Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
J Biol Chem ; 295(16): 5496-5508, 2020 04 17.
Artigo em Inglês | MEDLINE | ID: mdl-32161116

RESUMO

Previous studies have shown that sphingosine kinase interacting protein (SKIP) inhibits sphingosine kinase (SK) function in fibroblasts. SK phosphorylates sphingosine producing the potent signaling molecule sphingosine-1-phosphate (S1P). SKIP gene (SPHKAP) expression is silenced by hypermethylation of its promoter in acute myeloid leukemia (AML). However, why SKIP activity is silenced in primary AML cells is unclear. Here, we investigated the consequences of SKIP down-regulation in AML primary cells and the effects of SKIP re-expression in leukemic cell lines. Using targeted ultra-HPLC-tandem MS (UPLC-MS/MS), we measured sphingolipids (including S1P and ceramides) in AML and control cells. Primary AML cells had significantly lower SK activity and intracellular S1P concentrations than control cells, and SKIP-transfected leukemia cell lines exhibited increased SK activity. These findings show that SKIP re-expression enhances SK activity in leukemia cells. Furthermore, other bioactive sphingolipids such as ceramide were also down-regulated in primary AML cells. Of note, SKIP re-expression in leukemia cells increased ceramide levels 2-fold, inactivated the key signaling protein extracellular signal-regulated kinase, and increased apoptosis following serum deprivation or chemotherapy. These results indicate that SKIP down-regulation in AML reduces SK activity and ceramide levels, an effect that ultimately inhibits apoptosis in leukemia cells. The findings of our study contrast with previous results indicating that SKIP inhibits SK function in fibroblasts and therefore challenge the notion that SKIP always inhibits SK activity.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Leucemia Mieloide Aguda/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Ceramidas/metabolismo , Humanos , Células K562 , Células Tumorais Cultivadas
3.
Artigo em Inglês | MEDLINE | ID: mdl-25195021

RESUMO

A simple and rapid ultra-high performance liquid chromatography-mass spectrometry/mass spectrometry (UPLC-MS/MS) method has been developed for measuring intracellular concentrations of the anticancer agent 7-ethyl-10-hydroxycamptothecin (SN-38) in tumour cells using camptothecin (CPT) as internal standard. SN-38 extraction was carried out using acidified acetonitrile. SN-38 and CPT were separated on a PFP column using gradient elution with acidified water and acetonitrile. SN-38 and CPT were quantified using a triple quadrupole mass spectrometry system. Least square regression calibration lines were obtained with average correlation coefficients of R(2)=0.9993±0.0016. The lower limit of detection (LOD) and lower limit of quantification (LOQ) for SN-38 were 0.1 and 0.3ng/ml, respectively. CPT recovery was 98.5±13% and SN-38 recoveries at low quality control (LQC, 5ng/ml) and high quality control (HQC, 500ng/ml) were 89±6% and 95±8%, respectively. The intra- and inter-day imprecision for LQC was 5.8 and 8.5%, and for HQC was 6.3 and 4.4%, respectively. The method was compared to a validated high performance liquid chromatography-fluorescent method. In addition, the method has been successfully applied to determine the intracellular accumulation of SN-38 investigating the transport through ABCB1 (P-gp) and ABCG2 (BCRP) efflux pumps in colorectal cancer cell lines.


Assuntos
Camptotecina/análogos & derivados , Cromatografia Líquida de Alta Pressão/métodos , Neoplasias do Colo/química , Espectrometria de Massas em Tandem/métodos , Acetonitrilas , Camptotecina/análise , Linhagem Celular Tumoral , Estabilidade de Medicamentos , Células HCT116 , Células HT29 , Humanos , Espaço Intracelular/química , Irinotecano , Análise dos Mínimos Quadrados , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Espectrometria de Fluorescência
4.
Brain ; 137(Pt 1): 92-108, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24287115

RESUMO

Progressive multiple sclerosis is associated with metabolic failure of the axon and excitotoxicity that leads to chronic neurodegeneration. Global sodium-channel blockade causes side effects that can limit its use for neuroprotection in multiple sclerosis. Through selective targeting of drugs to lesions we aimed to improve the potential therapeutic window for treatment. This was assessed in the relapsing-progressive experimental autoimmune encephalomyelitis ABH mouse model of multiple sclerosis using conventional sodium channel blockers and a novel central nervous system-excluded sodium channel blocker (CFM6104) that was synthesized with properties that selectively target the inflammatory penumbra in experimental autoimmune encephalomyelitis lesions. Carbamazepine and oxcarbazepine were not immunosuppressive in lymphocyte-driven autoimmunity, but slowed the accumulation of disability in experimental autoimmune encephalomyelitis when administered during periods of the inflammatory penumbra after active lesion formation, and was shown to limit the development of neurodegeneration during optic neuritis in myelin-specific T cell receptor transgenic mice. CFM6104 was shown to be a state-selective, sodium channel blocker and a fluorescent p-glycoprotein substrate that was traceable. This compound was >90% excluded from the central nervous system in normal mice, but entered the central nervous system during the inflammatory phase in experimental autoimmune encephalomyelitis mice. This occurs after the focal and selective downregulation of endothelial p-glycoprotein at the blood-brain barrier that occurs in both experimental autoimmune encephalomyelitis and multiple sclerosis lesions. CFM6104 significantly slowed down the accumulation of disability and nerve loss in experimental autoimmune encephalomyelitis. Therapeutic-targeting of drugs to lesions may reduce the potential side effect profile of neuroprotective agents that can influence neurotransmission. This class of agents inhibit microglial activity and neural sodium loading, which are both thought to contribute to progressive neurodegeneration in multiple sclerosis and possibly other neurodegenerative diseases.


Assuntos
Benzamidas/uso terapêutico , Indazóis/uso terapêutico , Esclerose Múltipla/tratamento farmacológico , Fármacos Neuroprotetores/uso terapêutico , Oxidiazóis/uso terapêutico , Bloqueadores dos Canais de Sódio/uso terapêutico , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Animais , Bancos de Espécimes Biológicos , Encéfalo/patologia , Carbamazepina/farmacologia , Proteínas de Transporte/metabolismo , Proliferação de Células/efeitos dos fármacos , Cromatografia Líquida de Alta Pressão , Sistemas de Liberação de Medicamentos , Encefalomielite Autoimune Experimental/metabolismo , Feminino , Imuno-Histoquímica , Espectrometria de Massas , Camundongos , Camundongos Endogâmicos C57BL , Esclerose Múltipla/fisiopatologia , Neurite Óptica/fisiopatologia , Linfócitos T/efeitos dos fármacos , Uveíte/fisiopatologia , Canais de Sódio Disparados por Voltagem/metabolismo
6.
Genome Biol ; 14(4): R37, 2013 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-23628362

RESUMO

BACKGROUND: Tumor classification based on their predicted responses to kinase inhibitors is a major goal for advancing targeted personalized therapies. Here, we used a phosphoproteomic approach to investigate biological heterogeneity across hematological cancer cell lines including acute myeloid leukemia, lymphoma, and multiple myeloma. RESULTS: Mass spectrometry was used to quantify 2,000 phosphorylation sites across three acute myeloid leukemia, three lymphoma, and three multiple myeloma cell lines in six biological replicates. The intensities of the phosphorylation sites grouped these cancer cell lines according to their tumor type. In addition, a phosphoproteomic analysis of seven acute myeloid leukemia cell lines revealed a battery of phosphorylation sites whose combined intensities correlated with the growth-inhibitory responses to three kinase inhibitors with remarkable correlation coefficients and fold changes (> 100 between the most resistant and sensitive cells). Modeling based on regression analysis indicated that a subset of phosphorylation sites could be used to predict response to the tested drugs. Quantitative analysis of phosphorylation motifs indicated that resistant and sensitive cells differed in their patterns of kinase activities, but, interestingly, phosphorylations correlating with responses were not on members of the pathway being targeted; instead, these mainly were on parallel kinase pathways. CONCLUSION: This study reveals that the information on kinase activation encoded in phosphoproteomics data correlates remarkably well with the phenotypic responses of cancer cells to compounds that target kinase signaling and could be useful for the identification of novel markers of resistance or sensitivity to drugs that target the signaling network.


Assuntos
Leucemia Mieloide Aguda/metabolismo , Linfoma/metabolismo , Mieloma Múltiplo/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Processamento de Proteína Pós-Traducional , Proteoma/metabolismo , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Humanos , Fosforilação , Proteoma/química , Proteoma/efeitos dos fármacos
7.
Sci Signal ; 6(268): rs6, 2013 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-23532336

RESUMO

Kinases determine the phenotypes of many cancer cells, but the frequency with which individual kinases are activated in primary tumors remains largely unknown. We used a computational approach, termed kinase-substrate enrichment analysis (KSEA), to systematically infer the activation of given kinase pathways from mass spectrometry-based phosphoproteomic analysis of acute myeloid leukemia (AML) cells. Experiments conducted in cell lines validated the approach and, furthermore, revealed that DNA-dependent protein kinase (DNA-PK) was activated as a result of inhibiting the phosphoinositide 3-kinase (PI3K)-mammalian target of rapamycin (mTOR) signaling pathway. Application of KSEA to primary AML cells identified PI3K, casein kinases (CKs), cyclin-dependent kinases (CDKs), and p21-activated kinases (PAKs) as the kinase substrate groups most frequently enriched in this cancer type. Substrates phosphorylated by extracellular signal-regulated kinase (ERK) and cell division cycle 7 (CDC7) were enriched in primary AML cells that were resistant to inhibition of PI3K-mTOR signaling, whereas substrates of the kinases Abl, Lck, Src, and CDK1 were increased in abundance in inhibitor-sensitive cells. Modeling based on the abundances of these substrate groups accurately predicted sensitivity to a dual PI3K and mTOR inhibitor in two independent sets of primary AML cells isolated from patients. Thus, our study demonstrates KSEA as an untargeted method for the systematic profiling of kinase pathway activities and for increasing our understanding of diseases caused by the dysregulation of signaling pathways.


Assuntos
Biologia Computacional/métodos , Proteína Quinase Ativada por DNA/metabolismo , Ativação Enzimática/fisiologia , Leucemia Mieloide Aguda/enzimologia , Modelos Biológicos , Transdução de Sinais/fisiologia , Ativação Enzimática/genética , Humanos , Espectrometria de Massas/métodos , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Proteômica/métodos , Transdução de Sinais/genética , Especificidade por Substrato , Serina-Treonina Quinases TOR/metabolismo
8.
Blood ; 121(12): 2274-84, 2013 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-23341541

RESUMO

Phosphoinositide-3 kinase (PI3K) pathway activation contributes to mantle cell lymphoma (MCL) pathogenesis, but early-phase studies of the PI3K p110δ inhibitor GS-1101 have reported inferior responses in MCL compared with other non-Hodgkin lymphomas. Because the relative importance of the class IA PI3K isoforms p110α, p110ß, and p110δ in MCL is not clear, we studied expression of these isoforms and assessed their contribution to PI3K signaling in this disease. We found that although p110δ was highly expressed in MCL, p110α showed wide variation and expression increased significantly with relapse. Loss of phosphatase and tensin homolog expression was found in 16% (22/138) of cases, whereas PIK3CA and PIK3R1 mutations were absent. Although p110δ inhibition was sufficient to block B-cell receptor-mediated PI3K activation, combined p110α and p110δ inhibition was necessary to abolish constitutive PI3K activation. In addition, GDC-0941, a predominantly p110α/δ inhibitor, was significantly more active compared with GS-1101 against MCL cell lines and primary samples. We found that a high PIK3CA/PIK3CD ratio identified a subset of primary MCLs resistant to GS-1101 and this ratio increased significantly with relapse. These findings support the use of dual p110α/p110δ inhibitors in MCL and suggest a role for p110α in disease progression.


Assuntos
Linfoma de Célula do Manto/tratamento farmacológico , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Inibidores de Proteínas Quinases/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Linhagem Celular Tumoral , Classe I de Fosfatidilinositol 3-Quinases , Análise Mutacional de DNA , Resistencia a Medicamentos Antineoplásicos/genética , Regulação Enzimológica da Expressão Gênica/fisiologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/genética , Regulação Neoplásica da Expressão Gênica/fisiologia , Humanos , Linfoma de Célula do Manto/genética , Linfoma de Célula do Manto/metabolismo , Linfoma de Célula do Manto/patologia , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/fisiologia , Inibidores de Proteínas Quinases/administração & dosagem , Inibidores de Proteínas Quinases/farmacologia , Purinas/administração & dosagem , Purinas/farmacologia , Purinas/uso terapêutico , Quinazolinonas/administração & dosagem , Quinazolinonas/farmacologia , Quinazolinonas/uso terapêutico , Recidiva , Transdução de Sinais/fisiologia , Especificidade por Substrato , Resultado do Tratamento
9.
J Pain Symptom Manage ; 45(4): 670-80, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22995672

RESUMO

CONTEXT: Morphine is the opioid of choice for cancer-related pain, but for many patients the benefits of morphine are outweighed by its side effect profile. Morphine is metabolized to morphine-3-glucuronide and morphine-6-glucuronide; however, little is known about the contribution of these metabolites to analgesia and morphine-related side effects. OBJECTIVES: We investigated the association between plasma morphine and metabolite concentrations and the clinical effects of morphine in cancer patients. METHODS: A prospective study was performed in cancer patients taking oral morphine for moderate-to-severe cancer pain. Subjects who responded well to morphine (responders) and subjects who failed to respond to morphine because of lack of analgesia and/or the presence of intolerable side effects (nonresponders/switchers) were recruited. Pain and toxicity scores were recorded and blood samples were analyzed for plasma morphine, morphine-3-glucuronide, and morphine-6-glucuronide concentrations. RESULTS: Results showed that 1) morphine responders have higher plasma morphine and metabolite concentrations compared with nonresponders, 2) lower pain scores are associated with higher plasma morphine and metabolite concentrations, 3) central side effects are associated with a higher metabolite:plasma morphine ratio, and 4) myoclonus is associated with extremely high concentrations of plasma morphine and metabolites. CONCLUSION: This study has shown that plasma morphine and metabolite concentrations are associated with the clinical effects of morphine therapy. These results are important because they demonstrate the relevance of measuring plasma metabolite concentrations in clinical trials and the potential for metabolite data to deepen our understanding of factors that influence an individual's response to morphine.


Assuntos
Morfina/sangue , Neoplasias/sangue , Neoplasias/epidemiologia , Transtornos Relacionados ao Uso de Opioides/sangue , Transtornos Relacionados ao Uso de Opioides/epidemiologia , Dor/sangue , Dor/prevenção & controle , Analgésicos Opioides/sangue , Analgésicos Opioides/uso terapêutico , Biomarcadores/sangue , Causalidade , Comorbidade , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Morfina/uso terapêutico , Neoplasias/enfermagem , Dor/epidemiologia , Medição da Dor/efeitos dos fármacos , Medição da Dor/estatística & dados numéricos , Prevalência , Fatores de Risco , Estatística como Assunto , Resultado do Tratamento , Reino Unido/epidemiologia
12.
Br J Haematol ; 154(4): 448-56, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21770918

RESUMO

We have previously reported presentation serum selenium level to be predictive of outcome in diffuse large B-cell lymphoma. This has now been studied in a further 430 patients, 163 with acute myeloid leukaemia (AML), 156 with Hodgkin Lymphoma (HL), and 111 with Follicular Lymphoma (FL). Serum selenium was below the UK normal reference range in 45% of patients, and correlated with serum albumin (r=0·24-0·46, P<0·001-0·003) in all tumour types. Independent predictors of presentation selenium were; French-American-British subtype and albumin (P<0·001 for both) in AML, haemoglobin (P=0·002) and B-symptoms (P=0·01) in HL, and albumin (P<0·001) in FL. In AML and HL, response to first line therapy was lower in patients with low serum selenium, but selenium was no longer predictive of response when other variables were entered into a multivariate model. Low selenium was also associated with a worse overall survival in FL [Hazard Ratio (HR) 2·3, 95% confidence interval (CI) 1·4, 4·0] and a trend to a worse overall survival in AML (HR 1·43, 95% CI 0·96, 2·13) by univariate Cox regression analysis, but not by multivariate analysis. In conclusion, low serum selenium is associated with a worse outcome in patients with haematological malignancies, but is not independently predictive, suggesting that it reflects other factors.


Assuntos
Biomarcadores Tumorais/sangue , Neoplasias Hematológicas/diagnóstico , Selênio/sangue , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Feminino , Seguimentos , Neoplasias Hematológicas/sangue , Neoplasias Hematológicas/terapia , Doença de Hodgkin/sangue , Doença de Hodgkin/diagnóstico , Doença de Hodgkin/terapia , Humanos , Estimativa de Kaplan-Meier , Leucemia Mieloide Aguda/sangue , Leucemia Mieloide Aguda/diagnóstico , Leucemia Mieloide Aguda/terapia , Linfoma Folicular/sangue , Linfoma Folicular/diagnóstico , Linfoma Folicular/terapia , Masculino , Pessoa de Meia-Idade , Prognóstico , Albumina Sérica/metabolismo , Resultado do Tratamento , Adulto Jovem
13.
Cancer Chemother Pharmacol ; 68(3): 815-21, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21519842

RESUMO

PURPOSE: Selenium is a trace element that is fundamental to human health. Research has mainly focussed on its role in cancer prevention, but recent evidence supports its role in established cancer, with high concentrations inducing tumour cell death and non-toxic concentrations sensitising cells to chemotherapy. However, the precise mechanism of selenium action is not clear. The effect of methylseleninic acid (MSA), an organic selenium compound, on histone deacetylase (HDAC) activity in diffuse large B-cell lymphoma cell lines is reported here. METHODS: Lymphoma cell lines were exposed to MSA under normoxic and hypoxic conditions. Protein expression was determined by western blotting, HDAC activity and VEGF concentration by fluorimetric and electrochemiluminescence assays, respectively, and intracellular selenium metabolites quantified by mass spectrometry. RESULTS: MSA inhibited HDAC activity, which resulted in the acetylation of histone H3 and α-tubulin. However, cellular metabolism of MSA to methylselenol was required for this effect. Dimethylselenide, the methylation product of methylselenol, was found to be the major intracellular metabolite. MSA also inhibited HIF-1α expression and VEGF secretion, a possible consequence of HDAC inhibition. CONCLUSION: The ability of methylselenol to inhibit HDAC activity has not been previously reported, thus providing a novel mechanism of selenium action.


Assuntos
Inibidores de Histona Desacetilases , Histona Desacetilases/metabolismo , Linfoma de Células B/enzimologia , Compostos Organosselênicos/farmacologia , Western Blotting , Linhagem Celular Tumoral , Cistina/efeitos dos fármacos , Cistina/metabolismo , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/antagonistas & inibidores , Selênio/metabolismo , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Fator A de Crescimento do Endotélio Vascular/metabolismo
14.
Anal Bioanal Chem ; 399(5): 1789-97, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21140135

RESUMO

The formation of intracellular dimethylselenide (DMSe) as a product of exposure of non-malignant (PBMCs) and lymphoma (RL and DHL-4) cell lines to methylseleninic acid (MSA) at clinical levels is suggested here for the first time. This was achieved by analysis of cell lysates by HPLC coupled to ICP-MS via APEX-Q nebulisation, enabling limits of detection for target methyl-Se species which are up to 12-fold lower than those obtained with conventional nebulisation. Methyl-Se-glutathione (CH3Se-SG), although detected in lysates of cells exposed to MSA, was found to be a reaction product of MSA with glutathione. This was confirmed by HPLC-ESI MS (MS) analysis of lysates of control cells (unexposed to Se) spiked with MSA. The MS/MS data obtained by collision-induced dissociation fragmentation of the ion m/z 402 (for [M+H](+) 8°Se) were consistent with the presence of CH3Se-SG. Formation of DMSe was not detected by HPLC-ICP-MS in these spiked lysates, and it was found to require live cells in cell media containing MSA. Interestingly, the ratio of DMSe to CH3Se-SG was significantly higher in lymphoma cells exposed to MSA in comparison to non-malignant cells. Moreover, maximum Se uptake levels in lymphoma cell lines seemed to be reached much earlier (after 10 min of MSA exposure) than in non-malignant cells. Finally, the GC-TOF-MS speciation data obtained for cell headspace suggested that the major Se species (dimethyldiselenide) appeared to be present in lymphoma cell headspace at significantly higher concentrations than in non-malignant cell headspace after only 10 min of exposure to MSA. Evidence for the presence of dimethylselenidesulfide in lymphoma cell headspace is also provided for the first time.


Assuntos
Linfoma de Células B/química , Selênio/análise , Selênio/farmacocinética , Linhagem Celular , Cromatografia Líquida de Alta Pressão , Humanos , Linfoma de Células B/metabolismo , Espectrometria de Massas , Compostos Organosselênicos/metabolismo , Compostos Organosselênicos/farmacologia , Distribuição Tecidual
15.
Mol Cancer Ther ; 9(12): 3322-9, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20889729

RESUMO

Lapatinib is a specific HER1 and 2 targeted tyrosine kinase inhibitor now widely used in combination with chemotherapy in the clinical setting. In this work, we investigated the interactions between lapatinib and specific chemotherapy agents (cisplatin, SN-38, topotecan) in a panel of cell lines [breast (n = 2), lung (n = 2), testis (n = 4)]. A high-sensitivity cell proliferation/cytotoxicity ATP assay and flow cytometry were used to determine cell viability, apoptosis, and the effect of the drugs on cell-cycle distribution. CalcuSyn analysis was employed to formally identify synergistic interactions between drugs. Intracellular concentrations of SN-38 were measured using a novel high-performance liquid chromatography (HPLC) technique. Flow cytometry and HPLC techniques were used to identify the effect of lapatinib on drug influx and efflux pumps, using specific substrates and inhibitors of these pumps. Results showed significant synergy between SN-38, and lapatinib in the majority of cell lines (combination index < 0.75), associated with increased apoptosis. This synergy was not universal but, when observed (Susa S/R, H1975, H358, and MDA-MB-231 cell lines), was related to SN-38 intracellular accumulation (2.2- to 4.8-fold increase, P < 0.05 for each), attributable to the inhibition of the breast cancer-related protein (BCRP) efflux pump by lapatinib. Flow cytometry analysis showed that lapatinib (10 µmol/L) inhibited the efflux of mitoxantrone, a specific substrate of the BCRP pump, in a manner similar to fumitremorgin C, a known BCRP inhibitor, confirming lapatinib as a BCRP inhibitor. This work shows that lapatinib has a direct inhibitory effect on BCRP accounting for the synergistic findings. The synergy is cell line dependent and related to the activity of specific efflux pumps.


Assuntos
Transportadores de Cassetes de Ligação de ATP/antagonistas & inibidores , Antineoplásicos/farmacologia , Proteínas de Neoplasias/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Quinazolinas/farmacologia , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/metabolismo , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Cromatografia Líquida de Alta Pressão , Cisplatino/farmacologia , Ensaios de Seleção de Medicamentos Antitumorais , Sinergismo Farmacológico , Citometria de Fluxo , Humanos , Indazóis , Espaço Intracelular/efeitos dos fármacos , Espaço Intracelular/metabolismo , Lapatinib , Proteínas de Neoplasias/metabolismo , Pirimidinas/farmacologia , Quinazolinas/química , Sulfonamidas/farmacologia
17.
Am J Pathol ; 176(6): 2607-15, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20395447

RESUMO

Development of chemoresistance limits the clinical efficiency of platinum-based therapy. Although many resistance mechanisms have been demonstrated, genetic/molecular alterations responsible for drug resistance in the majority of clinical cases have not been identified. We analyzed three pairs of testicular germ cell tumor cell lines using Affymetrix expression microarrays and revealed a limited number of differentially expressed genes across the cell lines when comparing the parental and resistant cells. Among them, CCND1 was the most significantly differentially expressed gene. Analysis of testicular germ cell tumor clinical samples by quantitative reverse transcription PCR analysis revealed that overall expression of CCND1 was significantly higher in resistant cases compared with sensitive samples (P < 0.0001). We also found that CCND1 was dramatically overexpressed both in induced and intrinsically resistant samples of ovarian and prostate cancer. Finally combined CCND1 knockdown using small-interfering RNA and cisplatin treatment inhibited cell growth in vitro significantly more effectively than any of these single treatments. Therefore, deregulation of CCND1 may be a major cause of cisplatin resistance in testicular germ cell tumors and may also be implicated in ovarian and prostate cancers. CCND1 could be potentially used as a marker for treatment stratification and as a molecular target to improve the treatment of platinum-resistant tumors.


Assuntos
Antineoplásicos/uso terapêutico , Cisplatino/uso terapêutico , Ciclina D1/genética , Resistencia a Medicamentos Antineoplásicos/genética , Neoplasias Embrionárias de Células Germinativas , Neoplasias Testiculares , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular , Hibridização Genômica Comparativa , Ciclina D1/metabolismo , Feminino , Perfilação da Expressão Gênica , Humanos , Masculino , Análise em Microsséries , Neoplasias Embrionárias de Células Germinativas/tratamento farmacológico , Neoplasias Embrionárias de Células Germinativas/patologia , Neoplasias Embrionárias de Células Germinativas/fisiopatologia , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/patologia , Neoplasias Ovarianas/fisiopatologia , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/patologia , Neoplasias da Próstata/fisiopatologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Neoplasias Testiculares/tratamento farmacológico , Neoplasias Testiculares/patologia , Neoplasias Testiculares/fisiopatologia
18.
Blood ; 115(19): 3939-48, 2010 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-20190189

RESUMO

GCS-100 is a galectin-3 antagonist with an acceptable human safety profile that has been demonstrated to have an antimyeloma effect in the context of bortezomib resistance. In the present study, the mechanisms of action of GCS-100 are elucidated in myeloma cell lines and primary tumor cells. GCS-100 induced inhibition of proliferation, accumulation of cells in sub-G(1) and G(1) phases, and apoptosis with activation of both caspase-8 and -9 pathways. Dose- and time-dependent decreases in MCL-1 and BCL-X(L) levels also occurred, accompanied by a rapid induction of NOXA protein, whereas BCL-2, BAX, BAK, BIM, BAD, BID, and PUMA remained unchanged. The cell-cycle inhibitor p21(Cip1) was up-regulated by GCS-100, whereas the procycling proteins CYCLIN E2, CYCLIN D2, and CDK6 were all reduced. Reduction in signal transduction was associated with lower levels of activated IkappaBalpha, IkappaB kinase, and AKT as well as lack of IkappaBalpha and AKT activation after appropriate cytokine stimulation (insulin-like growth factor-1, tumor necrosis factor-alpha). Primary myeloma cells showed a direct reduction in proliferation and viability. These data demonstrate that the novel therapeutic molecule, GCS-100, is a potent modifier of myeloma cell biology targeting apoptosis, cell cycle, and intracellular signaling and has potential for myeloma therapy.


Assuntos
Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Galectina 3/antagonistas & inibidores , Mieloma Múltiplo/patologia , Polissacarídeos/farmacologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Western Blotting , Caspases/metabolismo , Proliferação de Células/efeitos dos fármacos , Técnicas de Cocultura , Ativação Enzimática/efeitos dos fármacos , Galectina 3/metabolismo , Humanos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/metabolismo , Proteína de Sequência 1 de Leucemia de Células Mieloides , Células Estromais/efeitos dos fármacos , Células Tumorais Cultivadas
19.
Cancer Res ; 69(10): 4150-8, 2009 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-19366807

RESUMO

Aurora kinases play a critical role in regulating mitosis and cell division, and their overexpression has been implicated in the survival and proliferation of human cancer. In this study, we report the in vitro and in vivo activities of AZD1152, a compound that has selectivity for aurora B kinase, in acute myeloid leukemia (AML) cell lines, primary AML samples, and cord blood cells. AZD1152 exerted antiproliferative or cytotoxic effects in all cell lines studied, inhibited the phosphorylation of histone H3 (pHis H3) on Ser10 in a dose-dependent manner, and resulted in cells with >4N DNA content. THP-1 cells treated with AZD1152 accumulated in a state of polyploidy and showed a senescent response to the drug, in contrast to the apoptotic response seen in other cell lines. Accordingly, AZD1152 profoundly affected the growth of AML cell lines and primary AML in an in vivo xenotransplantation model. However, concentration-dependent effects on cell growth, apoptosis, and cell cycle progression were also observed when human cord blood and primary lineage-negative stem and progenitor cells were analyzed in vitro and in vivo. These data suggest that the inhibition of aurora B kinase may be a useful therapeutic strategy in the treatment of AML and that further exploration of dosing and treatment schedules is warranted in clinical trials.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Leucemia Mieloide Aguda/patologia , Organofosfatos/farmacologia , Quinazolinas/farmacologia , Animais , Anexina A5/metabolismo , Antineoplásicos/uso terapêutico , Aurora Quinase B , Aurora Quinases , Linhagem Celular Tumoral , Células HL-60/efeitos dos fármacos , Células-Tronco Hematopoéticas/citologia , Humanos , Leucemia Mieloide Aguda/tratamento farmacológico , Camundongos , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Células U937/efeitos dos fármacos
20.
Cancer Chemother Pharmacol ; 64(5): 925-33, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19263053

RESUMO

BACKGROUND: Germ cell tumours (GCT) can become resistant to cisplatin, which is associated with a relatively poor prognosis. Oxaliplatin and satraplatin have been developed to overcome cisplatin resistance in other cancers, but their effect in cisplatin resistant (cisR) GCTs is unclear. In this work we address this issue by comparing their efficacy in three paired sensitive and cisR GCT cell lines. METHODS: Three established cisplatin sensitive (cisS) and resistant cell line pairs were used (GCT27, GCT27r: SUSA, SUSAr: 833k, 833kr). Viability was assessed using a luciferase based ATP assay and EC(50) and EC(80) concentrations were calculated. Western blot analysis and flow cytometry was used for further assessment. RESULTS: Sensitivity to the three platinum compounds was broadly similar in the three cisS lines GCT cell lines (EC(50) = 0.27-0.51 microM for cisplatin, 0.52-0.79 microM for oxaliplatin, 0.31-1.26 microM for satraplatin). EC(50) values for cisplatin in the three cisR sub lines were 1.8- to 3.8-fold higher than in the sensitive parental lines. Cross resistance to satraplatin and oxaliplatin occurred in all three cisR cell lines (resistance factor 1.9-4.4), with the exception of oxaliplatin in the 833Kr (resistance factor 0.9). Differences in the effect of specific drugs on cell cycle distribution, p53, p21 and MDM2 were observed. CONCLUSIONS: These data suggest that satraplatin and oxaliplatin could theoretically be used in chemo-naive GCTs and support the further clinical evaluation of these agents in this setting. The mechanism of cross resistance to these drugs appears multifactorial.


Assuntos
Antineoplásicos/farmacologia , Cisplatino/farmacologia , Neoplasias Embrionárias de Células Germinativas/tratamento farmacológico , Compostos Organoplatínicos/farmacologia , Neoplasias Testiculares/tratamento farmacológico , Apoptose/efeitos dos fármacos , Western Blotting , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , DNA de Neoplasias/biossíntese , Resistencia a Medicamentos Antineoplásicos , Citometria de Fluxo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Neoplasias Embrionárias de Células Germinativas/patologia , Proteína Oncogênica p21(ras)/biossíntese , Oxaliplatina , Proteínas Proto-Oncogênicas c-mdm2/biossíntese , Neoplasias Testiculares/patologia , Proteína Supressora de Tumor p53/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...