Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
BMC Cancer ; 11: 358, 2011 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-21849059

RESUMO

BACKGROUND: The fusion protein VEGF(121)/rGel composed of the growth factor VEGF(121) and the plant toxin gelonin targets the tumor neovasculature and exerts impressive anti-vascular effects. We have previously shown that VEGF(121)/rGel is cytotoxic to endothelial cells overexpressing VEGFR-2 but not to endothelial cells overexpressing VEGFR-1. In this study, we examined the basis for the specific toxicity of this construct and assessed its intracellular effects in vitro and in vivo. METHODS: We investigated the binding, cytotoxicity and internalization profile of VEGF(121)/rGel on endothelial cells expressing VEGFR-1 or VEGFR-2, identified its effects on angiogenesis models in vitro and ex vivo, and explored its intracellular effects on a number of molecular pathways using microarray analysis. RESULTS: Incubation of PAE/VEGFR-2 and PAE/VEGFR-1 cells with (125)I-VEGF(121)/rGel demonstrated binding specificity that was competed with unlabeled VEGF(121)/rGel but not with unlabeled gelonin. Assessment of the effect of VEGF(121)/rGel on blocking tube formation in vitro revealed a 100-fold difference in IC(50) levels between PAE/VEGFR-2 (1 nM) and PAE/VEGFR-1 (100 nM) cells. VEGF(121)/rGel entered PAE/VEGFR-2 cells within one hour of treatment but was not detected in PAE/VEGFR-1 cells up to 24 hours after treatment. In vascularization studies using chicken chorioallantoic membranes, 1 nM VEGF(121)/rGel completely inhibited bFGF-stimulated neovascular growth. The cytotoxic effects of VEGF(121)/rGel were not apoptotic since treated cells were TUNEL-negative with no evidence of PARP cleavage or alteration in the protein levels of select apoptotic markers. Microarray analysis of VEGF(121)/rGel-treated HUVECs revealed the upregulation of a unique "fingerprint" profile of 22 genes that control cell adhesion, apoptosis, transcription regulation, chemotaxis, and inflammatory response. CONCLUSIONS: Taken together, these data confirm the selectivity of VEGF(121)/rGel for VEGFR-2-overexpressing endothelial cells and represent the first analysis of genes governing intoxication of mammalian endothelial cells by a gelonin-based targeted therapeutic agent.


Assuntos
Inibidores da Angiogênese/farmacologia , Células Endoteliais/efeitos dos fármacos , Proteínas Recombinantes de Fusão/farmacologia , Proteínas Inativadoras de Ribossomos Tipo 1/farmacologia , Fator A de Crescimento do Endotélio Vascular/farmacologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Aorta/citologia , Embrião de Galinha , Galinhas , Membrana Corioalantoide/irrigação sanguínea , Membrana Corioalantoide/efeitos dos fármacos , Sistemas de Liberação de Medicamentos , Células Endoteliais da Veia Umbilical Humana , Humanos , Neovascularização Fisiológica/efeitos dos fármacos , Proteínas Recombinantes de Fusão/genética , Proteínas Inativadoras de Ribossomos Tipo 1/genética , Suínos , Fator A de Crescimento do Endotélio Vascular/genética , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo
2.
J Cell Biol ; 175(6): 913-23, 2006 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-17158952

RESUMO

Cancer cells exhibit increased glycolysis for ATP production due, in part, to respiration injury (the Warburg effect). Because ATP generation through glycolysis is less efficient than through mitochondrial respiration, how cancer cells with this metabolic disadvantage can survive the competition with other cells and eventually develop drug resistance is a long-standing paradox. We report that mitochondrial respiration defects lead to activation of the Akt survival pathway through a novel mechanism mediated by NADH. Respiration-deficient cells (rho(-)) harboring mitochondrial DNA deletion exhibit dependency on glycolysis, increased NADH, and activation of Akt, leading to drug resistance and survival advantage in hypoxia. Similarly, chemical inhibition of mitochondrial respiration and hypoxia also activates Akt. The increase in NADH caused by respiratory deficiency inactivates PTEN through a redox modification mechanism, leading to Akt activation. These findings provide a novel mechanistic insight into the Warburg effect and explain how metabolic alteration in cancer cells may gain a survival advantage and withstand therapeutic agents.


Assuntos
Respiração Celular/fisiologia , Sobrevivência Celular , Glicólise/fisiologia , Mitocôndrias/metabolismo , Neoplasias/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Apoptose , Hipóxia Celular , DNA Mitocondrial/genética , DNA Mitocondrial/metabolismo , Ativação Enzimática , Humanos , NAD , Oxirredução , PTEN Fosfo-Hidrolase/genética , PTEN Fosfo-Hidrolase/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Células Tumorais Cultivadas
3.
Neuro Oncol ; 10(3): 275-91, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18443131

RESUMO

The p53 protein plays a critical role in inducing cell cycle arrest or apoptosis. Because p53 is inactivated in human gliomas, restoring p53 function is a major focus of glioma therapy. The most clinically tested strategy for replacing p53 has been adenoviral-mediated p53 gene therapy (Ad-p53). In addition to their therapeutic implications, investigations into Ad-p53 provide model systems for understanding p53's ability to induce cell cycle arrest versus apoptosis, particularly because wild-type p53 cells are resistant to Ad-p53-induced apoptosis. Here we use Ad-p53 constructs to test the hypothesis that simultaneous phosphorylation of p53 at threonine 18 (Thr18) and serine 20 (Ser20) is causally associated with p53-mediated apoptosis. Studies using phosphorylation-specific antibodies demonstrated that p53-induced apoptosis correlates with phosphorylation of p53 at Thr18 and Ser20 but not with carboxy-terminal phosphorylation (Ser392). To prove a causal relationship between apoptosis and Thr18 and Ser20 phosphorylation of p53, the effects of an adenoviral p53 construct that was not phosphorylated (Ad-p53) was compared with a Thr18/Ser20 phosphomimetic construct (Ad-p53-18D20D) in wild-type p53 gliomas. Whereas treatment with Ad-p53 resulted only in cell cycle arrest, treatment with Ad-p53-18D20D induced dramatic apoptosis. Microarray and Western blot analyses showed that only Ad-p53-18D20D was capable of inducing expression of apoptosis-inducing proteins. Chromatin immunoprecipitation assays indicated that the protein product of Ad-p53-18D20D, but not Ad-p53, was capable of binding to apoptosis-related genes. We thus conclude that phosphorylation of Thr18 and Ser20 is sufficient for inducing p53-mediated apoptosis in glioma cells. These results have implications for p53 gene therapy and inform other strategies that aim to restore p53 function.


Assuntos
Apoptose/fisiologia , Terapia Genética/métodos , Serina/metabolismo , Treonina/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Adenoviridae , Western Blotting , Ciclo Celular/fisiologia , Linhagem Celular Tumoral , Citometria de Fluxo , Imunofluorescência , Expressão Gênica , Regulação da Expressão Gênica , Vetores Genéticos , Humanos , Imunoprecipitação , Análise de Sequência com Séries de Oligonucleotídeos , Fosforilação , Reação em Cadeia da Polimerase
4.
Int J Radiat Oncol Biol Phys ; 71(1): 226-36, 2008 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-18406887

RESUMO

PURPOSE: The purpose of this study was to investigate early gene expression changes after chemoradiation in a human solid tumor, allowing identification of chemoradiation-induced gene expression changes in the tumor as well as the tumor microenvironment. In addition we aimed to identify a gene expression profile that was associated with clinical outcome. METHODS AND MATERIALS: Microarray experiments were performed on cervical cancer specimens obtained before and 48 h after chemoradiation from 12 patients with Stage IB2 to IIIB squamous cell carcinoma of the cervix treated between April 2001 and August 2002. RESULTS: A total of 262 genes were identified that were significantly changed after chemoradiation. Genes involved in DNA repair were identified including DDB2, ERCC4, GADD45A, and XPC. In addition, significantly regulated cell-to-cell signaling pathways included insulin-like growth factor-1 (IGF-1), interferon, and vascular endothelial growth factor signaling. At a median follow-up of 41 months, 5 of 12 patients had experienced either local or distant failure. Supervised clustering analysis identified a 58-gene set from the pretreatment samples that were differentially expressed between patients with and without recurrence. Genes involved in integrin signaling and apoptosis pathways were identified in this gene set. Immortalization-upregulated protein (IMUP), IGF-2, and ARHD had particularly marked differences in expression between patients with and without recurrence. CONCLUSIONS: Genetic profiling identified genes regulated by chemoradiation including DNA damage and cell-to-cell signaling pathways. Genes associated with recurrence were identified that will require validation in an independent patient data set to determine whether the 58-gene set associated with clinical outcome could be useful as a prognostic assay.


Assuntos
Carcinoma de Células Escamosas/genética , Reparo do DNA/genética , Perfilação da Expressão Gênica/métodos , Expressão Gênica/genética , Transdução de Sinais/genética , Neoplasias do Colo do Útero/genética , Adulto , Carcinoma de Células Escamosas/tratamento farmacológico , Carcinoma de Células Escamosas/patologia , Carcinoma de Células Escamosas/radioterapia , Feminino , Seguimentos , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/efeitos da radiação , Humanos , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Análise de Sequência com Séries de Oligonucleotídeos , Projetos Piloto , Reação em Cadeia da Polimerase , Estudos Prospectivos , Neoplasias do Colo do Útero/tratamento farmacológico , Neoplasias do Colo do Útero/patologia , Neoplasias do Colo do Útero/radioterapia
5.
Leuk Res ; 32(11): 1684-97, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18339423

RESUMO

Busulfan (Bu) resistance is a major obstacle to hematopoietic stem cell transplantation (HSCT) of patients with chronic or acute myelogenous leukemia (CML or AML). We used gene expression analysis to identify cellular factors underlying Bu resistance. Two Bu-resistant leukemia cell lines were established, characterized and analyzed for differentially expressed genes. The CML B5/Bu250(6) cells are 4.5-fold more resistant to Bu than their parental B5 cells. The AML KBM3/Bu250(6) cells are 4.0-fold more Bu-resistant than KBM3 parental cells. Both resistant sublines evade Bu-mediated G2-arrest and apoptosis with altered regulations of CHK2 and CDC2 proteins, constitutively up-regulated anti-apoptotic genes (BCL-X(L), BCL2, BCL2L10, BAG3 and IAP2/BIRC3) and down-regulated pro-apoptotic genes (BIK, BNIP3, and LTBR). Bu-induced apoptosis is partly mediated by activation of caspases; use of the inhibitor Z-VAD-FMK completely abrogated PARP1 cleavage and reduced apoptosis by approximately 50%. Furthermore, Bu resistance in these cells may be attributed in part to up-regulation of HSP90 protein and activation of STAT3. The inhibition of HSP90 with geldanamycin attenuated phosphorylated STAT3 and made B5/Bu250(6) and KBM3/Bu250(6) more Bu-sensitive. The analysis of cells derived from patients classified as either clinically resistant or sensitive to high-dose Bu-based chemotherapy indicated alterations in gene expression that were analogous to those observed in the in vitro model cell lines, confirming the potential clinical relevance of this model for Bu resistance.


Assuntos
Antineoplásicos Alquilantes/farmacologia , Bussulfano/farmacologia , Resistencia a Medicamentos Antineoplásicos/genética , Regulação Leucêmica da Expressão Gênica/efeitos dos fármacos , Leucemia Mieloide/genética , Apoptose/efeitos dos fármacos , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Caspases/metabolismo , Ativação Enzimática/efeitos dos fármacos , Fase G2/efeitos dos fármacos , Perfilação da Expressão Gênica , Proteínas de Choque Térmico HSP90/genética , Proteínas de Choque Térmico HSP90/metabolismo , Humanos , Leucemia Mieloide/patologia , Poli(ADP-Ribose) Polimerase-1 , Poli(ADP-Ribose) Polimerases/genética , Poli(ADP-Ribose) Polimerases/metabolismo , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Células Tumorais Cultivadas
6.
Chem Biol ; 13(3): 241-9, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16638529

RESUMO

Insulin-like growth factor 2 (IGF2) is a potent mitogen whose deregulation plays a role in developing liver, breast, and prostate cancers. Here, we take a small-molecule approach to investigate molecular pathways that modulate IGF2 signaling, by using chromeceptin, a synthetic molecule that selectively impairs the viability and growth of IGF2-overexpressing hepatocellular carcinoma cells. Affinity purification revealed that chromeceptin binds to multifunctional protein 2 (MFP-2), a seemingly multifunctional enzyme implicated in peroxisomal beta-oxidation. The small molecule-protein interaction stimulates the expression of IGF binding protein 1 (IGFBP-1) and suppressor of cytokine signaling-3 (SOCS-3), two cellular attenuators of the IGF signals, through activation of signal transducers and activators of transcription 6 (STAT6). The results underline the importance of STATs in IGF/insulin regulation, and they implicate a new pathway for STAT6 activation that is amenable to small-molecule intervention.


Assuntos
17-Hidroxiesteroide Desidrogenases/farmacologia , Enoil-CoA Hidratase/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Fator de Crescimento Insulin-Like II/metabolismo , Complexos Multienzimáticos/farmacologia , Fator de Transcrição STAT6/farmacologia , Transdução de Sinais/efeitos dos fármacos , Animais , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/metabolismo , Fator de Crescimento Insulin-Like II/genética , Neoplasias Hepáticas/patologia , Camundongos , Dados de Sequência Molecular , Oxirredução , Proteína Multifuncional do Peroxissomo-2 , Peroxissomos/metabolismo , Proteínas Supressoras da Sinalização de Citocina/metabolismo
7.
Cancer Res ; 65(23): 10767-75, 2005 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-16322222

RESUMO

Galectin-3 is a multifunctional beta-galactoside-binding protein implicated in apoptosis, malignant transformation, and tumor progression. The mechanisms by which galectin-3 contributes to malignant progression are not fully understood. In this study, we found that the introduction of wild-type galectin-3 into nontumorigenic, galectin-3-null BT549 human breast epithelial cells conferred tumorigenicity and metastatic potential in nude mice, and that galectin-3 expressed by the cells was phosphorylated. In contrast, BT549 cells expressing galectin-3 incapable of being phosphorylated (Ser6-->Glu Ser6-->Ala) were nontumorigenic. A microarray analysis of 10,000 human genes, comparing BT549 transfectants expressing wild-type and those expressing phosphomutant galectin-3, identified 188 genes that were differentially expressed (>2.5-fold). Genes affected by introduction of wild-type phosphorylated but not phosphomutant galectin-3 included those involved in oxidative stress, a novel noncaspase lysosomal apoptotic pathway, cell cycle regulation, transcriptional activation, cytoskeleton remodeling, cell adhesion, and tumor invasion. The reliability of the microarray data was validated by real-time reverse transcription-PCR (RT-PCR) and by Western blot analysis, and clinical relevance was evaluated by real-time RT-PCR screening of a panel of matched pairs of breast tumors. Differentially regulated genes in breast cancers that are also predicted to be associated with phospho-galectin-3 in transformed BT549 cells include C-type lectin 2, insulin-like growth factor-binding protein 5, cathepsins L2, and cyclin D1. These data show the functional diversity of galectin-3 and suggest that phosphorylation of the protein is necessary for regulation (directly or indirectly) of unique sets of genes that play a role in malignant transformation.


Assuntos
Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Galectina 3/metabolismo , Animais , Western Blotting , Linhagem Celular , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Células Epiteliais/fisiologia , Feminino , Galectina 3/biossíntese , Galectina 3/genética , Regulação da Expressão Gênica/fisiologia , Humanos , Camundongos , Camundongos Nus , Análise de Sequência com Séries de Oligonucleotídeos , Fosforilação , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção
8.
PLoS One ; 12(9): e0184154, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28880927

RESUMO

We demonstrate that SCF-KIT signaling induces synthesis and secretion of endothelin-3 (ET3) in human umbilical vein endothelial cells and melanoma cells in vitro, gastrointestinal stromal tumors, human sun-exposed skin, and myenteric plexus of human colon post-fasting in vivo. This is the first report of a physiological mechanism of ET3 induction. Integrating our finding with supporting data from literature leads us to discover a previously unreported pathway of nitric oxide (NO) generation derived from physiological endothelial NO synthase (eNOS) or neuronal NOS (nNOS) activation (referred to as the KIT-ET3-NO pathway). It involves: (1) SCF-expressing cells communicate with neighboring KIT-expressing cells directly or indirectly (cleaved soluble SCF). (2) SCF-KIT signaling induces timely local ET3 synthesis and secretion. (3) ET3 binds to ETBR on both sides of intercellular space. (4) ET3-binding-initiated-ETBR activation increases cytosolic Ca2+, activates cell-specific eNOS or nNOS. (5) Temporally- and spatially-precise NO generation. NO diffuses into neighboring cells, thus acts in both SCF- and KIT-expressing cells. (6) NO modulates diverse cell-specific functions by NO/cGMP pathway, controlling transcriptional factors, or other mechanisms. We demonstrate the critical physiological role of the KIT-ET3-NO pathway in fulfilling high demand (exceeding basal level) of endothelium-dependent NO generation for coping with atherosclerosis, pregnancy, and aging. The KIT-ET3-NO pathway most likely also play critical roles in other cell functions that involve dual requirement of SCF-KIT signaling and NO. New strategies (e.g. enhancing the KIT-ET3-NO pathway) to harness the benefit of endogenous eNOS and nNOS activation and precise NO generation for correcting pathophysiology and restoring functions warrant investigation.


Assuntos
Endotelina-3/metabolismo , Óxido Nítrico/metabolismo , Proteínas Proto-Oncogênicas c-kit/metabolismo , Receptor de Endotelina B/metabolismo , Fator de Células-Tronco/metabolismo , Aterosclerose/patologia , Linhagem Celular Tumoral , Endotélio Vascular/metabolismo , Ensaio de Imunoadsorção Enzimática , Motilidade Gastrointestinal , Tumores do Estroma Gastrointestinal/metabolismo , Tumores do Estroma Gastrointestinal/patologia , Tumores do Estroma Gastrointestinal/fisiopatologia , Homeostase , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Imuno-Histoquímica , Melanoma/patologia , Plexo Mientérico/metabolismo , Invasividade Neoplásica , Óxido Nítrico Sintase Tipo I/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Transdução de Sinais , Pele/metabolismo , Luz Solar , Fatores de Tempo , Regulação para Cima/genética , Vasodilatação
9.
Int J Radiat Oncol Biol Phys ; 64(3): 670-7, 2006 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-16289374

RESUMO

PURPOSE: The local-regional control rate for advanced head-and-neck squamous cell carcinoma (HNSCC) remains poor and is unpredictable for a given individual. This study examined whether gene expression patterns developed from tumors from surgicopathologic, criteria-defined, high-risk HNSCC patients could be correlated with clinical outcomes, namely, metastasis or nonrecurrent disease. METHODS AND MATERIALS: Fifteen primary tumors from patients treated with a consistent protocol of surgery followed by radiotherapy were examined. Seven of these tumors were from high-risk patients who developed distant metastasis (DM), and eight tumors were from patients with no recurrence (NR) (median follow-up, 59 months). RESULTS: Unsupervised clustering of gene expression did not separate the two groups from one another, but when supervised methodologies were applied, 205 genes discriminated the two groups. Within the DM group, genes associated with cell growth and proliferation; DNA replication, recombination, and repair; antiapoptotic pathways; cell adhesion; and angiogenesis were identified. For NR samples, discriminatory genes were associated with the onset of apoptosis. CONCLUSIONS: Our data suggest that gene expression analysis of surgically excised HNSCC tumors from patients considered at high risk for recurrence has the potential to identify individuals susceptible to metastasis on the basis of distinct gene-expression patterns. These patients would be ideal candidates for testing systemic therapy.


Assuntos
Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/secundário , Expressão Gênica , Neoplasias de Cabeça e Pescoço/genética , Carcinoma de Células Escamosas/radioterapia , Carcinoma de Células Escamosas/cirurgia , Ciclo Celular/genética , Proliferação de Células , Forma Celular/genética , Reparo do DNA/genética , Replicação do DNA , Neoplasias de Cabeça e Pescoço/patologia , Neoplasias de Cabeça e Pescoço/radioterapia , Neoplasias de Cabeça e Pescoço/cirurgia , Humanos , Metástase Linfática , Recidiva Local de Neoplasia/genética , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Transdução de Sinais/genética , Regulação para Cima
10.
Methods Mol Biol ; 319: 261-73, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16719360

RESUMO

Of the technologies available for measuring gene expression, microarrays using cDNA targets is one of the most common and well-developed high-throughput techniques. With this technique, the expression levels of thousands of genes are measured simultaneously. DNA probes are immobilized on solid surfaces, either membrane-based or chemically coated glass surfaces. On glass arrays, the probes are hybridized with fluorescent-labeled target samples. Fluorescence intensities, which reflect gene expression levels, are detected by imaging the array using a laser or white-light source and capturing the image using photomultiplier tube detection or a charge-coupled device camera. Different laser-based scanners are used in laboratories to scan microarray images. This chapter discusses the imaging process and the protocols being developed.


Assuntos
Perfilação da Expressão Gênica , Processamento de Imagem Assistida por Computador/métodos , Análise de Sequência com Séries de Oligonucleotídeos , Carbocianinas/metabolismo , Sondas de DNA/metabolismo , Corantes Fluorescentes/metabolismo , Humanos , Lasers , Análise de Sequência com Séries de Oligonucleotídeos/instrumentação , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Reprodutibilidade dos Testes , Estatística como Assunto
11.
Cancer Res ; 63(15): 4315-21, 2003 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-12907597

RESUMO

Comparison of gene expressing profiles between gliomas with different grades revealed frequent overexpression of insulin-like growth factor binding protein 2 (IGFBP2) in glioblastoma (GBM), the most advanced stage of glioma. To determine whether IGFBP2 is involved in the proliferative and invasive nature of GBM, we established stable SNB19 GBM cell lines that overexpress IGFBP2. Although there was no marked difference in the cell growth between IGFBP2 overexpressing SNB19(BP2) lines when compared with the control cells, these clones showed significantly increased invasive rates when compared with the parental or vector transfected SNB19 cells. Total RNAs from controls and SNB19(BP2) clones were used for microarray analysis to detect IGFBP2-mediated alterations in gene expression. When compared with parental or vector-transfected control cells, SNB19(BP2) cells consistently showed 3-5-fold increase in the expression of matrix metalloproteinase-2 (MMP-2) as well as other invasion related genes. Increased MMP-2 expression in SNB19(BP2) cells was subsequently confirmed by real time reverse-transcription PCR, Western blotting, and gelatin zymography. Furthermore, consistent with increased MMP-2 expression in SNB19(BP2) cells, transient transfection of a MMP-2 promoter/luciferase reporter also resulted in 3-6-fold higher luciferase activity in SNB19(BP2) cells than in parental or vector-transfected control cells. Finally, tissue microarray analysis of 68 GBM tissue specimens showed a significant correlation between the overexpression of IGFBP2 and elevated MMP-2 expression. Taken together, our data provide evidence that IGFBP2 contributes to glioma progression in part by enhancing MMP-2 gene transcription and in turn tumor cell invasion.


Assuntos
Glioblastoma/genética , Glioblastoma/patologia , Proteína 2 de Ligação a Fator de Crescimento Semelhante à Insulina/fisiologia , Regulação Neoplásica da Expressão Gênica , Glioblastoma/metabolismo , Humanos , Proteína 2 de Ligação a Fator de Crescimento Semelhante à Insulina/biossíntese , Proteína 2 de Ligação a Fator de Crescimento Semelhante à Insulina/genética , Metaloproteinase 2 da Matriz/biossíntese , Metaloproteinase 2 da Matriz/genética , Invasividade Neoplásica , Análise de Sequência com Séries de Oligonucleotídeos , Transfecção , Células Tumorais Cultivadas
12.
Cancer Res ; 64(17): 5913-9, 2004 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-15342366

RESUMO

KIT gain of function mutations play an important role in the pathogenesis of gastrointestinal stromal tumors (GISTs). Imatinib is a selective tyrosine kinase inhibitor of ABL, platelet-derived growth factor receptor (PDGFR), and KIT and represents a new paradigm of targeted therapy against GISTs. Here we report for the first time that, after imatinib treatment, an additional specific and novel KIT mutation occurs in GISTs as they develop resistance to the drug. We studied 12 GIST patients with initial near-complete response to imatinib. Seven harbored mutations in KIT exon 11, and 5 harbored mutations in exon 9. Within 31 months, six imatinib-resistant rapidly progressive peritoneal implants (metastatic foci) developed in five patients. Quiescent residual GISTs persisted in seven patients. All six rapidly progressive imatinib-resistant implants from five patients show an identical novel KIT missense mutation, 1982T-->C, that resulted in Val654Ala in KIT tyrosine kinase domain 1. This novel mutation has never been reported before, is not present in pre-imatinib or post-imatinib residual quiescent GISTs, and is strongly correlated with imatinib resistance. Allelic-specific sequencing data show that this new mutation occurs in the allele that harbors original activation mutation of KIT.


Assuntos
Antineoplásicos/farmacologia , Neoplasias Gastrointestinais/tratamento farmacológico , Neoplasias Gastrointestinais/genética , Mutação de Sentido Incorreto , Piperazinas/farmacologia , Proteínas Proto-Oncogênicas c-kit/genética , Pirimidinas/farmacologia , Alelos , Sequência de Bases , Benzamidas , Ensaios Clínicos Fase III como Assunto , Progressão da Doença , Resistencia a Medicamentos Antineoplásicos , Éxons , Neoplasias Gastrointestinais/enzimologia , Neoplasias Gastrointestinais/patologia , Humanos , Mesilato de Imatinib , Estrutura Terciária de Proteína , Ensaios Clínicos Controlados Aleatórios como Assunto , Células Estromais/patologia
13.
BMC Genomics ; 5(1): 35, 2004 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-15196312

RESUMO

BACKGROUND: DNA microarrays using long oligonucleotide probes are widely used to evaluate gene expression in biological samples. These oligonucleotides are pre-synthesized and sequence-optimized to represent specific genes with minimal cross-hybridization to homologous genes. Probe length and concentration are critical factors for signal sensitivity, particularly when genes with various expression levels are being tested. We evaluated the effects of oligonucleotide probe length and concentration on signal intensity measurements of the expression levels of genes in a target sample. RESULTS: Selected genes of various expression levels in a single cell line were hybridized to oligonucleotide arrays of four lengths and four concentrations of probes to determine how these critical parameters affected the intensity of the signal representing their expression. We found that oligonucleotides of longer length significantly increased the signals of genes with low-expression in the target. High-expressing gene signals were also boosted but to a lesser degree. Increasing the probe concentration, however, did not linearly increase the signal intensity for either low- or high-expressing genes. CONCLUSIONS: We conclude that the longer the oligonuclotide probe the better the signal intensities of low expressing genes on oligonucleotide arrays.


Assuntos
Perfilação da Expressão Gênica/métodos , Regulação Neoplásica da Expressão Gênica/genética , Genes Neoplásicos/genética , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Linhagem Celular Tumoral , Neoplasias Colorretais/genética , Sondas de DNA/genética , Sondas de DNA/metabolismo , DNA de Neoplasias/genética , DNA de Neoplasias/metabolismo , Humanos
14.
Cancer Biol Ther ; 2(5): 572-8, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-14614330

RESUMO

5-fluorouracil (5-FU) is the major chemotherapeutic agent for treatment of colorectal carcinoma, but the molecular mechanisms of response and resistance are not understood completely. We therefore studied the 5-FU dose response and time course of gene expression transcriptome changes in colon carcinoma cell lines that are relatively sensitive to or resistant to 5-FU (RKO and HT29, respectively. We identified cellular pathways and corroborated functions of selected pathways. Expression of genes for polyamine biosynthesis, i.e., ornithine decarboxylase (ODC) and spermine and spermidine synthases, was repressed in the sensitive line, while the biosynthesis-inhibiting gene ODC antizyme was induced in the resistant line. The rate-limiting gene in catabolism, spermine/spermidine acetyltransferase, was induced in both lines. Polyamine levels showed corresponding drastic decreases after 5-FU treatment, and polyamine replenishment interfered with 5-FU-induced apoptosis. In the sensitive cells which have wild-type p53, the p53 gene and its downstream genes including p21/WAF1, mdm2, Fas, mic-1, EphA2, and ferredoxin reductase as well as genes in the tumor necrosis factor (TNF) pathway including TNF receptor 2 (TNFR2) were induced, but not Fas ligand (FasL). Exposure to exogenous FasL increased 5-FU-induced apoptosis, and anti-TNFR2 antibody, but not anti-TNFR1, partially protected the sensitive cells. Our combination of gene expression profiling and corroborative functional studies revealed that reduced polyamine levels, non-autocrine FasL originating exogenous to tumor cells, and induced TNFR2 are all functional mediators of apoptosis caused by 5-FU in colon carcinoma cells.


Assuntos
Antígenos CD/metabolismo , Apoptose/efeitos dos fármacos , Neoplasias do Colo/patologia , Fluoruracila/farmacologia , Poliaminas/metabolismo , Receptores do Fator de Necrose Tumoral/metabolismo , Receptor fas/metabolismo , Acetiltransferases/metabolismo , Antimetabólitos Antineoplásicos/farmacologia , Neoplasias do Colo/genética , Neoplasias do Colo/metabolismo , Perfilação da Expressão Gênica , Humanos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Ornitina Descarboxilase/metabolismo , Receptores Tipo II do Fator de Necrose Tumoral , Espermidina Sintase/metabolismo , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/metabolismo
15.
Head Neck ; 31(5): 642-54, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19260130

RESUMO

BACKGROUND: The expression of miRNA in head and neck squamous cell carcinomas (HNSCCs) that had been classified as high risk by surgical pathologic features and validated by trial outcome for disease recurrence was determined and compared with matched adjacent normal tissues. METHODS: miRNA and corresponding gene expression were determined using miRNA bioarrays and gene expression arrays. RESULTS: Twenty miRNAs were determined to be differentially regulated in the HNSCC samples when compared with their normal tissue counterparts. Quantitative reverse transcriptase-polymerase chain reaction confirmed differential regulation of miRNA expression, and gene expression analysis on these same-paired samples confirmed the loss of putative mRNA targets including genes such as adenomatous polyposis coli, programmed cell death protein 4, and TGF beta receptor 3 in the tumor samples. CONCLUSIONS: These data suggest a role for the upregulation of specific miRNAs in high-risk HNSCC. Furthermore, upregulation of these miRNAs may be responsible for the elimination of mRNAs that lead to the growth and progression of HNSCC.


Assuntos
Carcinoma de Células Escamosas/genética , Perfilação da Expressão Gênica , Neoplasias de Cabeça e Pescoço/genética , MicroRNAs/metabolismo , Proteína da Polipose Adenomatosa do Colo/genética , Proteínas Reguladoras de Apoptose/genética , Quimiocinas C/genética , Regulação para Baixo , Humanos , Peroxirredoxinas/genética , Proteínas de Ligação a RNA/genética , Receptores de Fatores de Crescimento Transformadores beta/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Canais de Cátion TRPP/genética , Regulação para Cima
16.
Mod Pathol ; 20(8): 811-20, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17571080

RESUMO

Acute myeloid leukemia with inv(16)(p13q22), also known as M4Eo, is a distinct type of leukemia with characteristic clinicopathologic and cytogenetic features. Patients with M4Eo have monocytosis, high blast counts, and abnormal bone marrow eosinophils that contain large basophilic granules. The inv(16)(p13q22) or, less commonly, the t(16;16)(p13;q22) causes fusion of the CBFbeta gene at 16q22 and the MYH11 gene at 16p13, creating the novel chimeric protein CBFbeta-MYH11. To understand the underlying molecular mechanisms unique to M4Eo biology, we determined the gene expression profile of M4Eo cases by using cDNA and long oligonucleotide microarrays. Cases of acute myelomonocytic leukemia without CBFbeta-MYH11 (M4) acted as our control. We found that in the gene expression profile of M4Eo, NF-kappaB activators and inhibitors were upregulated and downregulated, respectively, suggesting that the NF-kappaB signaling pathway is activated at a higher level in M4Eo than in acute myelomonocytic leukemia M4. In addition, the gene expression profile of M4Eo indicates high cell proliferation and low apoptosis. We used real-time PCR, immunohistochemistry, and flow cytometry immunophenotyping to confirm some of our microarray data. These findings most likely represent the functional consequences of the abnormal chimeric protein CBFbeta-MYH11, which is unique to this disease, and suggest that NF-kappaB is a potential therapeutic target for treating M4Eo patients.


Assuntos
Cromossomos Humanos Par 16 , Citometria de Fluxo , Perfilação da Expressão Gênica/métodos , Imuno-Histoquímica , Imunofenotipagem/métodos , Leucemia Mielomonocítica Aguda/genética , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase , Adulto , Idoso , Apoptose/genética , Medula Óssea/química , Medula Óssea/imunologia , Medula Óssea/patologia , Proliferação de Células , Inversão Cromossômica , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Leucemia Mielomonocítica Aguda/imunologia , Leucemia Mielomonocítica Aguda/metabolismo , Leucemia Mielomonocítica Aguda/patologia , Masculino , Pessoa de Meia-Idade , Proteínas de Fusão Oncogênica/genética , Transdução de Sinais/genética , Fator de Transcrição RelA/análise
17.
Cancer ; 107(8): 1898-908, 2006 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-16986125

RESUMO

BACKGROUND: Imatinib has demonstrated marked clinical efficacy against gastrointestinal stromal tumor (GIST). Microarray technology, real-time polymerase chain reaction (PCR) validation, and fluorodeoxyglucose-positron emission tomography (FDG-PET) imaging were used to study the early molecular effects of imatinib antitumor activity in GIST. METHODS: After exposure of sensitive and resistant sarcoma cell lines to imatinib for 24 to 48 hours, the changes in gene expression were evaluated using a 1146 unique pathway array with Western blot validation. Real-time PCR was used to confirm changes in gene expression in human GIST samples (preimatinib biopsy and postimatinib surgical specimen after 3-7 days of therapy). FDG-PET was performed to correlate radiographic findings with the effects of imatinib on gene expression in GIST. RESULTS: In all, 55 genes demonstrated a > or = 2-fold change after imatinib treatment of the GIST882 cells. Among these genes there was up-regulation of insulin-like growth factor binding protein-3 (IGFBP-3), a protein that modulates proliferation and apoptosis. Western blot analysis confirmed the increase of IGFBP-3 only in imatinib-sensitive GIST882 cells. Up to a 7-fold induction (49% mean increase; P = .08) of IGFBP-3 mRNA was found in tumor samples from patients with low residual FDG uptake, whereas there was an up to 12-fold reduction (-102% mean decrease; P = .03) in IGFBP-3 in those patients with high residual FDG uptake after imatinib therapy. CONCLUSIONS: In the current study, imatinib appears to regulate numerous genes and specifically induces IGFBP-3 in GIST cells and tumor samples. IGFBP-3 levels also were found to be inversely correlated with residual FDG uptake in GIST patients early in imatinib therapy. These initial observations suggest that IGFBP-3 is an important early marker of antitumor activity of imatinib in GIST.


Assuntos
Antineoplásicos/farmacologia , Tumores do Estroma Gastrointestinal/metabolismo , Piperazinas/farmacologia , Tomografia por Emissão de Pósitrons , Pirimidinas/farmacologia , Apoptose , Benzamidas , Sobrevivência Celular , Tumores do Estroma Gastrointestinal/diagnóstico por imagem , Tumores do Estroma Gastrointestinal/genética , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Mesilato de Imatinib , Proteína 3 de Ligação a Fator de Crescimento Semelhante à Insulina , Proteínas de Ligação a Fator de Crescimento Semelhante a Insulina/metabolismo , Radiografia , Sarcoma/genética , Regulação para Cima
18.
J Biol Chem ; 280(5): 3295-304, 2005 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-15546879

RESUMO

The thymidylate synthase inhibitor 5-fluorouracil (5-FU) is used widely for chemotherapy of colorectal carcinoma. Recent studies showed that 5-FU affects polyamine metabolism in colon carcinoma cells. We therefore examined whether combinations of 5-FU with drugs that specifically target polyamine metabolism, i.e. N1,N11-diethylnorspermine (DENSPM) or alpha-difluoromethylornithine (DFMO), have synergistic effects in killing HCT116 colon carcinoma cells with wild-type or absent p53. Our results showed that simultaneous 5-FU and DENSPM, a spermine analogue, synergistically increased transcript levels of the polyamine catabolism enzyme spermidine/spermine N1-acetyltransferase, depleted spermine and spermidine, increased acetylated spermidine, and produced synergistic tumor cell apoptosis in both p53 wild-type and p53-null variants. By contrast, simultaneous combination of 5-FU with DFMO, an inhibitor of the polyamine biosynthetic enzyme ornithine decarboxylase, depleted putrescine but did not produce synergistic cell killing. Some pre-treatment and post-treatment regimens of DENSPM and DFMO were antagonistic to 5-FU depending on cellular p53 status. Protein and transcriptome expression analysis showed that combined 5-FU and DENSPM treatment activated caspase 9, but not caspase 3, and significantly suppressed NADH dehydrogenases and cytochrome c oxidases, consistent with the observed increase in hydrogen peroxide, loss of mitochondrial membrane potential, and release of cytochrome c. Our findings demonstrate the importance of the polyamine pathway in 5-FU effects and suggest that the combination of 5-FU with DENSPM has potential for development as therapy for colorectal carcinoma.


Assuntos
Acetiltransferases/metabolismo , Antimetabólitos Antineoplásicos/farmacologia , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Fluoruracila/farmacologia , Poliaminas/metabolismo , Espermina/análogos & derivados , Espermina/farmacologia , Acetiltransferases/genética , Antioxidantes/metabolismo , Caspase 9 , Caspases/metabolismo , Citocromos c/metabolismo , Sinergismo Farmacológico , Deleção de Genes , Células HCT116 , Humanos , Peróxido de Hidrogênio/metabolismo , Mitocôndrias/metabolismo , Transcrição Gênica/efeitos dos fármacos , Proteína Supressora de Tumor p53/genética
19.
Arch Biochem Biophys ; 397(1): 11-7, 2002 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-11747305

RESUMO

The function of autophosphorylation in Src family protein tyrosine kinases is not fully understood. In this paper we compared the catalytic and ligand-binding properties of autophosphorylated and nonautophosphorylated (control) Src. The following are the main differences we found. First, while both forms had the same K(m) for ATP-Mg, autophosphorylated Src had significantly higher K(m) values for the phosphate-accepting substrates, polyE(4)Y, and RCM-lysozyme. The autophosphorylated form also had significantly higher V(max) values than the control. The substrate specificity, as measured by V(max)/K(m) ratio, was altered by autophosphorylation and was dependent on the phosphate-accepting substrate. Second, while autophosphorylation did not affect Src activation by free Mg(2+), Zn(2+), which inhibited Src by competing against an essential Mg(2+) activator, inhibited the control threefold more potently than the autophosphorylated form. Third, autophosphorylation significantly reduced the ability of its SH2 domain to bind phosphotyrosine. Fourth, a Pro-rich Src SH3 domain binding peptide activated the control, but not the autophosphorylated Src even though the apparent binding affinity was not significantly affected by autophosphorylation. These differences indicated that autophosphorylation induced significant and widespread changes in the catalytic and regulatory properties of Src. The implications of these findings relative to Src biological regulation are discussed.


Assuntos
Proteínas Tirosina Quinases/química , Proteína Tirosina Quinase CSK , Catálise , Cloretos/química , Relação Dose-Resposta a Droga , Concentração Inibidora 50 , Cinética , Ligantes , Magnésio/química , Muramidase/química , Peptídeos/química , Fosforilação , Fosfotirosina/química , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas Recombinantes/química , Sefarose/química , Cloreto de Sódio/farmacologia , Fatores de Tempo , Zinco/química , Compostos de Zinco/química , Domínios de Homologia de src , Quinases da Família src
20.
Cancer ; 94(7): 2069-75, 2002 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-11932911

RESUMO

Leiomyosarcomas are malignant smooth muscle tumors characterized by a spectrum of histopathologic features and clinical behavior. Gene expression profiling of leiomyosarcomas may identify differential gene signatures that may allow for the clinical stratification of the tumors. Typically, surgical specimens from these tumors are large and manifest a variegated macroscopic appearance. Because of their large size at the time of diagnosis, sufficient tissue is available for regional and clonal heterogeneity assessment. However, if the gene expression profiles of samples taken from different locations in the tumors are drastically different, biologic classification on the basis of random sample analysis may not be adequate. Therefore, to assess intertumor and intratumor heterogeneity, the authors performed a gene expression study using leiomyosarcoma specimens from three excised sarcomas from an equal number of different patients. Comparisons between tumor and normal samples from the three patients as well as between carefully mapped peripheral and core specimens from the same tumor (excised from one of the patients), were performed. Analysis of the expression profiles demonstrated minimal intratumor variations compared with intertissue variations, indicating homogeneous tumor specific gene expression profiles. The authors also identified genes that are expressed differentially in tumor and normal tissue.


Assuntos
Biomarcadores Tumorais/genética , Expressão Gênica , Leiomiossarcoma/genética , Neoplasias Musculares/genética , Proteínas de Neoplasias/genética , Biomarcadores Tumorais/metabolismo , Análise por Conglomerados , Primers do DNA/química , Perfilação da Expressão Gênica/métodos , Heterogeneidade Genética , Humanos , Leiomiossarcoma/metabolismo , Leiomiossarcoma/patologia , Neoplasias Musculares/metabolismo , Neoplasias Musculares/patologia , Proteínas de Neoplasias/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro/metabolismo , RNA Neoplásico/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA