regulation. The past decade has witnessed the identification of numerous signaling intermediates implicated in TNFalpha cellular responses. This article reviews the molecular mechanisms of TNFalpha signal transduction. In particular, pathways involved in cell death and transcription factor activation are discussed.


Assuntos
Transdução de Sinais/fisiologia , Fator de Necrose Tumoral alfa/fisiologia , Animais , Humanos , Ligantes , Transdução de Sinais/genética , Fator de Necrose Tumoral alfa/genética
14.
J Biol Chem ; 275(24): 18099-107, 2000 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-10849436

RESUMO

Tumor necrosis factor (TNF) does not cause endothelial apoptosis unless the expression of cytoprotective genes is blocked. We have previously demonstrated that one of the TNF-inducible cytoprotective genes is the Bcl-2 family member, A1. A1 is induced by the action of the transcription factor, NFkappaB, in response to inflammatory mediators. In this report we demonstrate that, as with other cell types, inhibition of NFkappaB initiates microvascular endothelial apoptosis in response to TNF. A1 is able to inhibit this apoptosis over 24 h. We demonstrate that A1 is localized to and functions at the mitochondria. Whereas A1 is able to inhibit mitochondrial depolarization, loss of cytochrome c, cleavage of caspase 9, BID, and poly(ADP-ribose) polymerase, it does not block caspase 8 or caspase 3 cleavage. In contrast, A1 is not able to prevent endothelial apoptosis by TNF over 72 h, when NFkappaB signaling is blocked. On the other hand, the caspase inhibitor, benzyloxycarbonyl-VAD-formylmethyl ketone, completely blocks TNF-induced endothelial apoptosis over 72 h. Our findings indicate that A1 is able to maintain temporary survival of endothelial cells in response to TNF by maintaining mitochondrial viability and function. However, a mitochondria-independent caspase pathway eventually results in endothelial death despite mitochondrial protection by A1.


Assuntos
Apoptose , Replicação do DNA , Proteínas de Ligação a DNA/fisiologia , Proteínas de Homeodomínio , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Repressoras , Proteínas de Saccharomyces cerevisiae , Fator de Necrose Tumoral alfa/farmacologia , Caspase 3 , Caspase 8 , Caspase 9 , Caspases/metabolismo , Células Cultivadas , Cicloeximida/farmacologia , Grupo dos Citocromos c/metabolismo , Dactinomicina/farmacologia , Endotélio/citologia , Humanos , Microcirculação , Microscopia Imunoeletrônica , Antígenos de Histocompatibilidade Menor , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , NF-kappa B/efeitos dos fármacos , NF-kappa B/metabolismo , Poli(ADP-Ribose) Polimerases/metabolismo , Inibidores da Síntese de Proteínas/farmacologia , Proteína de Replicação C
15.
Int J Mol Med ; 5(5): 447-56, 2000 May.
Artigo em Inglês | MEDLINE | ID: mdl-10762646

RESUMO

Vasculogenesis and angiogenesis are the mechanisms responsible for the development of the blood vessels. Angiogenesis refers to the formation of capillaries from pre-existing vessels in the embryo and adult organism, while vasculogenesis is the development of new blood vessels from the differentiation of endothelial precursors (angioblasts) in situ. Vascular endothelial growth factor (VEGF) family members are major mediators of vasculogenesis and angiogenesis both during development and in pathological conditions. VEGF has a variety of effects on vascular endothelium, including the ability to promote endothelial cell viability, mitogenesis, chemotaxis, and vascular permeability. It mediates its activity mainly via two tyrosine kinase receptors, VEGFR-1 (flt-1) and VEGFR-2 (flk-1/KDR), although other receptors, such as neuropilin-1 and -2, can also bind VEGF. Another tyrosine kinase receptor, VEGFR-3 (flt-4) binds VEGF-C and VEGF-D and is more important in the development of lymphatic vessels. While the functional effects of VEGF on endothelial cells has been well studied, not as much is known about VEGF signaling. This review summarizes the different pathways known to be involved in VEGF signal transduction and the biological responses triggered by the VEGF signaling cascade.


Assuntos
Fatores de Crescimento Endotelial/metabolismo , Linfocinas/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Receptores de Fatores de Crescimento/metabolismo , Transdução de Sinais , Animais , Fatores de Crescimento Endotelial/genética , Regulação da Expressão Gênica , Humanos , Linfocinas/genética , Neovascularização Fisiológica , Receptores Proteína Tirosina Quinases/genética , Receptores de Fatores de Crescimento/genética , Receptores de Fatores de Crescimento do Endotélio Vascular , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
16.
J Biol Chem ; 274(40): 28808-15, 1999 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-10497254

RESUMO

In the presence of a protein synthesis inhibitor, cycloheximide, tumor necrosis factor-alpha (TNF-alpha), interleukin 1-beta (IL-1beta), or lipopolysaccharide (LPS) induces human umbilical vein endothelial cells (HUVECs) to undergo apoptosis, suggesting that constitutive or inducible cytoprotective pathways are required for cell survival. We studied the correlation between nuclear factor-kappaB (NF-kappaB) activation and cell death induced by TNF-alpha, IL-1beta, or LPS. Adenovirus-mediated overexpression of a dominant-negative IkappaBalpha (inhibitor of kappaB) mutant blocked NF-kappaB activation by gel shift assay and blocked induction of vascular cell adhesion molecule-1 protein by TNF-alpha, IL-1beta, and LPS, a NF-kappaB-dependent response. In cells overexpressing the IkappaBalpha mutant, TNF-alpha induced cell death, whereas IL-1beta or LPS did not. We conclude that cell survival following TNF-alpha stimulation is NF-kappaB-dependent but that a constitutive or inducible NF-kappaB-independent pathway(s) protects IL-1beta- or LPS-treated HUVECs from cell death.


Assuntos
Endotélio Vascular/metabolismo , Interleucina-1/farmacologia , Lipopolissacarídeos/farmacologia , NF-kappa B/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Adenoviridae/fisiologia , Apoptose/efeitos dos fármacos , Sobrevivência Celular , Células Cultivadas , Endotélio Vascular/citologia , Humanos , Proteínas I-kappa B/genética , Mutação
17.
J Immunol ; 162(8): 4842-8, 1999 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-10202028

RESUMO

Adherence of leukocytes to cells undergoing apoptosis has been reported to be dependent on a variety of recognition pathways. These include alpha V beta 3 (CD51/CD61, vitronectin receptor), CD36 (thrombospondin receptor), macrophage class A scavenger receptor, phosphatidylserine translocated to the outer leaflet of apoptotic cell membranes, and CD14 (LPS-binding protein). We investigated the mechanism by which leukocytes adhere to apoptotic endothelial cells (EC). Peripheral blood mononuclear leukocytes and U937 monocytic cells adhered to human or bovine aortic EC induced to undergo apoptosis by withdrawal of growth factors, treatment with the promiscuous protein kinase inhibitor staurosporine, with the protein synthesis inhibitor and protein kinase activator anisomycin, or with the combination of cycloheximide and TNF-alpha. Expression of endothelial adherence molecules such as CD62E (E-selectin), CD54 (ICAM-1), and CD106 (VCAM-1) was not induced or increased by these treatments. A mAb to alpha V beta 3, exogenous thrombospondin, or blockade of phosphatidylserine by annexin V did not inhibit leukocyte adherence. Further, leukocyte binding to apoptotic EC was completely blocked by treatment of leukocytes but not EC with mAb to beta 1 integrin. These results define a novel pathway for the recognition of apoptotic cells.


Assuntos
Apoptose/imunologia , Integrina beta1/fisiologia , Leucócitos Mononucleares/imunologia , Neutrófilos/imunologia , Apoptose/efeitos dos fármacos , Biomarcadores/análise , Adesão Celular/efeitos dos fármacos , Adesão Celular/imunologia , Moléculas de Adesão Celular/fisiologia , Células Cultivadas , Endotélio Vascular/citologia , Endotélio Vascular/fisiologia , Matriz Extracelular/fisiologia , Humanos , Leucócitos Mononucleares/efeitos dos fármacos , Neutrófilos/efeitos dos fármacos , Fosfatidilserinas/fisiologia , Receptores de Vitronectina/fisiologia , Estaurosporina/farmacologia , Células U937
18.
J Biol Chem ; 274(12): 8039-45, 1999 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-10075703

RESUMO

Endothelial cell death may contribute to tissue injury from ischemia. Little is known, however, about the characteristics of endothelial cell death in response to hypoxia. Using an in vitro model, we found that human umbilical vein endothelial cells were resistant to hypoxia-induced cell death with only a 2% reduction in viability at 24 h and 45% reduction in viability at 48 h. Overexpression of a mutant, IkappaBalpha, via adenoviral vector did not potentiate cell death in hypoxia, indicating that nuclear factor-kappaB activation was not involved in cytoprotection. Cell death in hypoxia was determined to be apoptotic by 3' labeling of DNA using terminal deoxynucleotidyl transferase staining and reversibility of cell death with a caspase inhibitor. Exposure of endothelial cells to hypoxia did not alter levels of proapoptotic and antiapoptotic Bcl-2 family members Bax and Bcl-XL by immunoblot analysis. In contrast, changes in p53 protein levels correlated with the induction of apoptosis in hypoxic endothelial cells. Inhibition of the proteasome increased p53 protein levels and accelerated cell death in hypoxia. Overexpression of p53 by adenoviral transduction was sufficient to initiate apoptosis of normoxic endothelial cells. These data provide a framework for the study of factors regulating endothelial cell survival and death in hypoxia.


