Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Mol Pharmacol ; 82(3): 510-20, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22710939

RESUMO

The Sec61 protein translocon is a multimeric complex that transports proteins across lipid bilayers. We discovered that the Sec61ß subunit modulates cellular sensitivity to chemotherapeutic agents, particularly the platinum drugs. To investigate the mechanism, expression of Sec61ß was constitutively knocked down in 2008 ovarian cancer cells. Sec61ß knockdown (KD) resulted in 8-, 16.8-, and 9-fold resistance to cisplatin (cDDP), carboplatin, and oxaliplatin, respectively. Sec61ß KD reduced the cellular accumulation of cDDP to 67% of that in parental cells. Baseline copper levels, copper uptake, and copper cytotoxicity were also reduced. Because copper transporters and chaperones regulate platinum drug accumulation and efflux, their expression in 2008 Sec61ß-KD cells was analyzed; ATP7A was found to be 2- to 3-fold overexpressed, whereas there was no change in ATP7B, ATOX1, CTR1, or CTR2 levels. Cells lacking ATP7A did not exhibit increased cDDP resistance upon knockdown of Sec61ß. Sec61ß-KD cells also exhibited altered ATP7A cellular distribution. We conclude that Sec61ß modulates the cytotoxicity of many chemotherapeutic agents, with the largest effect being on the platinum drugs. This modulation occurs through effects of Sec61ß on the expression and distribution of ATP7A, which was shown previously to control platinum drug sequestration and cytotoxicity.


Assuntos
Adenosina Trifosfatases/metabolismo , Antineoplásicos/farmacologia , Proteínas de Transporte de Cátions/metabolismo , Proteínas de Membrana/metabolismo , Compostos Organoplatínicos/farmacologia , Adenosina Trifosfatases/genética , Transporte Biológico/efeitos dos fármacos , Proteínas de Transporte de Cátions/genética , Cobre/efeitos adversos , ATPases Transportadoras de Cobre , Resistencia a Medicamentos Antineoplásicos , Feminino , Homeostase/efeitos dos fármacos , Humanos , Proteínas de Membrana/genética , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/metabolismo , Canais de Translocação SEC , Células Tumorais Cultivadas
2.
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
3.
Mol Pharmacol ; 77(6): 887-94, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20159940

RESUMO

Multiple lines of evidence indicate that the platinum-containing cancer drugs enter cells, are distributed to various subcellular compartments, and are exported from cells via transporters that evolved to manage copper homeostasis. The cytotoxicity of the platinum drugs is directly related to how much drug enters the cell, and almost all cells that have acquired resistance to the platinum drugs exhibit reduced drug accumulation. The major copper influx transporter, copper transporter 1 (CTR1), has now been shown to control the tumor cell accumulation and cytotoxic effect of cisplatin, carboplatin, and oxaliplatin. There is a good correlation between change in CTR1 expression and acquired cisplatin resistance among ovarian cancer cell lines, and genetic knockout of CTR1 renders cells resistant to cisplatin in vivo. The expression of CTR1 is regulated at the transcriptional level by copper via Sp1 and at the post-translational level by the proteosome. Copper and cisplatin both trigger the down-regulation of CTR1 via a process that involves ubiquitination and proteosomal degradation and requires the copper chaperone antioxidant protein 1 (ATOX1). The cisplatin-induced degradation of CTR1 can be blocked with the proteosome inhibitor bortezomib, and this increases the cellular uptake and the cytotoxicity of cisplatin in a synergistic manner. Copper and platinum(II) have similar sulfur binding characteristics, and the presence of stacked rings of methionines and cysteines in the CTR1 trimer suggest a mechanism by which CTR1 selectively transports copper and the platinum-containing drugs via sequential transchelation reactions similar to the manner in which copper is passed from ATOX1 to the copper efflux transporters.


Assuntos
Antineoplásicos/farmacologia , Carboplatina/farmacologia , Proteínas de Transporte de Cátions/fisiologia , Cisplatino/farmacologia , Compostos Organoplatínicos/farmacologia , Proteínas de Transporte de Cátions/química , Proteínas de Transporte de Cátions/genética , Cobre/metabolismo , Cobre/farmacologia , Transportador de Cobre 1 , Regulação da Expressão Gênica/efeitos dos fármacos , Homeostase , Humanos , Oxaliplatina , Conformação Proteica
4.
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
5.
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
6.
Clin Cancer Res ; 15(2): 553-60, 2009 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-19147760

RESUMO

PURPOSE: The copper transporter 1 (CTR1) is a major influx transporter for platinum drugs. However, the accumulation of cisplatin in human ovarian carcinoma cells is limited by the fact that cisplatin triggers the down-regulation and proteasomal degradation of CTR1, thereby limiting its own uptake. We sought to determine whether proteasome inhibition using bortezomib would prevent human CTR1 (hCTR1) degradation and increase platinum accumulation in ovarian cancer cells. EXPERIMENTAL DESIGN: The effects of bortezomib on human hCTR1 expression and cisplatin accumulation were measured by Western blot, flow cytometric, and confocal digital imaging analyses. Platinum accumulation was measured by inductively coupled plasma mass spectrometry and bortezomib concentrations by liquid chromatography/mass spectrometry. RESULTS: Bortezomib blocked the cisplatin-induced down-regulation of hCTR1 in a concentration-dependent manner and increased cisplatin uptake 1.6- to 2.4-fold. Median effect analysis showed a combination index of 0.37 at 50% cell kill, indicating a high level of synergy. The effect of bortezomib was muted in cells lacking both alleles of CTR1, showing that bortezomib was working primarily through its effect on blocking hCTR1 degradation. I.p. administration of bortezomib produced a peritoneal/plasma area under the curve ratio of 252 in a murine model. I.p. administration of bortezomib before i.p. cisplatin increased platinum accumulation in peritoneal tumors by 33% (P = 0.006). CONCLUSIONS: Proteasomal inhibition prevented cisplatin-induced down-regulation of hCTR1 in ovarian cancer cells and enhanced drug uptake and cell killing in a synergistic manner. Bortezomib shows a large pharmacologic advantage when administered i.p. There is a strong rationale for the combined i.p. administration of bortezomib and cisplatin.


Assuntos
Antineoplásicos/farmacologia , Ácidos Borônicos/farmacologia , Proteínas de Transporte de Cátions/metabolismo , Cisplatino/farmacologia , Sistemas de Liberação de Medicamentos , Regulação Neoplásica da Expressão Gênica , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Peritoneais/tratamento farmacológico , Pirazinas/farmacologia , Animais , Bortezomib , Linhagem Celular Tumoral , Transportador de Cobre 1 , Feminino , Humanos , Camundongos , Camundongos Nus , Complexo de Endopeptidases do Proteassoma/metabolismo
7.
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
8.
Obes Surg ; 15(3): 323-9, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15832432

RESUMO

BACKGROUND: Fellowships in advanced laparoscopy with emphasis in laparoscopic gastric bypass (LGBP) are available for obtaining experience in performing LGBP. The following is the first report in the literature prospectively documenting a single surgeon's experience with LGBP outcomes following completion of an advanced laparoscopic surgical fellowship. METHODS: Outcomes measured prospectively included length of stay, length of operation, complications, reduction in obesity-related co-morbidities, and percentage excess weight loss. Outcomes were analyzed by quartile to see if there was difference over time. Complications were also compared to outcomes in the literature. RESULTS: 175 patients (147 female, 28 male) underwent LGBP. The mean BMI was 49.2. Mean operative time was 123 minutes, and mean length of stay was 2.2 days. The percentage excess weight loss at 1 year was 73% (n = 79). One patient developed an internal hernia (0.6%) and 1 patient developed an anastomotic leak (0.6%). Post operative transfusion rate was 4.6%. There were no deep venous thromboses or pulmonary emboli detected. There were no conversions to open, and there was no mortality. Upon quartile analysis, there was no difference in complication rates. Complication rates were comparable to published outcomes in the literature. CONCLUSION: Fellowships in advanced laparoscopy with emphasis on LGBP provide the optimal training environment for acquisition of skills necessary to safely and effectively perform LGBP. With fellowship training, complication rates were comparable to published outcomes in the literature without a period of higher complications (the learning curve).


Assuntos
Bariatria , Bolsas de Estudo , Cirurgia Geral/educação , Laparoscopia , Adulto , Anastomose em-Y de Roux/métodos , Bariatria/métodos , Feminino , Seguimentos , Derivação Gástrica/métodos , Humanos , Laparoscopia/métodos , Tempo de Internação , Masculino , Pessoa de Meia-Idade , Procedimentos Cirúrgicos Minimamente Invasivos/educação , Obesidade/cirurgia , Complicações Pós-Operatórias , Estudos Prospectivos , Fatores de Tempo , Resultado do Tratamento , Redução de Peso
9.
Biochem Pharmacol ; 90(4): 379-87, 2014 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-24967972

RESUMO

The copper influx transporter CTR1 is also a major influx transporter for cisplatin (cDDP) in tumor cells. It influences the cytotoxicity of cDDP both in vivo and in vitro. Whereas Cu triggers internalization of CTR1 from the plasma membrane, cDDP does not. To investigate the mechanisms of these effects, myc-tagged forms of wild type hCTR1 and variants in which Y103 was converted to alanine, C189 was converted to serine, or the K178/K179 dilysine motif was converted to alanines were re-expressed in mouse embryo cells in which both alleles of CTR1 had been knocked out and also in HEK293T cells. The Y103A mutation and to a lesser extent the C189S mutation reduced internalization of CTR1 induced by Cu while the K178A/K179A had little effect. Both Y103 and C189 were required for Cu and cDDP transport whereas the K178/K179 motif was not. While Y103 lies in an YXXM motif that, when phosphorylated, is a potential docking site for phosphatidylinositol 3-kinase and other proteins involved in endocytosis, Western blot analysis of immunoprecipitated myc-CTR1, and proteomic analysis of peptides derived from CTR1, failed to identify any basal or Cu-induced phosphorylation. However, proteomic analysis did identify an interaction of CTR1 with IRS-4 and this was confirmed by co-immunoprecipitation from HEK cells expressing either FLAG-CTR1 or myc-CTR1. The interaction was greater in the Y103A-expressing cells. We conclude that Y103 is required for the internalization of hCTR1 in response to Cu, that this occurs by a mechanism other than phosphorylation and that mutation of Y103 modulates the interaction with IRS-4.


Assuntos
Proteínas de Transporte de Cátions/metabolismo , Cisplatino/metabolismo , Cobre/metabolismo , Proteínas Substratos do Receptor de Insulina/metabolismo , Sequência de Aminoácidos , Animais , Biotina/metabolismo , Proteínas de Transporte de Cátions/química , Proteínas de Transporte de Cátions/genética , Transportador de Cobre 1 , Imunoprecipitação , Camundongos , Dados de Sequência Molecular , Mutação , Fosforilação , Ligação Proteica , Proteômica
10.
Cell Biochem Biophys ; 63(3): 223-34, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22569840

RESUMO

Human copper transporter 1 (hCTR1) is the major high affinity copper influx transporter in mammalian cells that also mediates uptake of the cancer chemotherapeutic agent cisplatin. A low resolution structure of hCTR1 determined by cryoelectron microscopy was recently published. Several protein structure simulation techniques were used to create an all-atom model of this important transporter using the low resolution structure as a starting point. The all-atom model provides new insights into the roles of specific residues of the N-terminal extracellular domain, the intracellular loop, and C-terminal region in metal ion transport. In particular, the model demonstrates that the central region of the pore contains four sets of methionine triads in the intramembranous region. The structure confirms that two triads of methionine residues delineate the intramembranous region of the transporter, and further identifies two additional methionine triads that are located in the extracellular N-terminal part of the transporter. Together, the four triads create a structure that promotes stepwise transport of metal ions into and then through the intramembranous channel of the transporter via transient thioether bonds to methionine residues. Putative copper-binding sites in the hCTR1 trimer were identified by a program developed by us for prediction of metal-binding sites. These sites correspond well with the known effects of mutations on the ability of the protein to transport copper and cisplatin.


Assuntos
Proteínas de Transporte de Cátions/química , Cobre/química , Simulação de Dinâmica Molecular , Sequência de Aminoácidos , Sítios de Ligação , Proteínas de Transporte de Cátions/metabolismo , Cisplatino/química , Transportador de Cobre 1 , Cristalografia por Raios X , Humanos , Dados de Sequência Molecular , Estrutura Terciária de Proteína , Alinhamento de Sequência
11.
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
12.
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
13.
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
14.
Retina ; 24(1): 120-31, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15076953

RESUMO

PURPOSE: Little is known about the ocular oxygen consumption rate (QO2) in human diseases. Alterations in QO2 must occur in many conditions, such as retinal ischemia. We present a method of estimating QO2 that eventually could be used in patients during vitrectomy surgery. METHODS: We performed vitreoperfusion (i.e., perfusion of the vitreous cavity after vitrectomy) in 14 cat eyes with no ocular blood flow. The solution contained nutrients at a high partial pressure of oxygen (PO2). In eight eyes, the retinas were undisturbed (Group 1), and in six eyes, we excised the retinas (Group 2). We estimated QO2 in both groups on the basis of the temporal decline of PO2 in the vitreoperfusion solution according to a pharmacokinetic model. RESULTS: The mean and standard deviation of QO2 was 3.2 +/- 0.8 microL/min in Group 1 and 0.4+/- 0.7 microL/min in Group 2, with the difference being the retinal contribution, 88%. In Group 1, metabolism, bulk flow, and diffusion accounted for 82, 13, and 5%, respectively, of the oxygen loss from the vitreoperfusion solution. CONCLUSION: We estimated ocular oxygen consumption by means of vitreoperfusion. Eventually, the pathophysiology of human diseases may be clarified by similar measurements during vitrectomy.


Assuntos
Hipóxia/metabolismo , Consumo de Oxigênio/fisiologia , Perfusão , Retina/metabolismo , Animais , Cateterismo/métodos , Gatos , Metabolismo Energético , Feminino , Isquemia/metabolismo , Masculino , Vitrectomia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA