Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Death Differ ; 19(3): 501-10, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21941369

RESUMO

Suppression of anoikis after detachment of cancer cells from the extracellular matrix is a key step during metastasis. Here we show that, after detachment, mouse embryonic fibroblasts (MEFs) transformed by K-Ras(V12) or ETV6-NTRK3 (EN) activate a transcriptional response overrepresented by genes related to bioenergetic stress and the AMP-activated protein kinase (AMPK) energy-sensing pathway. Accordingly, AMPK is activated in both transformed and non-transformed cells after detachment, and AMPK deficiency restores anoikis to transformed MEFs. However, AMPK activation represses the mTOR complex-1 (mTORC1) pathway only in transformed cells, suggesting a key role for AMPK-mediated mTORC1 inhibition in the suppression of anoikis. Consistent with this, AMPK-/- MEFs transformed by EN or K-Ras show sustained mTORC1 activation after detachment and fail to suppress anoikis. Transformed TSC1-/- MEFs, which are incapable of suppressing mTORC1, also undergo anoikis after detachment, which is reversed by mTORC1 inhibitors. Furthermore, transformed AMPK-/- and TSC1-/- MEFs both have higher total protein synthesis rates than wild-type controls, and translation inhibition using cycloheximide partially restores their anoikis resistance, indicating a mechanism whereby mTORC1 inhibition suppresses anoikis. Finally, breast carcinoma cell lines show similar detachment-induced AMPK/mTORC1 activation and restoration of anoikis by AMPK inhibition. Our data implicate AMPK-mediated mTORC1 inhibition and suppression of protein synthesis as a means for bioenergetic conservation during detachment, thus promoting anoikis resistance.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Anoikis/fisiologia , Embrião de Mamíferos/metabolismo , Fibroblastos/metabolismo , Biossíntese de Proteínas , Serina-Treonina Quinases TOR/metabolismo , Proteínas Quinases Ativadas por AMP/genética , Animais , Linhagem Celular Transformada , Linhagem Celular Tumoral , Embrião de Mamíferos/citologia , Fibroblastos/citologia , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Knockout , Complexos Multiproteicos , Células NIH 3T3 , Proteínas/genética , Proteínas/metabolismo , Serina-Treonina Quinases TOR/antagonistas & inibidores , Serina-Treonina Quinases TOR/genética
2.
Clin Cancer Res ; 7(8): 2590-6, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11489843

RESUMO

BTO-956 [methyl-3,5-diiodo-4-(4'-methoxyphenoxy)benzoate], a novel tubulin-binding drug and thyroid hormone analogue, was originally found to inhibit human carcinoma cell proliferation in vitro and to have potent growth delay activity in human breast and ovarian carcinoma xenografts in nude mice. Here we report that BTO-956 given to Fischer 344 rats also inhibits corneal angiogenesis and the growth and neovascularization of the R3230Ac rat mammary carcinoma tumor implanted in skin-fold window chambers. Hydron pellets containing recombinant human basic fibroblast growth factor (50 ng) and Sucralfate (20 microg) were implanted into surgically created corneal micropockets (day 0). BTO-956 was administrated by oral gavage (500 mg/kg, twice a day for 6 days) on days 1-6 (controls received vehicle alone). On day 7, rats received retrograde infusions of India ink via the thoracic aorta to visualize the corneal vasculature. Digitized images of slide-mounted corneas from control and treated animals were taken with a microscope. For the tumor growth and angiogenesis study, small pieces of R3230Ac tumor from a donor rat were implanted into surgically prepared window chambers (day 0). BTO-956 was given during days 5-11, and images of the tumors and their vasculature were recorded on day 12. No body weight loss was observed in either study. BTO-956 significantly inhibited corneal angiogenesis (by 50-80%), as assessed by measurements of limbal circumference displaying neovascularization, vessel length, vascularized area, and vascular area density. In the window chamber assay, tumors from treated animals were >50% smaller than tumors in control animals. In addition, vascular length densities in peripheral tumor zones were 30% less in treated compared with control animals. Together, these findings demonstrate that BTO-956 can inhibit angiogenesis induced by a growth factor in the rat cornea and in the peripheral area of implanted tumors, where tumor angiogenesis is most active.


Assuntos
Antineoplásicos/farmacologia , Iodobenzoatos/farmacologia , Neoplasias Mamárias Experimentais/tratamento farmacológico , Neovascularização Patológica/prevenção & controle , Tubulina (Proteína)/metabolismo , Animais , Antineoplásicos/metabolismo , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Neovascularização da Córnea/patologia , Neovascularização da Córnea/prevenção & controle , Relação Dose-Resposta a Droga , Feminino , Humanos , Neoplasias Mamárias Experimentais/irrigação sanguínea , Neoplasias Mamárias Experimentais/patologia , Neovascularização Patológica/patologia , Ligação Proteica , Ratos , Ratos Endogâmicos F344
3.
Biochem Biophys Res Commun ; 284(5): 1134-9, 2001 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-11414701

RESUMO

The c-jun gene is a major regulator of proliferative and stress responses of both normal and transformed cells. In general, during immortalization/transformation c-jun cooperates with oncogenic signals rather than acting as an oncogene itself. Here we report a novel example of this cooperation, the requirement for c-jun to sustain expression of the matrix metalloproteinase-2 (MMP-2) gene in cells immortalized by SV40 large T-antigen (TAg). MMP-2 encodes a type IV collagenase that is secreted by cells within normal and tumor microenvironments. We used wild-type and c-jun null primary and TAg-immortalized mouse embryonic fibroblasts (mEFs) to investigate the importance of c-jun for the regulation of this activity, and observed that c-jun is essential for MMP-2 expression in immortalized but not primary mEFs. This finding directly demonstrates a cooperative interaction of c-jun with an oncogene, and suggests that TAg dependent immortalization/transformation may require other c-Jun/AP-1-dependent genes.


Assuntos
Antígenos Transformantes de Poliomavirus/farmacologia , Metaloproteinase 2 da Matriz/biossíntese , Proteínas Proto-Oncogênicas c-jun/farmacologia , Animais , Linhagem Celular Transformada , Transformação Celular Viral , Colagenases/metabolismo , Fibroblastos/efeitos dos fármacos , Fibroblastos/enzimologia , Fibroblastos/metabolismo , Expressão Gênica/efeitos dos fármacos , Camundongos , Peso Molecular , Retroviridae/genética , Transcrição Gênica/efeitos dos fármacos
4.
Cancer Res ; 61(6): 2696-703, 2001 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-11289150

RESUMO

Placenta growth factor (PlGF) is a mitogen for endothelial cells that can potentiate the growth and permeabilizing effects on endothelium of vascular endothelial growth factor. Here we report that hypoxia induces the expression of both PlGF mRNA and protein in immortalized/transformed mouse embryonic fibroblasts (mEFs) and in NIH 3T3 cells. Importantly, the magnitude of the induction of PlGF expression by hypoxia is enhanced by the presence of oncogenic Ras. To investigate the transcriptional component of hypoxia-inducible PlGF expression, we cloned and sequenced a 1350-bp fragment of the 5'-flanking region of the mouse gene. Analysis of the promoter region indicated the presence of putative consensus sequences for known hypoxia-responsive regulatory sites, including metal response elements and Sp1-like sites. In the present study, we show that the induction of PlGF expression by hypoxia is dependent on the presence of the metal response element-binding transcription factor 1 (MTF-1). Thus, in mEFs with targeted deletions of both MTF-1 alleles, hypoxia-induced increases of PIGF mRNA and protein levels were greatly attenuated compared with those in wild-type mEFs. Moreover, transient transfection of a PlGF promoter reporter gene into NIH 3T3 cells resulted in hypoxia-responsive transcriptional activation of the reporter. Finally, ectopic expression of MTF-1 resulted in increased basal transcriptional activity of a PlGF promoter reporter. Together, these findings demonstrate that the PlGF gene is responsive to hypoxia and that this response is mediated by MTF-1. It remains to be determined whether this activation is the result of direct and/or indirect transcriptional activation by MTF-1. The stimulatory effect of oncogenic Ras on the induction of PlGF expression in hypoxic cells suggests that PlGF could be an important proangiogenic factor in the tumor microenvironment.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Oxigênio/fisiologia , Proteínas da Gravidez/biossíntese , Fatores de Transcrição/fisiologia , Células 3T3 , Animais , Sequência de Bases , Hipóxia Celular , Linhagem Celular Transformada , Clonagem Molecular , DNA/genética , DNA/isolamento & purificação , Proteínas de Ligação a DNA , Embrião de Mamíferos , Fibroblastos/metabolismo , Fibroblastos/fisiologia , Regulação Neoplásica da Expressão Gênica , Genes ras/fisiologia , Humanos , Camundongos , Dados de Sequência Molecular , Fator de Crescimento Placentário , Proteínas da Gravidez/genética , Regiões Promotoras Genéticas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ativação Transcricional/fisiologia , Células Tumorais Cultivadas , Fator MTF-1 de Transcrição
5.
Clin Cancer Res ; 6(7): 2941-50, 2000 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10914744

RESUMO

Tumor angiogenesis, the development of new blood vessels during malignant progression, is a regulated process that has both genetic and physiological controls. Physiologically, angiogenesis is stimulated by decreases in tissue oxygenation (i.e., hypoxia). We investigated the effect of hypoxia on the expression of two angiogenic factors reported to be genetically regulated by the p53 tumor suppressor gene: (a) the angiogenic inhibitor thrombospondin 1 (TSP-1); and (b) the angiogenic inducer vascular endothelial growth factor (VEGF). Analysis of rodent cells that differ in their p53 genotype (p53+/+ or p53-/-) indicated that in vitro exposure to hypoxia simultaneously suppressed TSP-1 and induced VEGF expression, regardless of the p53 genotype. On transformation of these cells with E1A and oncogenic H-ras, the basal level of TSP-1 expression was strongly diminished, whereas that of VEGF could still be induced by hypoxia. Consistent with these in vitro findings, sections of tumors derived from the transformed p53+/+ and p53-/- cells showed that VEGF protein overlapped with regions of hypoxia, whereas TSP-1 protein was below the limits of detection in tumor tissue. Using a panel of normal/immortalized and transformed human cells, it was found that the ability of hypoxia to inhibit TSP-1 expression depends on the cell type and/or the degree of transformation. In contrast, VEGF expression was induced by hypoxia in all of the human cell types examined. Together, these findings suggest that hypoxic and oncogenic signals could interact in the tumor microenvironment to inhibit TSP-1 and induce VEGF expression, promoting the switch to the angiogenic phenotype.


Assuntos
Carcinoma de Células Escamosas/genética , Hipóxia Celular , Fatores de Crescimento Endotelial/genética , Regulação da Expressão Gênica , Genes p53 , Linfocinas/genética , Trombospondina 1/genética , Neoplasias do Colo do Útero/genética , Animais , Carcinoma de Células Escamosas/patologia , Divisão Celular , Células Cultivadas , Feminino , Humanos , Camundongos , Camundongos SCID , Transfecção , Proteína Supressora de Tumor p53/genética , Neoplasias do Colo do Útero/patologia , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular , Ensaios Antitumorais Modelo de Xenoenxerto
6.
J Biol Chem ; 274(18): 12890-7, 1999 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-10212278

RESUMO

Pathophysiological hypoxia is an important modulator of gene expression in solid tumors and other pathologic conditions. We observed that transcriptional activation of the c-jun proto-oncogene in hypoxic tumor cells correlates with phosphorylation of the ATF2 transcription factor. This finding suggested that hypoxic signals transmitted to c-jun involve protein kinases that target AP-1 complexes (c-Jun and ATF2) that bind to its promoter region. Stress-inducible protein kinases capable of activating c-jun expression include stress-activated protein kinase/c-Jun N-terminal protein kinase (SAPK/JNK) and p38 members of the mitogen-activated protein kinase (MAPK) superfamily of signaling molecules. To investigate the potential role of MAPKs in the regulation of c-jun by tumor hypoxia, we focused on the activation SAPK/JNKs in SiHa human squamous carcinoma cells. Here, we describe the transient activation of SAPK/JNKs by tumor-like hypoxia, and the concurrent transcriptional activation of MKP-1, a stress-inducible member of the MAPK phosphatase (MKP) family of dual specificity protein-tyrosine phosphatases. MKP-1 antagonizes SAPK/JNK activation in response to diverse environmental stresses. Together, these findings identify MKP-1 as a hypoxia-responsive gene and suggest a critical role in the regulation of SAPK/JNK activity in the tumor microenvironment.


Assuntos
Proteínas Quinases Dependentes de Cálcio-Calmodulina/antagonistas & inibidores , Proteínas de Ciclo Celular , Hipóxia Celular , Proteínas Imediatamente Precoces/metabolismo , Proteínas Quinases Ativadas por Mitógeno , Oxigênio/metabolismo , Fosfoproteínas Fosfatases , Proteínas Tirosina Fosfatases/metabolismo , Fator 2 Ativador da Transcrição , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Fosfatase 1 de Especificidade Dupla , Ativação Enzimática , Humanos , Proteínas Imediatamente Precoces/genética , Proteínas Quinases JNK Ativadas por Mitógeno , Fosforilação , Proteína Fosfatase 1 , Proteínas Tirosina Fosfatases/genética , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas c-jun/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fatores de Transcrição/metabolismo , Ativação Transcricional , Células Tumorais Cultivadas
7.
Cancer Res ; 59(6): 1315-22, 1999 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-10096565

RESUMO

Metallothioneins (MTs) are a family of stress-induced proteins with diverse physiological functions, including protection against metal toxicity and oxidants. They may also contribute to the regulation of cellular proliferation, apoptosis, and malignant progression. We reported previously that the human (h)MT-IIA isoform is induced in carcinoma cells (A431, SiHa, and HT29) exposed to low oxygen, conditions commonly found in solid tumors. The present study demonstrates that the genes for hMT-IIA and mouse (m)MT-I are transcriptionally activated by hypoxia through metal response elements (MREs) in their proximal promoter regions. These elements bind metal transcription factor-1 (MTF-1). Deletion and mutational analyses of the hMT-IIA promoter indicated that the hMRE-a element is essential for basal promoter activity and for induction by hypoxia, but that other elements contribute to the full transcriptional response. Functional studies of the mMT-I promoter demonstrated that at least two other MREs (mMRE-d and mMRE-c) are responsive to hypoxia. Multiple copies of either hMRE-a or mMRE-d conferred hypoxia responsiveness to a minimal MT promoter. Mouse MT-I gene transcripts in fibroblasts with targeted deletions of both MTF-1 alleles (MTF-1(-/-); dko7 cells) were not induced by zinc and showed low responsiveness to hypoxia. A transiently transfected MT promoter was unresponsive to hypoxia or zinc in dko7 cells, but inductions were restored by cotransfecting a mouse MTF-1 expression vector. Electrophoretic mobility shift assays detected a specific protein-DNA complex containing MTF-1 in nuclear extracts from hypoxic cells. Together, these results demonstrate that hypoxia activates MT gene expression through MREs and that this activation involves MTF-1.


Assuntos
Regulação Neoplásica da Expressão Gênica , Metalotioneína/genética , Oxigênio/metabolismo , Regiões Promotoras Genéticas , Fatores de Transcrição/metabolismo , Células 3T3 , Animais , Ligação Competitiva , Hipóxia Celular/genética , Proteínas de Ligação a DNA , Fibroblastos/metabolismo , Células HT29 , Humanos , Metalotioneína/biossíntese , Metais/metabolismo , Camundongos , Oxirredução , RNA Mensageiro/biossíntese , Fatores de Transcrição/genética , Células Tumorais Cultivadas , Fator MTF-1 de Transcrição
8.
Int J Cancer ; 78(6): 775-82, 1998 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-9833772

RESUMO

A significant portion of patients who present with non-muscle invasive "superficial" bladder cancer develop the muscle "invasive" life-threatening form of the disease during subsequent follow-up. In clinical studies, overexpression of the epidermal growth factor receptor (EGFR) and the p21 ras oncogene have been strongly associated with this phenotypic tumor transition. The marked difference in incidence of invasive bladder cancer in Asia compared to the United States has made us hypothesize that, among other factors, dietary influences have an impact on such tumor progression. A significantly higher dietary consumption of soy products exists in Asia and has led to the notion that the isoflavones present in this diet may contribute to a reduction in the number of invasive transitional cell bladder cancers. In this regard, we sought to determine the effect of genistein, a naturally occurring dietary protein tyrosine kinase (PTK) inhibitor, on the growth and motility of human bladder cancer cell lines with diverse EGFR and p21ras expression phenotypes and corresponding invasive behaviors. These effects were compared with those of tyrphostin, a pure synthetic EGFR inhibitor. Our results indicate that both genistein and tyrphostin are effective inhibitors of bladder cancer motility and growth, key factors in the development of muscle invasive disease. In addition, the growth and motility inhibitory effects of genistein and tyrphostin are observed preferentially in cells that overexpress the EGFR. Cells that have a mutated p21ras but do not overexpress the EGFR are less inhibited by these 2 compounds, suggesting that their effect is primarily directed at the EGFR signal transduction pathways proximal to the p21ras gene. Our results would seem to corroborate the notion that a high dietary intake of isoflavones is a likely explanation for the decreased incidence of invasive bladder cancer.


Assuntos
Inibidores Enzimáticos/farmacologia , Receptores ErbB/metabolismo , Genisteína/farmacologia , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Neoplasias da Bexiga Urinária/patologia , Movimento Celular , Expressão Gênica , Humanos , Pessoa de Meia-Idade , Invasividade Neoplásica , Fosforilação , Fatores de Tempo , Células Tumorais Cultivadas , Tirfostinas/farmacologia
9.
Cell Growth Differ ; 9(11): 919-28, 1998 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9831244

RESUMO

Although overexpression of the epidermal growth factor receptor (EGFR) has been strongly associated with the transition from superficial to invasive human bladder cancer, the exact molecular pathways by which this gene effectively triggers or facilitates the invasive process are not completely understood. Because enhanced cellular motility is a prerequisite for invasion, we chose to determine how EGFR signaling impacts cellular motility of human bladder cancer in vitro in a cell model of human bladder cancer that closely mimics the human disease. Using a stable antisense approach to diminish EGFR expression, we obtained data that support the role of EGFR in mediating bladder cancer motility. These results also demonstrate that EGFR plays an important role in bladder cancer motility, even in the presence of a mutated and overexpressing Ras protein, and suggest the possibility that Ras-independent EGFR motility signaling is a significant pathway used by bladder cancer cells. In support of this concept, using specific pharmacological inhibition of phosphatidylinositol 3-kinase, we show that this mediator is involved in EGFR motility signaling in this system. Knowledge of the pathways used by EGFR to induce motility and subsequent invasion may lead to development of methods to prevent or retard the progression of aggressive superficial bladder tumors.


Assuntos
Movimento Celular , Receptores ErbB/biossíntese , Fosfatidilinositol 3-Quinases/metabolismo , Neoplasias da Bexiga Urinária/metabolismo , Divisão Celular , Fator de Crescimento Epidérmico/farmacologia , Humanos , Invasividade Neoplásica , Oligonucleotídeos Antissenso , Fosforilação , Transfecção , Células Tumorais Cultivadas , Neoplasias da Bexiga Urinária/enzimologia
10.
Biochem Pharmacol ; 56(5): 623-33, 1998 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-9783731

RESUMO

We identified a structural analog of thyroid hormone, methyl-3,5-diiodo-4-(4'-methoxyphenoxy) benzoate (Oncocidin A1), that inhibits human carcinoma cell proliferation and the growth of human breast (MDA MB-231) and ovarian (OVCAR-3) carcinoma xenografts in nude mice. This novel antitumor agent is orally bioavailable and well tolerated by animals. Exposure of MCF-7 and MDA MB-231 breast carcinoma cells to Oncocidin A1 in vitro caused a cell-cycle arrest in prometaphase (a G2/M arrest) and apoptosis, suggesting a cytotoxic mechanism involving mitotic spindle function. The interaction of Oncocidin A1 with microtubules was demonstrated by: 1) immunofluorescence studies of microtubule assembly in the presence of the drug in cell-free and in cellular assays; and 2) in vitro binding inhibition studies involving radiolabeled Oncocidin A1 or colchicine and tubulin monomers. Taken together, these experiments indicate that Oncocidin A1 perturbs cellular microtubule assembly, possibly by binding to the colchicine site on tubulin. Three-dimensional structural modelling of Oncocidin A1 revealed that it can adopt a twisted conformation similar to that of combretastatin A-4, which binds to the colchicine site of tubulin. The novel structural features of Oncocidin A1 could guide the design of a new class of microtubule-binding antitumor agents having substantially reduced normal tissue toxicity upon oral administration.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Iodobenzoatos/uso terapêutico , Neoplasias Ovarianas/tratamento farmacológico , Éteres Fenílicos/uso terapêutico , Tubulina (Proteína)/metabolismo , Administração Oral , Animais , Antineoplásicos/metabolismo , Antineoplásicos/farmacocinética , Disponibilidade Biológica , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Divisão Celular/efeitos dos fármacos , Feminino , Humanos , Iodobenzoatos/metabolismo , Iodobenzoatos/farmacocinética , Metáfase/efeitos dos fármacos , Camundongos , Camundongos Nus , Mitose/efeitos dos fármacos , Transplante de Neoplasias , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Éteres Fenílicos/metabolismo , Éteres Fenílicos/farmacocinética , Ligação Proteica , Transplante Heterólogo
11.
Carcinogenesis ; 19(8): 1333-7, 1998 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9744525

RESUMO

Transient transfection studies of human HepG2 and mouse Hepa hepatocarcinoma cells with a reporter gene construct regulated by a human antioxidant responsive element (ARE) from the NQO1 gene demonstrated that the element is responsive to low oxygen conditions. The antioxidant N-acetyl L-cysteine (NAC) strongly inhibited basal aerobic reporter gene activity in HepG2 cells without obviously affecting the hypoxic induction, as is consistent with ARE sensitivity to oxidative stress in aerobic cultures. Electrophoretic mobility shift (EMS) assays of nuclear extracts of HepG2 and Hepa cells lysed under aerobic or hypoxic conditions or after exposure to the phenolic compound 3-(2)-tert-butyl-4-hydroxyanisole (BHA), showed specific and constitutive protein binding to the ARE under all of these conditions. Taken together, these findings show that the ARE can mediate gene expression in response to low oxygen conditions. Co-ordinately regulated expression of ARE-dependent genes, such as phase II detoxification enzymes, may be an important phenotype of solid tumors containing significant regions of pathophysiological hypoxia.


Assuntos
Cloranfenicol O-Acetiltransferase/metabolismo , Regulação Enzimológica da Expressão Gênica , NAD(P)H Desidrogenase (Quinona)/metabolismo , Transativadores/fisiologia , Acetilcisteína/farmacologia , Animais , Hidroxianisol Butilado/farmacologia , Hipóxia Celular , Cloranfenicol O-Acetiltransferase/antagonistas & inibidores , Cloranfenicol O-Acetiltransferase/genética , Sequestradores de Radicais Livres/farmacologia , Genes Reporter , Humanos , Camundongos , NAD(P)H Desidrogenase (Quinona)/genética , Oxirredução , Células Tumorais Cultivadas
12.
Blood ; 90(9): 3322-31, 1997 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-9345014

RESUMO

Tumor angiogenesis, the development of new blood vessels, is a highly regulated process that is controlled genetically by alterations in oncogene and tumor suppressor gene expression and physiologically by the tumor microenvironment. Previous studies indicate that the angiogenic switch in Ras-transformed cells may be physiologically promoted by the tumor microenvironment through the induction of the angiogenic mitogen, vascular endothelial growth factor (VEGF). In this report, we show Ras-transformed cells do not use the downstream effectors c-Raf-1 or mitogen activated protein kinases (MAPK) in signaling VEGF induction by hypoxia as overexpression of kinase-defective alleles of these genes does not inhibit VEGF induction under low oxygen conditions. In contrast to the c-Raf-1/MAP kinase pathway, hypoxia increases phosphatidylinositol 3-kinase (PI 3-kinase) activity in a Ras-dependent manner, and inhibition of PI 3-kinase activity genetically and pharmacologically results in inhibition of VEGF induction. We propose that hypoxia modulates VEGF induction in Ras-transformed cells through the activation of a stress inducible PI 3-kinase/Akt pathway and the hypoxia inducible factor-1 (HIF-1) transcriptional response element.


Assuntos
Transformação Celular Neoplásica , Proteínas de Ligação a DNA/fisiologia , Fatores de Crescimento Endotelial/fisiologia , Genes ras , Linfocinas/fisiologia , Neovascularização Patológica , Proteínas Nucleares/fisiologia , Fosfatidilinositol 3-Quinases/fisiologia , Transdução de Sinais , Células 3T3 , Animais , Hipóxia Celular , Regulação da Expressão Gênica , Fator 1 Induzível por Hipóxia , Subunidade alfa do Fator 1 Induzível por Hipóxia , Camundongos , Fatores de Transcrição/fisiologia , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
13.
Circ Res ; 80(3): 336-44, 1997 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-9048653

RESUMO

Hypoxia and reoxygenation are principal components of myocardial ischemia and reperfusion and have distinctive effects on the tissue. Both conditions have been associated with inflammation, necrosis, apoptosis, and myocardial infarction. Using a cell culture model of ischemia and reperfusion in which cardiac myocytes were exposed to cycles of hypoxia and reoxygenation, we report here that reoxygenation, but not hypoxia alone, caused sustained approximately 10-fold increases in phosphorylation of the amino-terminal domain of the c-jun transcription factor. The activation was similar to treatments with anisomycin or okadaic acid and correlated with the hypoxia-mediated depression of intracellular glutathione. Reoxygenation-induced c-Jun kinase activity was reduced by preincubating myocytes during the hypoxia phase with the spin-trap agent alpha-phenyl N-tert-butylnitrone or with N-acetylcysteine. The kinase activation was also inhibited by the tyrosine kinase inhibitor genistein but not by other protein kinase inhibitors. These results implicate unquenched reactive oxygen intermediates as the stimulus that initiates a kinase pathway involving the stress-activated protein kinases (JNKs/SAPKs) in reoxygenated cardiac myocytes.


Assuntos
Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Hipóxia Celular , Proteínas Quinases Ativadas por Mitógeno , Miocárdio/enzimologia , Oxirredução , Animais , Anisomicina/farmacologia , Antioxidantes/farmacologia , Western Blotting , Butionina Sulfoximina/farmacologia , Células Cultivadas , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Fibroblastos , Genisteína , Glutationa/análogos & derivados , Coração/efeitos dos fármacos , Isoflavonas/farmacologia , Proteínas Quinases JNK Ativadas por Mitógeno , Proteína Quinase 3 Ativada por Mitógeno , Ácido Okadáico/farmacologia , Oxigênio/metabolismo , Oxigênio/farmacologia , Inibidores da Síntese de Proteínas/farmacologia , Proteínas Tirosina Quinases/antagonistas & inibidores , Ratos , Espécies Reativas de Oxigênio/fisiologia , Fatores de Tempo
14.
Cancer Res ; 56(15): 3436-40, 1996 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-8758908

RESUMO

Hypoxia can select for cells that have lost their apoptotic potential, thereby making them resistant to adverse conditions. However, long-term survival of transformed cells which have diminished apoptotic sensitivity when exposed to low oxygen conditions would require the activation of their angiogenic program to compensate for an insufficient oxygen supply. In this report, we show that the activity (of oncogenic Ha-ras, either constitutively or transiently, enhances the induction of the angiogenic mitogen, vascular endothelial growth factor (VEGF), by hypoxia. Analysis of the 5' flanking region of the VEGF promoter indicates that a HIF-1-like sequence is to promote a 15-fold increase in reporter gene activity in Ha-ras-transformed cells when exposed to hypoxia, whereas mutations in the same site totally inhibited VEGF induction. Under low oxygen conditions, VEGF induction is inhibited in cells expressing a mutant inhibitory allele of Ha-ras (RasN17), indicating a direct role for Ras in modulating VEGF activity. We propose that the angiogenic switch in Ras-transformed cells may be physiologically promoted by the tumor microenvironment through VEGF induction.


Assuntos
Transformação Celular Neoplásica/genética , Fatores de Crescimento Endotelial/biossíntese , Genes ras , Linfocinas/biossíntese , Células 3T3/metabolismo , Células 3T3/fisiologia , Animais , Hipóxia Celular/fisiologia , Fatores de Crescimento Endotelial/genética , Expressão Gênica , Linfocinas/genética , Camundongos , Regiões Promotoras Genéticas , RNA Mensageiro/biossíntese , Ratos , Estresse Fisiológico/metabolismo , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular , Proteínas ras/biossíntese , Proteínas ras/fisiologia
15.
Int J Cancer ; 66(6): 772-8, 1996 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-8647648

RESUMO

The ATDC gene was originally identified by its ability to complement the radiosensitivity defect of an ataxia telangiectasia (AT) fibroblast cell line. Because hypersensitivity to ionizing radiation is an important feature of the AT phenotype, we reasoned that ATDC may function generally in the suppression of radiosensitivity. Previous work in our laboratory focused on radiosensitization mechanisms in human squamous carcinoma (SC) cells, especially A431 cells. To establish a basis for investigating the role of ATDC in radiation-responsive signaling pathways in human SC cells, we characterized ATDC message and protein expressions in A431 cells. ATDC message expression was also compared among human epidermoid cells (A431 cells, HaCaT spontaneously immortalized human keratinocytes and normal human epidermal keratinocytes) and a normal human fibroblast cell line (LM217). We made the following major observations: (i) the relative abundance of ATDC message is substantially higher in the epidermoid cells than in the fibroblast cell line, which has a message level comparable to those reported for other fibroblast lines; (ii) ATDC is constitutively phosphorylated on serine/threonine in A431 cells; (iii) in A431 cells, ATDC is a substrate for the serine/threonine protein kinase C (PKC) but not the epidermal growth factor (EGF) receptor tyrosine kinase; and (iv) EGF decreases ATDC message and protein expressions in A431 cells after a 24-hr exposure. The phosphorylation studies suggest that the ability of ATDC to modulate cellular radiosensitivity may be mediated in part through a PKC signaling pathway.


Assuntos
Carcinoma de Células Escamosas/metabolismo , Proteínas de Ligação a DNA/biossíntese , Fator de Crescimento Epidérmico/farmacologia , Regulação Neoplásica da Expressão Gênica , Proteínas de Neoplasias/biossíntese , Ataxia Telangiectasia/genética , Ataxia Telangiectasia/patologia , Sequência de Bases , Carcinoma de Células Escamosas/patologia , Linhagem Celular Transformada , Transformação Celular Viral , DNA Complementar/genética , Proteínas de Ligação a DNA/genética , Fibroblastos , Fase G1/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Queratinócitos , Dados de Sequência Molecular , Proteínas de Neoplasias/genética , Fosforilação , Proteína Quinase C/metabolismo , Processamento de Proteína Pós-Traducional , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , RNA Neoplásico/biossíntese , RNA Neoplásico/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes/farmacologia , Vírus 40 dos Símios/fisiologia , Pele/citologia , Fatores de Transcrição , Células Tumorais Cultivadas/efeitos dos fármacos
16.
Int J Radiat Oncol Biol Phys ; 34(1): 79-84, 1996 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-12118568

RESUMO

PURPOSE: To determine what structural moieties of the fused pyrazine mono-N-oxides are determining factors in their in vitro cytotoxicity and oncogenicity. METHODS AND MATERIALS: A new series of experimental bioreductive drugs, fused pyrazine mono-N-oxides, was evaluated in vitro for aerobic and hypoxic cytotoxicity in the HT29 human colon adenocarcinoma cell line by using clonogenic assays. The relative oncogenicities of these compounds were also determined in aerobic cultures of C3H 10T1/2 mouse embryo fibroblasts by using a standard transformation assay. RESULTS: Removal of the 4-methyl piperazine side chain from the parent compound, RB 90740, reduced the potency of the hypoxic cytotoxin. Reduction of the N-oxide function increased the aerobic cytotoxicity and eliminated most of the hypoxic/aerobic cytotoxic differential. The reduced N-oxide also had significant oncogenicity, consistent with a mechanism of genotoxicity following bioreduction of RB 90740. CONCLUSION: This new series of bioreductive compounds may be effective in cancer therapy, particularly the lead compound RB 90740. The oncogenic potential of these compounds is similar to that for other cancer therapies. Further studies should include evaluation of these compounds in vivo and the development of analogs with reduced oncogenic potential and retention of the hypoxic/aerobic cytotoxicity differential.


Assuntos
Antineoplásicos/efeitos adversos , Pirazinas/efeitos adversos , Animais , Hipóxia Celular/efeitos dos fármacos , Transformação Celular Neoplásica/induzido quimicamente , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Células HT29/efeitos dos fármacos , Humanos , Camundongos , Camundongos Endogâmicos C3H , Radiobiologia , Relação Estrutura-Atividade , Células Tumorais Cultivadas/efeitos dos fármacos
17.
Cancer Res ; 55(24): 6222-6, 1995 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-8521417

RESUMO

We have investigated the hypoxia inducibility of vascular endothelial growth factor (VEGF) in multicellular tumor spheroids of HT29 cells using a monoclonal antibody to a fluorinated bioreductive drug, EF5 [2-(2-nitro-1H-imidazol-1-yl)-N-(2,2,3,3,3-pentafluoropropyl)aceta mide], a chemical probe for hypoxia. We have shown that VEGF expression is predominantly localized in interior spheroid cells that are sufficiently hypoxic to bioreductively activate the 2-nitroimidazole and produce immunologically detectable adducts of the EF5 compound. Northern blotting analyses demonstrated that VEGF165 is the predominant form of VEGF produced by HT29 cells and that the phorbol ester 12-O-tetradecanoyl-phorbol-13-acetate did not induce VEGF expression. This study demonstrates that VEGF expression is up-regulated in response to hypoxia and in the microenvironments found in human multicellular tumor spheroids. This investigation also illustrates the utility of the EF5 binding in multi-cellular tumor spheroids as a means of studying the expression and regulation of hypoxia-inducible genes.


Assuntos
Carcinoma/irrigação sanguínea , Neoplasias do Colo/irrigação sanguínea , Fatores de Crescimento Endotelial/genética , Linfocinas/genética , Neovascularização Patológica , Etanidazol/análogos & derivados , Regulação Neoplásica da Expressão Gênica , Humanos , Hidrocarbonetos Fluorados , Hipóxia/metabolismo , Hibridização In Situ , Indicadores e Reagentes , Organoides , RNA Mensageiro/genética , RNA Neoplásico/genética , Células Tumorais Cultivadas , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
18.
Cancer Res ; 54(22): 5808-10, 1994 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-7954405

RESUMO

The expression of metallothionein IIA (MT-IIA) was investigated in A431 human squamous carcinoma cells exposed to hypoxia (pO < or = 0.01% of atmospheric pO2) and subsequent reoxygenation. Northern analysis showed that MT-IIA mRNA levels were significantly increased during 14 h of hypoxia and during reoxygenation. Western blotting confirmed that total MT protein levels were also increased in response to these stresses. Evidence of the transcriptional control of MT-IIA expression in hypoxic and in reoxygenated A431 cells was found using a 0.2-kilobase sequence of the proximal 5'-regulatory region of the MT-IIA gene in a chloramphenicol acetyltransferase reporter gene construct. Thus the proximal promoter of the human MT-IIA gene appears to contain a hypoxic response element(s). These observations indicate that MT-IIA may have an important role in the stress responses of cells in solid tumors.


Assuntos
Carcinoma de Células Escamosas/metabolismo , Hipóxia Celular , Metalotioneína/metabolismo , Regulação para Cima , Carcinoma de Células Escamosas/genética , Cloranfenicol O-Acetiltransferase/metabolismo , Proteínas de Choque Térmico HSP70 , Humanos , Metalotioneína/genética , Proteínas/metabolismo , RNA Mensageiro/metabolismo , RNA Neoplásico/metabolismo , Fatores de Tempo , Transfecção , Células Tumorais Cultivadas
19.
Mol Cell Biol ; 14(8): 5032-42, 1994 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-8035787

RESUMO

Hypoxic stress in tumor cells has been implicated in malignant progression and in the development of therapeutic resistance. We have investigated the effects of acute hypoxic exposure on regulation of the proto-oncogene c-jun in SiHa cells, a human squamous carcinoma cell line. Hypoxic exposure produced increased levels of c-jun mRNA resulting from both message stabilization and transcriptional activation. A superinduction of c-jun message resulted during simultaneous oxygen and glucose deprivation, with several characteristics of an induction mediated by oxidative-stress pathways. This superinduction was blocked by preincubation of cells with the glutathione precursor N-acetyl cysteine or with phorbol 12-myristate 13-acetate, which indicates redox control of c-jun expression and probable involvement of protein kinase C. By gel retardation assay, no increase in AP-1 DNA binding activity was found to be concomitant with the transcriptional activation of c-jun. A lack of increased DNA binding was observed for the consensus AP-1 sequence and for the two AP-1 sequence variants found within the c-Jun promoter. Additionally, hypoxic and low-glucose stress produced no activation of stably transfected AP-1 reporter sequences. Taken together, these results indicate that the transcriptional activation of c-jun during hypoxic and low-glucose stress involves redox control and is unlikely to be mediated by AP-1 recognition elements within the c-jun promoter.


Assuntos
Genes jun , Glucose/metabolismo , Hipóxia/genética , Acetilcisteína/farmacologia , Sequência de Bases , Regulação da Expressão Gênica , Genes fos , Humanos , Dados de Sequência Molecular , Oligodesoxirribonucleotídeos/química , Proteína Quinase C/metabolismo , Proto-Oncogene Mas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transcrição Gênica
20.
Cancer Res ; 54(11): 2845-7, 1994 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-8187065

RESUMO

This study examined the efficacy of combining cyclophosphamide and the hypoxic cytotoxin, tirapazamine, in the treatment of human breast cancer xenografts grown in nude mice. A single dose of tirapazamine was followed 2 h later by a single dose of cyclophosphamide. As determined from tumor regrowth delay, the effectiveness of combined therapy was greater than the additive effects of each treatment given alone. Possible mechanisms of this synergistic interaction include enhancement of DNA damage, inhibition of repair of DNA damage, or induction of apoptosis. Apart from some loss in body weight, the only other toxicity of interest in mice treated with tirapazamine was necrosis of the skin on the distal tail, which appeared to be vascular in origin.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Animais , Peso Corporal/efeitos dos fármacos , Ciclofosfamida/administração & dosagem , Ensaios de Seleção de Medicamentos Antitumorais , Sinergismo Farmacológico , Feminino , Humanos , Camundongos , Camundongos Nus , Cauda/efeitos dos fármacos , Tirapazamina , Transplante Heterólogo , Triazinas/administração & dosagem , Triazinas/efeitos adversos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...