Assuntos
Apoptose , Endotélio Vascular/fisiologia , Proteínas I-kappa B , Proteína Supressora de Tumor p53/fisiologia , Adenoviridae/genética , Hipóxia Celular , DNA Nucleotidilexotransferase/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Endotélio Vascular/citologia , Ensaio de Imunoadsorção Enzimática , Humanos , Leucina/análogos & derivados , Leucina/metabolismo , Mutagênese , Inibidor de NF-kappaB alfa , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Transfecção , Células Tumorais Cultivadas , Proteína X Associada a bcl-2 , Proteína bcl-X
19.
Am J Hematol ; 59(4): 279-87, 1998 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9840908

RESUMO

We have defined an in vitro model for the study of microvascular endothelial cell (EC) apoptosis mediated by plasma from patients with various forms of thrombotic thrombocytopenic purpura (TTP) and hemolytic-uremic syndrome (HUS). This system reproduces a variety of histopathologic and ultrastructural features of tissue EC involved in TTP/sporadic HUS, suggesting that apoptotic EC injury is a primary pathophysiologic event in the thrombotic microangiopathies. We now document the ability of tetrapeptide-based inhibitors of interleukin 1beta-converting enzyme (ICE)-like caspase 1 and cysteine protease protein (CPP)-32-like caspase 3, two members of a novel class of cysteine proteases involved in final pathways to apoptosis, to block TTP/sporadic HUS plasma-mediated apoptosis. Overexpression of Bcl-X(L) via gene transfer suppressed this apoptosis by 70%. Transduction of EC with the Bcl-2 homolog A1 had a more limited protective effect. These findings support a role for apoptosis-linked cysteine proteases in the pathophysiology of TTP and sporadic HUS, and raise the possibility that specific apoptosis inhibitors may have a role in the experimental therapeutics of these syndromes.


Assuntos
Apoptose/fisiologia , Doenças Autoimunes/metabolismo , Caspase 1/fisiologia , Caspases/fisiologia , Endotélio Vascular/patologia , Síndrome Hemolítico-Urêmica/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/fisiologia , Púrpura Trombocitopênica Idiopática/metabolismo , Doenças Autoimunes/patologia , Caspase 3 , Inibidores de Caspase , Células Cultivadas , Endotélio Vascular/metabolismo , Infecções por HIV/metabolismo , Infecções por HIV/patologia , Síndrome Hemolítico-Urêmica/patologia , Humanos , Inibidores de Proteases/farmacologia , Proteínas Proto-Oncogênicas c-bcl-2/genética , Púrpura Trombocitopênica Idiopática/patologia , Proteínas Recombinantes de Fusão/fisiologia , Proteína bcl-X
20.
Blood ; 92(8): 2759-65, 1998 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-9763560

RESUMO

The effect of lipopolysaccharide (LPS) on endothelial cells is a key component of the inflammatory response seen in Gram-negative sepsis. LPS does not cause death of cultured human endothelial cells. However, when the expression of new proteins is inhibited by cycloheximide, microvascular endothelial cells in culture undergo apoptosis. This finding suggests that LPS induces apoptotic and antiapoptotic pathways, with the antiapoptotic response being dependent on the synthesis of new proteins. Concurrent activation of apoptotic and antiapoptotic pathways has previously been documented for tumor necrosis factor (TNF). In the case of TNF, the antiapoptotic signal has been attributed to at least two cytoprotective proteins: the Bcl-2 homologue, A1, and the zinc-finger protein, A20. In this study, we demonstrate that both these molecules are induced in microvascular endothelial cells by LPS. Enforced overexpression of either A1 or A20 inhibits LPS and cycloheximide-initiated apoptosis. Induction of A1 and A20 does not require synthesis of intermediary proteins, but is dependent on the presence of soluble CD14. In addition, we show that inhibition of signaling by the transcription factor, NF-kappaB, blocks accumulation of A1 and A20 mRNA. Taken together, our findings suggest that LPS directly induces expression of the cytoprotective proteins, A1 and A20, via a CD14-dependent pathway requiring activation of NF-kappaB.


Assuntos
Apoptose/fisiologia , Cicloeximida/farmacologia , Proteínas de Ligação a DNA/biossíntese , Endotélio Vascular/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas de Homeodomínio , Lipopolissacarídeos/farmacologia , Biossíntese de Proteínas , Proteínas Proto-Oncogênicas c-bcl-2 , Proteínas Repressoras , Proteínas de Saccharomyces cerevisiae , Apoptose/efeitos dos fármacos , Capilares , Células Cultivadas , Proteínas de Ligação a DNA/genética , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Receptores de Lipopolissacarídeos/fisiologia , Antígenos de Histocompatibilidade Menor , NF-kappa B/fisiologia , Proteínas Nucleares , Inibidores da Síntese de Proteínas/farmacologia , Proteínas/genética , Proteína de Replicação C , Transdução de Sinais , Proteína 3 Induzida por Fator de Necrose Tumoral alfa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA