Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 111(49): 17606-11, 2014 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-25422431

RESUMO

Tamoxifen is effective for treating estrogen receptor-alpha (ER) positive breast cancers. However, few molecular mediators of tamoxifen resistance have been elucidated. Here we describe a previously unidentified gene, MACROD2 that confers tamoxifen resistance and estrogen independent growth. We found MACROD2 is amplified and overexpressed in metastatic tamoxifen-resistant tumors. Transgene overexpression of MACROD2 in breast cancer cell lines results in tamoxifen resistance, whereas RNAi-mediated gene knock down reverses this phenotype. MACROD2 overexpression also leads to estrogen independent growth in xenograft assays. Mechanistically, MACROD2 increases p300 binding to estrogen response elements in a subset of ER regulated genes. Primary breast cancers and matched metastases demonstrate MACROD2 expression can change with disease evolution, and increased expression and amplification of MACROD2 in primary tumors is associated with worse overall survival. These studies establish MACROD2 as a key mediator of estrogen independent growth and tamoxifen resistance, as well as a potential novel target for diagnostics and therapy.


Assuntos
Neoplasias da Mama/metabolismo , Enzimas Reparadoras do DNA/metabolismo , Resistencia a Medicamentos Antineoplásicos , Estrogênios/metabolismo , Hidrolases/metabolismo , Tamoxifeno/farmacologia , Sequência de Bases , Linhagem Celular Tumoral , Proliferação de Células , Epigênese Genética , Feminino , Deleção de Genes , Dosagem de Genes , Humanos , Dados de Sequência Molecular , Transplante de Neoplasias , Fenótipo , Prognóstico , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Receptores de Estrogênio/metabolismo , Transgenes , Resultado do Tratamento
2.
Proteomics ; 15(2-3): 318-26, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25367220

RESUMO

The PIK3CA gene encodes for the p110 alpha isoform of PI3 kinase and is one of the most frequently mutated oncogenes in human cancers. However, the mechanisms by which PIK3CA mutations activate cell signaling are not fully understood. Here we used a phosphoproteomic approach to compare differential phosphorylation patterns between human breast epithelial cells and two isogenic somatic cell knock in derivatives, each harboring a distinct PIK3CA mutation. We demonstrated differential phosphorylation patterns between isogenic cell lines containing a PIK3CA helical domain mutation (E545K) compared to cells with a PIK3CA kinase domain mutation (H1047R). In particular, the receptor tyrosine kinase, HER3, showed increased phosphorylation at tyrosine 1328 in H1047R cells versus E545K cells. Genetic studies using shRNA demonstrated that H1047R cells have a profound decrease in growth factor independent proliferation upon HER3 knock down, but this effect was attenuated in E545K cells. In addition, HER3 knock down led to reductions in both PI3 kinase and MAP kinase pathway activation in H1047R cells, but in E545K cells only PI3 kinase pathway diminution was observed. These studies demonstrate the power of using paired isogenic cell lines for proteomic analysis to gain new insights into oncogenic signal transduction pathways.


Assuntos
Neoplasias da Mama/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteômica , Receptor ErbB-3/metabolismo , Transdução de Sinais , Neoplasias da Mama/genética , Linhagem Celular , Linhagem Celular Tumoral , Proliferação de Células , Classe I de Fosfatidilinositol 3-Quinases , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Mutação , Fosfatidilinositol 3-Quinases/química , Fosfatidilinositol 3-Quinases/genética , Fosforilação , Estrutura Terciária de Proteína , Interferência de RNA , Receptor ErbB-3/genética
3.
J Proteome Res ; 14(9): 3882-3891, 2015 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-26267517

RESUMO

The human oncogene PIK3CA is frequently mutated in human cancers. Two hotspot mutations in PIK3CA, E545K and H1047R, have been shown to regulate widespread signaling events downstream of AKT, leading to increased cell proliferation, growth, survival, and motility. We used quantitative mass spectrometry to profile the global phosphotyrosine proteome of isogenic knock-in cell lines containing these activating mutations, where we identified 824 unique phosphopeptides. Although it is well understood that these mutations result in hyperactivation of the serine/threonine kinase AKT, we found a surprisingly widespread modulation of tyrosine phosphorylation levels of proteins in the mutant cells. In the tyrosine kinome alone, 29 tyrosine kinases were altered in their phosphorylation status. Many of the regulated phosphosites that we identified were located in the kinase domain or the canonical activation sites, indicating that these kinases and their downstream signaling pathways were activated. Our study demonstrates that there is frequent and unexpected cross-talk that occurs between tyrosine signaling pathways and serine/threonine signaling pathways activated by the canonical PI3K-AKT axis.


Assuntos
Fosfatidilinositol 3-Quinases/genética , Fosfoproteínas/genética , Proteoma/genética , Transdução de Sinais/genética , Tirosina/metabolismo , Linhagem Celular Tumoral , Classe I de Fosfatidilinositol 3-Quinases , Humanos , Fosfatidilinositol 3-Quinases/química , Fosfatidilinositol 3-Quinases/metabolismo , Fosfoproteínas/análise , Fosfoproteínas/química , Fosfoproteínas/metabolismo , Fosforilação , Proteoma/análise , Proteoma/química , Proteoma/metabolismo , Proteômica , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo
4.
J Pathol ; 232(2): 244-54, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24114654

RESUMO

Recent advances in genetics and genomics have revealed new genes and pathways that are somatically altered in human malignancies. This wealth of knowledge has translated into molecularly defined targets for therapy over the past two decades, serving as key examples that translation of laboratory findings can have great impact on the ability to treat patients with cancer. However, given the genetic instability and heterogeneity that are characteristic of all human cancers, drug resistance to virtually all therapies has emerged, posing further and future challenges for clinical oncology. Here we review the history of targeted therapies, including examples of genetically defined cancer targets and their approved therapies. We also discuss resistance mechanisms that have been uncovered, with an emphasis on somatic genetic alterations that lead to these phenotypes.


Assuntos
Antineoplásicos/uso terapêutico , Biomarcadores Tumorais/genética , Resistencia a Medicamentos Antineoplásicos/genética , Terapia de Alvo Molecular , Neoplasias/tratamento farmacológico , Neoplasias/genética , Animais , Descoberta de Drogas , Predisposição Genética para Doença , Humanos , Neoplasias/metabolismo , Neoplasias/patologia , Fenótipo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética
5.
Mol Pharmacol ; 79(1): 157-66, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20930109

RESUMO

Copper transporter 2 (CTR2) is one of the four copper transporters in mammalian cells that influence the cellular pharmacology of cisplatin and carboplatin. CTR2 was knocked down using a short hairpin RNA interference. Robust expression of CTR2 was observed in parental tumors grown in vivo, whereas no staining was found in the tumors formed from cells in which CTR2 had been knocked down. Knockdown of CTR2 reduced growth rate by 5.8-fold, increased the frequency of apoptotic cells, and decreased the vascular density, but it did not change copper content. Knockdown of CTR2 increased the tumor accumulation of cis-diamminedichloroplatinum(II) [cisplatin (cDDP)] by 9.1-fold and greatly increased its therapeutic efficacy. Because altered endocytosis has been implicated in cDDP resistance, uptake of dextran was used to quantify the rate of macropinocytosis. Knockdown of CTR2 increased dextran uptake 2.5-fold without reducing exocytosis. Inhibition of macropinocytosis with either amiloride or wortmannin blocked the increase in macropinocytosis mediated by CTR2 knockdown. Stimulation of macropinocytosis by platelet-derived growth factor coordinately increased dextran and cDDP uptake. Knockdown of CTR2 was associated with activation of the Rac1 and cdc42 GTPases that control macropinocytosis but not activation of the phosphoinositide-3 kinase pathway. We conclude that CTR2 is required for optimal tumor growth and that it is an unusually strong regulator of cisplatin accumulation and cytotoxicity. CTR2 regulates the transport of cDDP in part through control of the rate of macropinocytosis via activation of Rac1 and cdc42. Selective knockdown of CTR2 in tumors offers a strategy for enhancing the efficacy of cDDP.


Assuntos
Proteínas de Transporte de Cátions/fisiologia , Cisplatino/metabolismo , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Pinocitose/fisiologia , Animais , Linhagem Celular , Cisplatino/uso terapêutico , Feminino , Técnicas de Silenciamento de Genes , Camundongos , Camundongos Knockout , Camundongos Nus , Neoplasias Experimentais/tratamento farmacológico , Proteínas SLC31 , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Proc Natl Acad Sci U S A ; 105(1): 288-93, 2008 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-18162533

RESUMO

Tamoxifen is widely used for the treatment of hormonally responsive breast cancers. However, some resistant breast cancers develop a growth proliferative response to this drug, as evidenced by tumor regression upon its withdrawal. To elucidate the molecular mediators of this paradox, tissue samples from a patient with tamoxifen-stimulated breast cancer were analyzed. These studies revealed that loss of the cyclin-dependent kinase inhibitor p21 was associated with a tamoxifen growth-inducing phenotype. Immortalized human breast epithelial cells with somatic deletion of the p21 gene were then generated and displayed a growth proliferative response to tamoxifen, whereas p21 wild-type cells demonstrated growth inhibition upon tamoxifen exposure. Mutational and biochemical analyses revealed that loss of p21's cyclin-dependent kinase inhibitory property results in hyperphosphorylation of estrogen receptor-alpha, with subsequent increased gene expression of estrogen receptor-regulated genes. These data reveal a previously uncharacterized molecular mechanism of tamoxifen resistance and have potential clinical implications for the management of tamoxifen-resistant breast cancers.


Assuntos
Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Receptor alfa de Estrogênio/metabolismo , Tamoxifeno/farmacologia , Linhagem Celular Tumoral , Proliferação de Células , Metilação de DNA , Análise Mutacional de DNA , Resistencia a Medicamentos Antineoplásicos/genética , Feminino , Humanos , Pessoa de Meia-Idade , Moduladores Seletivos de Receptor Estrogênico/farmacologia , Resultado do Tratamento
7.
Mol Pharmacol ; 78(3): 333-9, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20519567

RESUMO

Mammalian copper transporter 1 (CTR1) is a high-affinity copper influx transporter that also mediates the uptake of platinum-containing chemotherapeutic agents including cisplatin (cDDP). Methionines 150, 154, and histidine 139 have been proposed to form a series of stacked rings in the pore formed by the CTR1 homotrimer, each of which is required for maximal copper transport. To examine the mechanism by which hCTR1 also transports cDDP, variant forms of hCTR1 in which methionines 150 and 154 were converted to isoleucines or in which histidine 139 was converted to alanine were re-expressed in cells in which both alleles of CTR1 had been knocked out. Each of these conversions disabled copper transport and increased cellular resistance to the cytotoxic effect of copper. In contrast, conversion of the methionines increased the uptake and cytotoxicity of cDDP well above that attained with wild-type hCTR1. Conversion of His139 to alanine did not impair cDDP uptake and actually enhanced cytotoxicity. Thus, although Met150 and Met154 facilitate the movement of copper through the pore, they serve to obstruct the passage of cDDP. None of the modifications altered the ability of cDDP to trigger the degradation of hCTR1, indicating that cDDP must interact with hCTR1 at other sites as well. Although both copper and cDDP may rely on a series of transchelation reactions to pass through the hCTR1 trimeric complex, the details of the molecular interactions must be different, which provides a potential basis for selective pharmacological modulation of copper versus cDDP cytotoxicity.


Assuntos
Cisplatino/metabolismo , Animais , Antineoplásicos/farmacologia , Transporte Biológico/efeitos dos fármacos , Proteínas de Transporte de Cátions , Células/metabolismo , Cobre/metabolismo , Cobre/farmacologia , Transportador de Cobre 1 , Interações Medicamentosas , Histidina/metabolismo , Histidina/farmacologia , Mamíferos/metabolismo , Metionina/metabolismo , Metionina/farmacologia , Camundongos , Camundongos Knockout , Platina/farmacologia
8.
Mol Pharmacol ; 77(6): 912-21, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20194531

RESUMO

Down-regulation of copper transporter 1 (CTR1) reduces uptake and sensitivity, whereas down-regulation of CTR2 enhances both. Cisplatin (DDP) triggers the rapid degradation of CTR1 and thus limits its own accumulation. We sought to determine the effect of DDP and copper on the expression of CTR2. Changes in CTR1 and CTR2 mRNA and protein levels in human ovarian carcinoma 2008 cells and ATOX1(+/+) and ATOX1(-/-) mouse embryo fibroblasts in response to exposure to DDP and copper were measured by quantitative reverse transcriptase-polymerase chain reaction, Western blot analysis, and deconvolution microscopy. DDP triggered rapid degradation of CTR1 in 2008 human ovarian cancer cells. However, it increased the expression of CTR2 mRNA and protein levels. Expression of CTR2 was heavily modulated by changes in intracellular copper concentration; copper depletion produced rapid disappearance of CTR2, whereas excess copper increased the level of CTR2 protein. This increase was associated with an increase in CTR2 mRNA and prolongation of the CTR2 half-life. Consistent with prior observations that short hairpin RNA interference-mediated knockdown of CTR2 enhanced DDP uptake and tumor cell kill, reduction of CTR2 by copper starvation also enhanced DDP uptake and cytotoxicity. Comparison of the ability of copper and DDP to modulate the expression of CTR1 in ATOX1(+/+) and ATOX1(-/-) indicated that ATOX1 participates in the regulation of CTR2 expression. Unlike CTR1, the expression of CTR2 is increased rather than decreased by DDP. Therefore, these two copper transporters have opposite effects on DDP sensitivity. CTR2 expression is regulated by copper availability via the copper-dependent regulator ATOX1.


Assuntos
Antineoplásicos/farmacologia , Proteínas de Transporte de Cátions/farmacologia , Cisplatino/farmacologia , Cobre/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Neoplasias Ovarianas/genética , Animais , Western Blotting , Proteínas de Transporte de Cátions/fisiologia , Linhagem Celular Tumoral , Células Cultivadas , Proteínas de Transporte de Cobre , Feminino , Regulação Neoplásica da Expressão Gênica/fisiologia , Humanos , Metalochaperonas , Camundongos , Chaperonas Moleculares/fisiologia , Neoplasias Ovarianas/patologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas SLC31
9.
Mol Pharmacol ; 75(2): 324-30, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18996970

RESUMO

The mammalian copper transporter 1 (CTR1) is responsible for the uptake of copper from the extracellular space. In this study, we used an isogenic pair of CTR1(+/+) and CTR1(-/-) mouse embryo fibroblasts to examine the contribution of CTR1 to the influx of cisplatin (DDP), carboplatin (CBDCA), oxaliplatin (L-OHP), and transplatin. Exposure to DDP triggered the rapid degradation of CTR1, suggesting that its contribution to influx was likely to be on the initial phase of drug entry. Loss of CTR1 decreased the initial binding of DDP to cells and reduced influx measured over the first 5 min of drug exposure by 81%. Loss of CTR1 almost completely eliminated the initial influx of CBDCA and reduced the initial uptake of L-OHP by 68% but had no effect on the influx of transplatin. Loss of CTR1 rendered cells resistant to even high concentrations of DDP when measured in vitro, and re-expression of CTR1 in the CTR1(-/-) cells restored both DDP uptake and cytotoxicity. The growth of CTR1(-/-) tumor xenografts in which CTR1 levels were restored by infection with a lentivirus expressing wild-type CTR1 was reduced by a single maximum tolerated dose of DDP in vivo, whereas the CTR1(-/-) xenografts failed to respond at all. We conclude that CTR1 mediates the initial influx of DDP, CBDCA, and L-OHP and is a major determinant of responsiveness to DDP both in vitro and in vivo.


Assuntos
Proteínas de Transporte de Cátions/fisiologia , Platina/farmacocinética , Animais , Carboplatina/farmacocinética , Linhagem Celular Tumoral , Células Cultivadas , Cisplatino/farmacocinética , Transportador de Cobre 1 , Camundongos , Compostos Organoplatínicos/farmacocinética , Oxaliplatina , Platina/química
10.
Cancer Biol Ther ; 3(2): 221-5, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14726675

RESUMO

We investigated the mechanism by which cancers evade the growth inhibitory effects of TGF-beta. Using two p21-/- somatically deleted human epithelial cell lines, we find that TGF-beta serves as a growth stimulator rather than a growth suppressor to cells lacking p21. In addition, TGF-beta stimulated p21-/- cells exhibited a mesenchymal phenotype, demonstrated by an upregulation of vimentin and decreased expression of E-cadherin. Analysis of primary human breast cancers by immunohistochemical labeling confirmed a correlation between p21 loss and positive vimentin expression. These data provide a molecular mechanism explaining how nongastrointestinal cancers can escape the anti-proliferative effects of this cytokine and simultaneously use this pathway for growth advantage.


Assuntos
Neoplasias da Mama/patologia , Neoplasias do Colo/patologia , Ciclinas/metabolismo , Células Epiteliais/efeitos dos fármacos , Fator de Crescimento Transformador beta/farmacologia , Neoplasias da Mama/metabolismo , Caderinas/metabolismo , Divisão Celular , Neoplasias do Colo/metabolismo , Inibidor de Quinase Dependente de Ciclina p21 , Inibidores Enzimáticos/metabolismo , Células Epiteliais/citologia , Feminino , Regulação da Expressão Gênica , Humanos , Células Tumorais Cultivadas , Vimentina/metabolismo
11.
Cancer Biol Ther ; 3(8): 772-5, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15254419

RESUMO

The phosphatidylinositol 3-kinases (PI3Ks) are known regulators of cellular growth and proliferation. It has recently been reported that somatic mutations within the PI3K subunit p110alpha (PIK3CA) are present in human colorectal and other cancers. Here we show that thirteen of fifty-three breast cancers (25%) contain somatic mutations in PIK3CA, with the majority of mutations located in the kinase domain. These results demonstrate that PIK3CA is the most mutated oncogene in breast cancer and support a role for PIK3CA in epithelial carcinogenesis.


Assuntos
Biomarcadores Tumorais/genética , Neoplasias da Mama/genética , Regulação Enzimológica da Expressão Gênica , Mutação de Sentido Incorreto , Fosfatidilinositol 3-Quinases/genética , Biomarcadores Tumorais/metabolismo , Mama/metabolismo , Neoplasias da Mama/enzimologia , Classe I de Fosfatidilinositol 3-Quinases , Éxons/genética , Feminino , Amplificação de Genes , Humanos , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , Fosfatidilinositol 3-Quinases/metabolismo , Células Tumorais Cultivadas
12.
Clin Cancer Res ; 20(10): 2643-2650, 2014 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-24504125

RESUMO

PURPOSE: Detecting circulating plasma tumor DNA (ptDNA) in patients with early-stage cancer has the potential to change how oncologists recommend systemic therapies for solid tumors after surgery. Droplet digital polymerase chain reaction (ddPCR) is a novel sensitive and specific platform for mutation detection. EXPERIMENTAL DESIGN: In this prospective study, primary breast tumors and matched pre- and postsurgery blood samples were collected from patients with early-stage breast cancer (n = 29). Tumors (n = 30) were analyzed by Sanger sequencing for common PIK3CA mutations, and DNA from these tumors and matched plasma were then analyzed for PIK3CA mutations using ddPCR. RESULTS: Sequencing of tumors identified seven PIK3CA exon 20 mutations (H1047R) and three exon 9 mutations (E545K). Analysis of tumors by ddPCR confirmed these mutations and identified five additional mutations. Presurgery plasma samples (n = 29) were then analyzed for PIK3CA mutations using ddPCR. Of the 15 PIK3CA mutations detected in tumors by ddPCR, 14 of the corresponding mutations were detected in presurgical ptDNA, whereas no mutations were found in plasma from patients with PIK3CA wild-type tumors (sensitivity 93.3%, specificity 100%). Ten patients with mutation-positive ptDNA presurgery had ddPCR analysis of postsurgery plasma, with five patients having detectable ptDNA postsurgery. CONCLUSIONS: This prospective study demonstrates accurate mutation detection in tumor tissues using ddPCR, and that ptDNA can be detected in blood before and after surgery in patients with early-stage breast cancer. Future studies can now address whether ptDNA detected after surgery identifies patients at risk for recurrence, which could guide chemotherapy decisions for individual patients.


Assuntos
Neoplasias da Mama/diagnóstico , Neoplasias da Mama/genética , DNA de Neoplasias/sangue , DNA de Neoplasias/genética , Adulto , Idoso , Neoplasias da Mama/cirurgia , Classe I de Fosfatidilinositol 3-Quinases , Análise Mutacional de DNA/métodos , DNA de Neoplasias/química , Éxons/genética , Feminino , Humanos , Pessoa de Meia-Idade , Mutação , Estadiamento de Neoplasias , Fosfatidilinositol 3-Quinases/genética , Reação em Cadeia da Polimerase/métodos , Período Pós-Operatório , Período Pré-Operatório , Estudos Prospectivos , Reprodutibilidade dos Testes , Sensibilidade e Especificidade
13.
Cancer Res ; 73(11): 3248-61, 2013 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-23580570

RESUMO

The selective pressures leading to cancers with mutations in both KRAS and PIK3CA are unclear. Here, we show that somatic cell knockin of both KRAS G12V and oncogenic PIK3CA mutations in human breast epithelial cells results in cooperative activation of the phosphoinositide 3-kinase (PI3K) and mitogen-activated protein kinase (MAPK) pathways in vitro, and leads to tumor formation in immunocompromised mice. Xenografts from double-knockin cells retain single copies of mutant KRAS and PIK3CA, suggesting that tumor formation does not require increased copy number of either oncogene, and these results were also observed in human colorectal cancer specimens. Mechanistically, the cooperativity between mutant KRAS and PIK3CA is mediated in part by Ras/p110α binding, as inactivating point mutations within the Ras-binding domain of PIK3CA significantly abates pathway signaling. In addition, Pdk1 activation of the downstream effector p90RSK is also increased by the combined presence of mutant KRAS and PIK3CA. These results provide new insights into mutant KRAS function and its role in carcinogenesis.


Assuntos
Neoplasias da Mama/genética , Transformação Celular Neoplásica/genética , Células Epiteliais/patologia , Fosfatidilinositol 3-Quinases/genética , Proteínas Proto-Oncogênicas/genética , Proteínas ras/genética , Animais , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Neoplasias da Mama/enzimologia , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Processos de Crescimento Celular/fisiologia , Transformação Celular Neoplásica/patologia , Classe I de Fosfatidilinositol 3-Quinases , Células Epiteliais/enzimologia , Células Epiteliais/metabolismo , Feminino , Técnicas de Introdução de Genes , Xenoenxertos , Humanos , Hospedeiro Imunocomprometido , Sistema de Sinalização das MAP Quinases , Camundongos , Camundongos Nus , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Mutação Puntual , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas p21(ras) , Piruvato Desidrogenase Quinase de Transferência de Acetil , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Proteínas Quinases S6 Ribossômicas 90-kDa/metabolismo , Proteínas ras/metabolismo
14.
Biochem Pharmacol ; 80(4): 448-54, 2010 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-20451502

RESUMO

The mammalian copper transporter 1 (CTR1) is responsible for the uptake of copper (Cu) from the extracellular space, and has been shown to play a major role in the initial accumulation of platinum-based drugs. In this study we re-expressed wild type and structural variants of hCTR1 in mouse embryo fibroblasts in which both alleles of mCTR1 had been knocked out (CTR1(-/-)) to examine the role of the N-terminal extracellular domain of hCTR1 in the accumulation of cisplatin (cDDP). Deletion of either the first 45 amino acids or just the (40)MXXM(45) motif in the N-terminal domain did not alter subcellular distribution or the amount of protein in the plasma membrane but it eliminated the ability of hCTR1 to mediate the uptake of Cu. In contrast it only partially reduced cDDP transport capacity. Neither of these structural changes prevented cDDP from triggering the rapid degradation of hCTR1. However, they did alter the potency of the cDDP that achieved cell entry, possibly reflecting the fact that hCTR1 may mediate the transport of cDDP both through the pore it forms in the plasma membrane and via endocytosis. We conclude that cDDP interacts with hCTR1 both at (40)MXXM(45) and at sites outside the N-terminal domain that produce the conformational changes that trigger degradation.


Assuntos
Transporte Biológico/efeitos dos fármacos , Proteínas de Transporte de Cátions/fisiologia , Células Cultivadas/metabolismo , Cisplatino/farmacocinética , Animais , Transporte Biológico/fisiologia , Proteínas de Transporte de Cátions/química , Linhagem Celular , Cobre/farmacologia , Transportador de Cobre 1 , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Humanos , Camundongos
15.
Clin Cancer Res ; 15(13): 4312-21, 2009 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-19509135

RESUMO

PURPOSE: Copper transporter 2 (CTR2) is known to mediate the uptake of Cu(+1) by mammalian cells. Several other Cu transporters, including the influx transporter CTR1 and the two efflux transporters ATP7A and ATP7B, also regulate sensitivity to the platinum-containing drugs. We sought to determine the effect of CTR2 on influx, intracellular trafficking, and efflux of cisplatin and carboplatin. EXPERIMENTAL DESIGN: The role of CTR2 was examined by knocking down CTR2 expression in an isogenic pair of mouse embryo fibroblasts consisting of a CTR1(+/+) line and a CTR1(-/-) line in which both CTR1 alleles had been deleted. CTR2 levels were determined by quantitative reverse transcription-PCR and Western blot analysis. Cisplatin (DDP) was quantified by inductively coupled plasma mass spectrometry and (64)Cu and [(14)C]carboplatin (CBDCA) accumulation by gamma and scintillation counting. RESULTS: Deletion of CTR1 reduced the uptake of Cu, DDP, and CBDCA and increased resistance to their cytotoxic effects by 2- to 3-fold. Knockdown of CTR2 increased uptake of Cu only in the CTR1(+/+) cells. In contrast, knockdown of CTR2 increased whole-cell DDP uptake and DNA platination in both CTR1(+/+) and CTR1(-/-) cells and proportionately enhanced cytotoxicity while producing no effect on vesicular accumulation or efflux. A significant correlation was found between CTR2 mRNA and protein levels and sensitivity to DDP in a panel of six ovarian carcinoma cell lines. CONCLUSIONS: CTR2 is a major determinant of sensitivity to the cytotoxic effects of DDP and CBDCA. CTR2 functions by limiting drug accumulation, and its expression correlates with the sensitivity of human ovarian carcinoma cell lines to DDP.


Assuntos
Carboplatina/farmacocinética , Proteínas de Transporte de Cátions/fisiologia , Cisplatino/farmacocinética , Citotoxinas/farmacocinética , Animais , Carboplatina/farmacologia , Carboplatina/uso terapêutico , Carcinoma/tratamento farmacológico , Carcinoma/genética , Carcinoma/patologia , Proteínas de Transporte de Cátions/genética , Proteínas de Transporte de Cátions/metabolismo , Células Cultivadas , Cisplatino/farmacologia , Cisplatino/uso terapêutico , Citotoxinas/farmacologia , Citotoxinas/uso terapêutico , Adutos de DNA/efeitos dos fármacos , Adutos de DNA/metabolismo , Resistencia a Medicamentos Antineoplásicos/genética , Feminino , Fibroblastos/metabolismo , Técnicas de Silenciamento de Genes , Humanos , Camundongos , Camundongos Knockout , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia , Proteínas SLC31 , Distribuição Tecidual/genética
16.
J Inorg Biochem ; 103(3): 333-41, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19124158

RESUMO

Previous work has demonstrated that the copper (Cu) transporters Ctr1, Atp7a and Atp7b regulate the cellular pharmacology of cisplatin (CDDP) by mediating its uptake and efflux. It was also shown that, in the process of uptake by Ctr1, CDDP triggers the rapid proteasomal degradation of its own transporter. The current study examined the role of the metallochaperone Atox1 in the regulation of uptake, efflux and subcellular distribution of CDDP by using a pair of fibroblast cell lines established from Atox1(+/+) and Atox1(-/-) mice. Atox1 is a metallochaperone that is known to play a central role in distributing Cu within the cells and was recently shown to act as a Cu-dependent transcription factor. Loss of Atox1 increased Cu accumulation and reduced efflux. In contrast, loss of Atox1 reduced the influx of CDDP and subsequent accumulation in vesicular compartments and in DNA. Loss of Atox1 was found to block the CDDP-induced down regulation of Ctr1. Ctr1 was found to be polyubiquitinated in an Atox1-dependent manner during CDDP exposure. In conclusion, Atox1 is required for the polyubiquitination of Ctr1 and the Ctr1-mediated uptake of CDDP.


Assuntos
Antineoplásicos/farmacologia , Proteínas de Transporte de Cátions/metabolismo , Cisplatino/farmacologia , Chaperonas Moleculares/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Animais , Antineoplásicos/metabolismo , Transporte Biológico , Proteínas de Transporte de Cátions/genética , Linhagem Celular , Cisplatino/metabolismo , Cobre/farmacologia , Proteínas de Transporte de Cobre , Adutos de DNA/metabolismo , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Camundongos , Camundongos Knockout , Chaperonas Moleculares/genética , Poliubiquitina/metabolismo , Ubiquitinação
17.
Breast Cancer Res Treat ; 99(1): 23-33, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16541319

RESUMO

Currently, a number of breast cancer cell lines exist that serve as models for both estrogen receptor alpha (ERalpha) positive and ERalpha negative disease. Models are also available for pre-neoplastic breast epithelial cells that do not express ERalpha; however, there are no ideal systems for studying pre-neoplastic cells that are ERalpha positive. This has been largely due to the inability to establish an estrogen growth stimulated, non-tumorigenic breast epithelial cell line, as most human breast epithelial cells engineered to overexpress ERalpha have been found to be growth inhibited by estrogens. We have developed independently derived clones from the non-cancerous MCF-10A human breast cell line that express ERalpha and are growth stimulated by 17-beta-estradiol (E2) in the absence of epidermal growth factor (EGF), a cytokine normally required for MCF-10A cell proliferation. This effect is blocked by the selective estrogen receptor modulator (SERM), Tamoxifen and the selective estrogen receptor downregulator, ICI 182,780 (Faslodex, Fulvestrant). Exposure of these cells to EGF and E2 results in a growth inhibitory phenotype similar to previous reports. These data present a reconciling explanation for the previously described paradoxical effects of ERalpha overexpression, and provide a model for examining the carcinogenic effects of estrogens in non-tumorigenic human breast epithelial cells.


Assuntos
Mama/metabolismo , Células Epiteliais/metabolismo , Receptor alfa de Estrogênio/metabolismo , Regulação da Expressão Gênica , Linhagem Celular Tumoral , Proliferação de Células , Estradiol/metabolismo , Estrogênios/metabolismo , Humanos , Fosforilação , Lesões Pré-Cancerosas , Elementos de Resposta , Análise de Sequência de DNA , Transdução de Sinais , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA