Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
1.
Clin Nutr ESPEN ; 30: 113-118, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30904210

RESUMO

BACKGROUND/AIM: Disease related malnutrition is a major problem in hospitals. Malnutrition in hospitalized patients is caused by many factors. Among these factors are decreased appetite and early satiety, and reaching nutritional requirements in nutritional risk patients is a challenge when using ordinary energy and protein dense food. The aim of this study was to examine if total protein and energy intake in medical and surgical patients at nutritional risk could be improved by protein fortified and energy rich in-between meals. METHODS: An assortment of fortified in-between meals including 10 g of protein was developed based on patient preferences and served in the Departments of Lung Medicine and Abdominal Surgery for a period of three months. Nutrition intake was recorded before and after intervention. RESULTS: Food intake records were collected from a total of 92 patients, (46 before and 46 after intervention). The total amount of protein intake per in-between meal was increased from 2,6 g to 10,3 g. Total daily protein intake increased from 49% to 88% (p < 0.00) and total energy intake from 74% to 109% (p < 0.00) of requirements. CONCLUSION: Protein and energy intake for surgical and medical patients at in-between meals as well as total daily intake increased significantly. Recommended average level for individually measured requirements was reached.


Assuntos
Proteínas Alimentares , Ingestão de Energia , Pacientes Internados , Refeições , Desnutrição Proteico-Calórica/prevenção & controle , Feminino , Serviço Hospitalar de Nutrição , Humanos , Masculino , Necessidades Nutricionais , Estado Nutricional , Resultado do Tratamento
2.
Ann Oncol ; 28(9): 2077-2085, 2017 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-28430862

RESUMO

Traditionally, the efficacy of cancer treatment in patients with advance or metastatic disease in clinical studies has been studied using overall survival and more recently tumor-based end points such as progression-free survival, measurements of response to treatment. However, these seem not to be the relevant clinical end points in current situation if such end points were no validated as surrogate of overall survival to demonstrate the clinical efficacy. Appropriate, meaningful, primary patient-oriented and patient-reported end points that adequately measure the effects of new therapeutic interventions are then crucial for the advancement of clinical research in metastatic colorectal cancer to complement the results of tumor-based end points. Health-related quality of life (HRQoL) is effectively an evaluation of quality of life and its relationship with health over time. HRQoL includes the patient report at least of the way a disease or its treatment affects its physical, emotional and social well-being. Over the past few years, several phase III trials in a variety of solid cancers have assessed the incremental value of HRQoL in addition to the traditional end points of tumor response and survival results. HRQoL could provide not only complementary clinical data to the primary outcomes, but also more precise predictive and prognostic value. This end point is useful for both clinicians and patients in order to achieve the dogma of precision medicine. The present article examines the use of HRQoL in phase III metastatic colorectal cancer clinical trials, outlines the importance of HRQoL assessment methods, analysis, and results presentation. Moreover, it discusses the relevance of including HRQoL as a primary/co-primary end point to support the progression-free survival results and to assess efficacy of treatment in the advanced disease setting.


Assuntos
Ensaios Clínicos como Assunto , Neoplasias Colorretais/terapia , Qualidade de Vida , Neoplasias Colorretais/fisiopatologia , Intervalo Livre de Doença , Humanos
3.
Ann Oncol ; 28(5): 922-930, 2017 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-28453700

RESUMO

Background: In respect of the principle of autonomy and the right of self-determination, obtaining an informed consent of potential participants before their inclusion in a study is a fundamental ethical obligation. The variations in national laws, regulations, and cultures contribute to complex informed consent documents for patients participating in clinical trials. Currently, only few ethics committees seem willing to address the complexity and the length of these documents and to request investigators and sponsors to revise them in a way to make them understandable for potential participants. The purpose of this work is to focus on the written information in the informed consent documentation for drug development clinical trials and suggests (i) to distinguish between necessary and not essential information, (ii) to define the optimal format allowing the best legibility of those documents. Methods: The Aide et Recherche en Cancérologie Digestive (ARCAD) Group, an international scientific committee involving oncologists from all over the world, addressed these issues and developed and uniformly accepted a simplified informed consent documentation for future clinical research. Results: A simplified form of informed consent with the leading part of 1200-1800 words containing all of the key information necessary to meet ethical and regulatory requirements and 'relevant supportive information appendix' of 2000-3000 words is provided. Conclusions: This position paper, on the basis of the ARCAD Group experts discussions, proposes our informed consent model and the rationale for its content.


Assuntos
Termos de Consentimento , Neoplasias/tratamento farmacológico , Ensaios Clínicos como Assunto , Conhecimentos, Atitudes e Prática em Saúde , Humanos , Consentimento Livre e Esclarecido , Participação do Paciente , Guias de Prática Clínica como Assunto
4.
Oncogene ; 34(17): 2261-71, 2015 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-24931170

RESUMO

The molecular basis for the resistance of tumor cells to cell-mediated cytotoxicity remains poorly understood and thus poses a major challenge for cancer immunotherapy. The present study was designed to determine whether the WNT1-inducible signaling pathway protein 2 (WISP2, also referred to as CCN5), a key regulator of tumor cell plasticity, interferes with tumor susceptibility to cytotoxic T-lymphocyte (CTL)-mediated lysis. We found that silencing WISP2 signaling in human breast adenocarcinoma MCF7 cells impairs CTL-mediated cell killing by a mechanism involving stem cell marker Kruppel-like factor-4 (KLF-4) induction and microRNA-7 (miR-7) downregulation. Inhibition of transforming growth factor beta (TGF-ß) signaling using the A83-01 inhibitor in MCF7-shWISP2 cells resulted in a significant reversal of the epithelial-to-mesenchymal-transitioned (EMT) phenotype, the expression of KLF-4 and a partial recovery of target susceptibility to CTLs. More importantly, we showed that silencing KLF-4 was accompanied by a reduction in MCF7-shWISP2 resistance to CTLs. Using human breast cancer tissues, we demonstrated the coexpression of KLF-4 with EMT markers and TGF-ß pathway signaling components. More importantly, we found that KLF-4 expression was accompanied by miR-7 inhibition, which is partly responsible for impairing CTL-mediated lysis. Thus, our data indicate that WISP2 has a role in regulating tumor cell susceptibility through EMT by inducing the TGF-ß signaling pathway, KLF-4 expression and miR-7 inhibition. These studies indicate for the first time that WISP2 acts as an activator of CTL-induced killing and suggests that the loss of its function promotes evasion of immunosurveillance and the ensuing progression of the tumor.


Assuntos
Neoplasias da Mama/imunologia , Proteínas de Sinalização Intercelular CCN/imunologia , Regulação Neoplásica da Expressão Gênica/imunologia , Imunidade Celular , Fatores de Transcrição Kruppel-Like/imunologia , MicroRNAs/imunologia , RNA Neoplásico/imunologia , Proteínas Repressoras/imunologia , Linfócitos T/imunologia , Neoplasias da Mama/patologia , Proteínas de Sinalização Intercelular CCN/genética , Linhagem Celular Tumoral , Feminino , Humanos , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/genética , MicroRNAs/genética , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/imunologia , RNA Neoplásico/genética , Proteínas Repressoras/genética , Linfócitos T/patologia , Via de Sinalização Wnt
5.
Genet Mol Res ; 12(2): 1440-56, 2013 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-23661467

RESUMO

Chromatin is thought to modulate access of repair proteins to DNA lesions, and may be altered by chromatin remodelers to facilitate repair. We investigated the participation of chromatin remodelers and DNA repair in 5-fluorouracil (5-FU) cytotoxicity in Saccharomyces cerevisiae. 5-FU is an antineoplastic drug commonly used in clinical settings. Among the several strains tested, only those with deficiencies in ATP-dependent chromatin remodeling (CR) and some histone acetyltransferases (HAT) exhibited sensitivity to 5-FU. CR and HAT double-mutants exhibited increased resistance to 5-FU in comparison to the wild-type mutant, but were still arrested in G2/M, as were the sensitive strains. The participation of Htz1p in 5-FU toxicity was also evaluated in single- and double-mutants of CR and HAT; the most significant effect was on cell cycle distribution. 5-FU lesions are repaired by different DNA repair machineries, including homologous recombination (HR) and post-replication repair (PRR). We investigated the role of CR and HAT in these DNA repair pathways. Deficiencies in Nhp10 and CR combined with deficiencies in HR or PRR increased 5-FU sensitivity; however, combined deficiencies of HAT, HR, and PRR did not. CRs are directly recruited to DNA damage and lead to chromatin relaxation, which facilitates access of HR and PRR proteins to 5-FU lesions. Combined deficiencies in HAT with defects in HR and PRR did not potentiate 5-FU cytotoxicity, possibly because they function in a common pathway.


Assuntos
Trifosfato de Adenosina/metabolismo , Montagem e Desmontagem da Cromatina , Fluoruracila/toxicidade , Histona Acetiltransferases/metabolismo , Saccharomyces cerevisiae/efeitos dos fármacos , Saccharomyces cerevisiae/metabolismo , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/genética , Montagem e Desmontagem da Cromatina/genética , Reparo do DNA , DNA Fúngico/genética , DNA Fúngico/metabolismo , Relação Dose-Resposta a Droga , Fluoruracila/metabolismo , Histona Acetiltransferases/genética , Recombinação Homóloga , Testes de Sensibilidade Microbiana , Mutação , Saccharomyces cerevisiae/genética
6.
Br J Pharmacol ; 152(6): 959-79, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17906685

RESUMO

BACKGROUND AND PURPOSE: Glycogen synthase kinase-3 (GSK-3) affects neuropathological events associated with Alzheimers disease (AD) such as hyperphosphorylation of the protein, tau. GSK-3beta expression, enzyme activity and tau phosphorylated at AD-relevant epitopes are elevated in juvenile rodent brains. Here, we assess five GSK-3beta inhibitors and lithium in lowering phosphorylated tau (p-tau) and GSK-3beta enzyme activity levels in 12-day old postnatal rats. EXPERIMENTAL APPROACH: Brain levels of inhibitors following treatment in vivo were optimized based on pharmacokinetic data. At optimal doses, p-tau (Ser(396)) levels in brain tissue was measured by immunoblotting and correlated with GSK-3beta enzyme activities in the same tissues. Effects of GSK inhibitors on p-tau, GSK-3beta activities and cell death were measured in a human neuronal cell line (LUHMES). KEY RESULTS: Lithium and CHIR98014 reduced tau phosphorylation (Ser(396)) in the cortex and hippocampus of postnatal rats, while Alsterpaullone and SB216763 were effective only in hippocampus. AR-A014418 and Indirubin-3'-monoxime were ineffective in either brain region. Inhibition of p-tau in brain required several-fold higher levels of GSK inhibitors than the IC(50) values obtained in recombinant or cell-based GSK-3beta enzyme activity assays. The inhibitory effect on GSK-3beta activity ex vivo correlated with protection against cell death and decrease of p-tau- in LUHMES cells, using low microM inhibitor concentrations. CONCLUSIONS AND IMPLICATIONS: Selective small-molecule inhibitors of GSK-3 reduce tau phosphorylation in vivo. These findings corroborate earlier suggestions that GSK-3beta may be an attractive target for disease-modification in AD and related conditions where tau phosphorylation is believed to contribute to disease pathogenesis.


Assuntos
Inibidores Enzimáticos/farmacologia , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Proteínas tau/metabolismo , Envelhecimento/metabolismo , Animais , Animais Recém-Nascidos , Western Blotting , Encéfalo/crescimento & desenvolvimento , Química Encefálica/fisiologia , Linhagem Celular , Feminino , Quinase 3 da Glicogênio Sintase/metabolismo , Humanos , Imuno-Histoquímica , Imunoprecipitação , Indóis/farmacologia , Cloreto de Lítio/farmacologia , Maleimidas/farmacologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fosforilação , Ratos , Ratos Wistar , Proteínas Recombinantes , Bibliotecas de Moléculas Pequenas , Tiazóis/farmacologia , Extratos de Tecidos/farmacologia
7.
Br J Pharmacol ; 141(6): 1015-23, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-14993096

RESUMO

1. Binding of the novel radioligand (3)H-2-(2-dimethylaminomethyl-phenylsulphanyl)-5-methyl-phenylamine ((3)H-MADAM) to the serotonin transporter (SERT) was used to characterise a range of selective serotonin re-uptake inhibitors (SSRIs) in vitro and in vivo. 2. (3)H-MADAM bound with high affinity in a saturable manner to both human SERT expressed in CHO cells (K(d)=0.20 nm (pK(d)=9.74+/-0.12), B(max)=35+/-4 fmol mg(-1) protein) and mouse cerebral cortex membranes (K(d)=0.21 nm (pK(d)=9.66+/-0.10), B(max)=50+/-24 fmol mg(-1) protein). 3. Binding of (3)H-MADAM was highly selective for SERT in vitro as demonstrated by the in vitro profile of MADAM tested at 75 different receptors, ion channels and transporters. This was further substantiated by the pharmacological profile of the binding. Hence, the binding of (3)H-MADAM was potently inhibited by SSRIs but not by selective inhibitors of noradrenaline transport and dopamine transport. Likewise, a 5-HT(2A/2C) receptor antagonist did not inhibit (3)H-MADAM binding. 4. (3)H-MADAM binding in vivo was inhibited only by compounds which also inhibited the binding of (3)H-MADAM in vitro (the SSRIs, mixed SERT/noradrenaline transport inhibitors and clomipramine), confirming the selectivity of (3)H-MADAM for SERT also in vivo. Moreover, compounds effective in inhibiting (3)H-MADAM binding were the only ones found to be active in the mouse 5-HTP potentiation test confirming the model as a behavioural correlate to in vivo 5-HT uptake. 5. Finally, it was found that a SERT occupancy of 85-95% was necessary to produce 50% of the maximum behavioural response (ED(50)).


Assuntos
Benzilaminas/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Inibidores Seletivos de Recaptação de Serotonina/metabolismo , 5-Hidroxitriptofano/administração & dosagem , 5-Hidroxitriptofano/antagonistas & inibidores , 5-Hidroxitriptofano/farmacologia , Animais , Antidepressivos Tricíclicos/farmacologia , Comportamento Animal/efeitos dos fármacos , Benzilaminas/farmacologia , Ligação Competitiva , Células CHO , Células Cultivadas , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Cricetinae , Sinergismo Farmacológico , Masculino , Camundongos , Camundongos Endogâmicos , Antagonistas da Serotonina/farmacologia , Proteínas da Membrana Plasmática de Transporte de Serotonina , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Sinaptossomos/efeitos dos fármacos , Sinaptossomos/metabolismo , Trítio
8.
FASEB J ; 15(12): 2288-90, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11511510

RESUMO

Although initiation of chromosome condensation during early prophase is linked temporally to the appearance of the mitotic cdc2 kinase in the nucleus, it is not known what targets the kinase to the nucleus and how this is coupled to chromatin remodeling. We now report that cdc2 kinase forms stable molecular complexes with the nuclear enzyme DNA topoisomerase II, which is associated with marked stimulation of both DNA binding and catalytic activity of topoisomerase II, albeit in a phosphorylation-independent manner. The molecular interaction is required for recruitment of cdc2 kinase, as shown by incubation of purified enzymes with chicken erythrocyte nuclei, which have neither endogenous topoisomerase II nor cdc2 kinase. The physical association between the two enzymes alters the DNA/topoisomerase II interaction as shown by pulse-field electrophoresis after incubation of intact nuclei with the specific topoisomerase II inhibitor VM-26. Furthermore, the presence of both enzymes, but not either enzyme alone, is accompanied by extensive chromatin remodeling converting the interphase nuclei into precondensation chromosomes with striking resemblance to early prophase structures. Our results reveal a novel property of cyclin-dependent kinases and demonstrate that the recruitment of cdc2 kinase by topoisomerase II is coupled to chromatin remodeling.


Assuntos
Proteína Quinase CDC2/metabolismo , Cromatina/fisiologia , DNA Topoisomerases Tipo II/metabolismo , DNA Topoisomerases Tipo II/fisiologia , Transporte Ativo do Núcleo Celular , Animais , Núcleo Celular/metabolismo , Células Cultivadas , DNA/metabolismo , Modelos Genéticos , Prófase
9.
Cancer Res ; 61(7): 2961-7, 2001 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-11306474

RESUMO

BN 80915 is the lead compound from a novel class of E-ring modified camptothecin analogues, the homocamptothecins, which show potent antitumor activities in animal models. Here, we report that BN 80915 induces up to 2-fold more cleavable complexes between plasmid DNA and purified human topoisomerase I than SN-38 and camptothecin. BN 80915 also induces DNA-topoisomerase I complexes in living HT-29 colon carcinoma cells, as shown by the in vivo link assay. BN 80915 is an extremely potent inducer of DNA-protein complexes in these cells starting at a concentration of 5 nM in the media. BN 80915 is clearly more potent than SN-38, because at least 20 times more SN-38 is needed to induce comparable levels of cleavable complexes. Kinetic experiments show that BN 80915 induces cleavable complexes within minutes that remain stable for at least 6 h in the presence of drug. Whereas the majority of the complexes are reversed within 15 min after drug removal, a substantial fraction (30%) persists for at least 4 h, in contrast with SN-38-treated cells, where all complexes have disappeared by this time. BN 80915 shows strong antiproliferative effects toward HT-29 cells with an IC50 of 0.3 nM compared with 20 nM for SN-38 and 40 nM for topotecan. BN 80915 is also potent against other colon carcinoma cells as well as toward cells growing in three dimensions as multicellular spheroids. HL-60 cells expressing functional P-glycoprotein or multidrug resistance protein show no cross-resistance toward BN 80915. Taken together, our results show that BN 80915 is unusually potent toward human colon carcinoma cells because of the formation of high levels of stable, covalent DNA-topoisomerase complexes.


Assuntos
Antineoplásicos/farmacologia , Camptotecina/farmacocinética , Neoplasias do Colo/tratamento farmacológico , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Transportadores de Cassetes de Ligação de ATP/metabolismo , Células CACO-2/efeitos dos fármacos , Camptotecina/análogos & derivados , Divisão Celular/efeitos dos fármacos , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , DNA Topoisomerases Tipo I/metabolismo , DNA Super-Helicoidal/efeitos dos fármacos , DNA Super-Helicoidal/metabolismo , Inibidores do Crescimento/farmacologia , Células HT29/efeitos dos fármacos , Humanos , Cinética , Proteínas Associadas à Resistência a Múltiplos Medicamentos , Esferoides Celulares/efeitos dos fármacos , Células Tumorais Cultivadas/efeitos dos fármacos
10.
J Biol Chem ; 275(44): 34710-8, 2000 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-10942766

RESUMO

DNA topoisomerase II alpha is required for chromatin condensation during prophase. This process is temporally linked with the appearance of mitosis-specific phosphorylation sites on topoisomerase IIalpha including one recognized by the MPM-2 monoclonal antibody. We now report that the ability of mitotic extracts to create the MPM-2 epitope on human topoisomerase II alpha is abolished by immunodepletion of protein kinase CK2. Furthermore, the MPM-2 phosphoepitope on topoisomerase II alpha can be generated by purified CK2. Phosphorylation of C-truncated topoisomerase II alpha mutant proteins conclusively shows, that the MPM-2 epitope is present in the last 163 amino acids. Use of peptides containing all conserved CK2 consensus sites in this region indicates that only the peptide containing Arg-1466 to Ala-1485 is able to compete with topoisomerase II alpha for binding of the MPM-2 antibody. Replacement of Ser-1469 with Ala abolishes the ability of the phosphorylated peptide to bind to the MPM-2 antibody while a peptide containing phosphorylated Ser-1469 binds tightly. Surprisingly, the MPM-2 phosphoepitope influences neither the catalytic activity of topoisomerase II alpha nor its ability to form molecular complexes with CK2 in vitro. In conclusion, we have identified protein kinase CK2 as a new MPM-2 kinase able to phosphorylate an important mitotic protein, topoisomerase II alpha, on Ser-1469.


Assuntos
Proteínas de Ciclo Celular , DNA Topoisomerases Tipo II , DNA Topoisomerases Tipo II/metabolismo , Isoenzimas/metabolismo , Mitose , Fosfoproteínas/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Serina/metabolismo , Sequência de Aminoácidos , Animais , Antígenos de Neoplasias , Caseína Quinase II , Catálise , Extratos Celulares , Cromossomos Humanos , DNA Topoisomerases Tipo II/química , Proteínas de Ligação a DNA , Guanosina Trifosfato/metabolismo , Células HeLa , Heparina/metabolismo , Humanos , Isoenzimas/antagonistas & inibidores , Isoenzimas/química , Cinesinas , Dados de Sequência Molecular , Fosforilação , Homologia de Sequência de Aminoácidos , Inibidores da Topoisomerase II
11.
Pharmacol Ther ; 85(3): 217-29, 2000 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10739876

RESUMO

The resistance of tumor cells to anticancer agents remains a major cause of treatment failure in cancer patients. The term multidrug resistance (MDR) is used to define a resistance phenotype where cells are resistant to multiple drugs with no obvious structural resemblance and with different molecular targets. It is now clear that MDR is always multifactorial. The intracellular drug distribution is modified in many MDR cell lines, leading to increased drug sequestration in acidic vesicles, such as the trans-Golgi apparatus, recycling endosomes, and lysosomes, followed by transport to the plasma membrane and extrusion into the external medium. Since most anticancer agents target DNA or nuclear enzymes, sequestration of drug in cytoplasmic organelles will lead to decreased drug-target interaction and thereby, decreased cytotoxicity. Altered intracellular drug distribution is usually associated with the expression of drug efflux pumps, such as the P-glycoprotein and the multidrug resistance protein. Another common modification in MDR cells is alkalization of the intracellular pH. The relationship between these different resistance mechanisms is reviewed and a model proposed that suggests why these different resistance mechanisms are co-expressed in multiple cell lines.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Transportadores de Cassetes de Ligação de ATP/metabolismo , Antineoplásicos/farmacologia , Resistência a Múltiplos Medicamentos , Organelas/química , Antineoplásicos/farmacocinética , Transporte Biológico , DNA de Neoplasias/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Proteínas de Neoplasias/metabolismo , Células Tumorais Cultivadas , Partículas de Ribonucleoproteínas em Forma de Abóbada/metabolismo
12.
Pathol Oncol Res ; 5(3): 171-8, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10491013

RESUMO

Mammalian DNA topoisomerase I is a multifunctional enzyme which is essential for embryonal development. In addition to its classical DNA nicking-closing activities which are needed for relaxation of supercoiled DNA, topoisomerase I can phosphorylate certain splicing factors. The enzyme is also involved in transcriptional regulation through its ability to associate with other proteins in the TFIID-, and possibly TFIIH-, transcription complexes, and is implicated in the recognition of DNA lesions. Finally, topoisomerase I is a recombinase which can mediate illegitimate recombination. A crucial reaction intermediate during relaxation of DNA is the formation of a DNA-topoisomerase I complex (the cleavable complex) where topoisomerase I is covalently linked to a 3 -end of DNA thereby creating a single stranded DNA break. Cleavable complexes are also formed in the vicinity of DNA lesions and in the presence of the antitumor agent, camptothecin. While formation of cleavable complexes may be necessary for the initial stages of the DNA damage response, these complexes are also potentially dangerous to the cell due to their ability to mediate illegitimate recombination, which can lead to genomic instability and oncogenesis. Thus the levels and stability of these complexes have to be strictly regulated. This is obtained by maintaining the enzyme levels relatively constant, by limiting the stability of the cleavable complexes through physical interaction with the oncogene suppressor protein p53 and by degradation of the topoisomerase I by the proteasome system. Emerging evidence suggest that these regulatory functions are perturbed in tumor cells, explaining at the same time why topoisomerase I activities so often are increased in certain human tumors, and why these cells are sensitized to the cytotoxic effects of camptothecins.


Assuntos
DNA Topoisomerases Tipo I/genética , DNA Topoisomerases Tipo I/metabolismo , Neoplasias/enzimologia , Cromossomos Humanos Par 20 , Dano ao DNA , DNA Super-Helicoidal/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Modelos Genéticos , Fosforilação , Recombinação Genética , Transcrição Gênica
13.
Biochimie ; 81(7): 771-9, 1999 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10492025

RESUMO

DNA topoisomerase II regulates the three-dimensional organisation of DNA and is the principal target of many important anticancer and antimicrobial agents. These drugs usually act on the DNA cleavage/religation steps of the catalytic cycle resulting in accumulation of covalent DNA-topoisomerase II complexes. We have studied the different steps of the catalytic cycle as a function of salt concentration, which is a classical way to evaluate the biochemical properties of proteins. The results show that the catalytic activity of topoisomerase II follows a bell-shaped curve with optimum between 100 and 225 mM KCl. No straight-forward correlation exists between DNA binding and catalytic activity. The highest levels of drug-induced covalent DNA-topoisomerase II complexes are observed between 100 and 150 mM KCl. Remarkably, at salt concentrations between 150 mM and 225 mM KCl, topoisomerase II is converted into a drug-resistant form with greatly reduced levels of drug-induced DNA-topoisomerase II complexes. This is due to efficient religation rather than to absence of DNA cleavage as witnessed by relaxation of the supercoiled DNA substrate. In the absence of DNA, ATP hydrolysis is strongest at low salt concentrations. Unexpectedly, the addition of DNA stimulates ATP hydrolysis at 100 and 150 mM KCl, but has little or no effect below 100 mM KCl in spite of strong non-covalent DNA binding at these salt concentrations. Therefore, DNA-stimulated ATP hydrolysis appears to be associated with covalent rather than non-covalent binding of DNA to topoisomerase II. Taken together, the results suggest that it is the DNA cleavage/religation steps that are most closely associated with the catalytic activities of topoisomerase II providing a unifying theme for the biological and pharmacological modulation of this enzyme.


Assuntos
DNA Topoisomerases Tipo II/metabolismo , DNA/metabolismo , Trifosfato de Adenosina/metabolismo , Catálise , Hidrólise , Concentração Osmolar , Ligação Proteica
14.
Proc Natl Acad Sci U S A ; 96(18): 10355-60, 1999 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-10468612

RESUMO

DNA topoisomerase I is a nuclear enzyme involved in transcription, recombination, and DNA damage recognition. Previous studies have shown that topoisomerase I interacts directly with the tumor-suppressor protein p53. p53 is a transcription factor that activates certain genes through binding to specific DNA sequences. We now report that topoisomerase I can be stimulated by both latent and activated wild-type p53 as well as by several mutant and truncated p53 proteins in vitro, indicating that sequence-specific DNA-binding and stimulation of topoisomerase I are distinct properties of p53. These assays also suggest that the binding site for topoisomerase I on p53 is between amino acids 302 and 321. In living cells, the interaction between p53 and topoisomerase I is strongly dependent on p53 status. In MCF-7 cells, which have wild-type p53, the association between the two proteins is tightly regulated in a spatial and temporal manner and takes place only during brief periods of genotoxic stress. In marked contrast, the two proteins are constitutively associated in HT-29 cells, which have mutant p53. These findings have important implications for both cellular stress response and genomic stability, given the ability of topoisomerase I to recognize DNA lesions as well as to cause illegitimate recombination.


Assuntos
DNA Topoisomerases Tipo I/metabolismo , DNA/metabolismo , Oligodesoxirribonucleotídeos/metabolismo , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Animais , Sequência de Bases , Sítios de Ligação , Neoplasias da Mama , Camptotecina/farmacologia , Catálise , Linhagem Celular , DNA/química , DNA Topoisomerases Tipo I/isolamento & purificação , Feminino , Cinética , Mitomicina/farmacologia , Mutagênese Sítio-Dirigida , Oligodesoxirribonucleotídeos/química , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Deleção de Sequência , Fatores de Transcrição/metabolismo , Transfecção , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/isolamento & purificação
15.
Int J Cancer ; 81(4): 580-7, 1999 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-10225448

RESUMO

We have previously shown that in myeloid leukemic cells, daunorubicin (DNR) induces apoptosis via the activation of the sphingomyelin-ceramide pathway. We have now investigated sphingomyelin (SM) hydrolysis, ceramide generation, and apoptosis in vincristine-selected multidrug resistant (MDR) HL-60 cells (HL-60/Vinc), compared with their parental counterparts. We show that DNR triggers the SM cycle (stimulation of neutral sphingomyelinase, SM hydrolysis, and ceramide generation) and apoptosis in both parental and MDR cells, when used at isotoxic doses (ie., 1 and 100 microM for HL-60 and HL-60/Vinc, respectively). However, in MDR cells treated with either 10 microM DNR or 1 microM DNR in association with the P-glycoprotein (P-gp) blocker verapamil (treatment conditions which yield an intracellular DNR concentration similar to that achieved with 1 microM in the parental cells), we were unable to detect SM hydrolysis, ceramide generation and apoptosis. This implies that inhibition of the DNR-induced SM cycle in MDR cells is not directly related to P-gp. We have also investigated the influence of intracellular drug localization on the DNR-induced SM-cycle in HL-60/Vinc cells. In these cells, DNR at 10 microM is mainly localized in cytoplasmic vesicles, while the drug is diffusely distributed when used at 100 microM. A diffuse distribution pattern was also observed when MDR cells were treated with 1 microM DNR in association with the cyclosporine derivative PSC-833, but not with verapamil. In parallel, PSC-833, but not verapamil, restored the induction of the SM cycle and the apoptotic potential of DNR, and markedly increased drug cytotoxicity in MDR cells. Our results suggest that altered intracellular drug transport plays an important role in limiting ceramide generation and cell death in MDR cells.


Assuntos
Apoptose/fisiologia , Ceramidas/metabolismo , Daunorrubicina/toxicidade , Resistência a Múltiplos Medicamentos/fisiologia , Células HL-60/fisiologia , Transdução de Sinais/fisiologia , Esfingomielinas/metabolismo , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Ceramidas/farmacologia , Ciclosporinas/farmacologia , Fragmentação do DNA , Células HL-60/citologia , Células HL-60/efeitos dos fármacos , Humanos , Cinética , Transdução de Sinais/efeitos dos fármacos , Esfingomielina Fosfodiesterase/metabolismo , Fatores de Tempo , Vincristina/toxicidade
16.
Br J Cancer ; 79(7-8): 1090-7, 1999 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10098741

RESUMO

Exposure of some acute myeloid leukaemia (AML) cells to daunorubicin leads to rapid cell death, whereas other AML cells show natural drug resistance. This has been attributed to expression of functional P-glycoprotein resulting in reduced drug accumulation. However, it has also been proposed that P-glycoprotein-expressing multidrug-resistant (MDR) cells are inherently defective for apoptosis. To distinguish between these different possibilities, we have compared the cell death process in a human AML cell line (HL-60) with a MDR subline (HL-60/Vinc) at doses that yield either similar intracellular daunorubicin concentrations or comparable cytotoxicity. Adjustment of the dose to obtain the same intracellular drug accumulation in the two cell lines did not result in equal cytotoxicity, suggesting the presence of additional resistance mechanisms in the P-glycoprotein-expressing HL-60/Vinc cells. However, at equitoxic doses, similar cell death pathways were observed. In HL-60 cells, daunorubicin induced rapid apoptosis at 0.5-1 microM and delayed mitotic cell death at 0.1 microM. These concentrations are within the clinical dose range. Similarly, HL-60/Vinc cells underwent apoptosis at 50-100 microM daunorubicin and mitotic cell death at 10 microM. These results show, for the first time, that anthracyclines can induce cell death by a dual mechanism in both sensitive and MDR cells. Our results also show that not only the cytotoxicity, but also the kinetics and mechanism of cell death, are dose dependent. Interestingly, regrowth was observed only in association with delayed cell death and the formation of enlarged, often polyploid, cells with micronucleation, suggesting that morphological criteria may be useful to evaluate treatment efficacy in patients with myeloid leukaemias.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Antibióticos Antineoplásicos/farmacologia , Morte Celular , Daunorrubicina/farmacologia , Resistência a Múltiplos Medicamentos , Células HL-60/efeitos dos fármacos , Mitose , Proteínas de Neoplasias/metabolismo , Antibióticos Antineoplásicos/farmacocinética , Apoptose/genética , Área Sob a Curva , Fragmentação do DNA , DNA Nucleotidilexotransferase/análise , Daunorrubicina/farmacocinética , Relação Dose-Resposta a Droga , Resistencia a Medicamentos Antineoplásicos , Células HL-60/metabolismo , Humanos
17.
Acta Biochim Pol ; 45(2): 535-44, 1998.
Artigo em Inglês | MEDLINE | ID: mdl-9821882

RESUMO

DNA topoisomerases regulate the organization of DNA and are important targets for many clinically used antineoplastic agents. In addition, DNA topoisomerases modulate the cellular sensitivity toward a number of DNA damaging agents. Increased topoisomerase II activities were shown to contribute to the resistance of both nitrogen mustard- and cisplatin-resistant cells. Similarly, cells with decreased topoisomerase II levels show increased sensitivity to cisplatin, carmustine, mitomycin C and nitrogen mustard. Recent studies propose that topoisomerases may be involved in damage recognition and DNA repair at several different levels including: 1) the initial recognition of DNA lesions; 2) DNA recombination; and 3) regulation of DNA structure. The stress-activated oncogene suppressor protein p53 can modulate the activity of at least three different human topoisomerases, either directly by molecular associations or by transcriptional regulation. Since DNA topoisomerases have considerable recombinase activities, inappropriately activated topoisomerases in tumor cells lacking functional p53 may contribute to the genetic instability of these cells.


Assuntos
Reparo do DNA , DNA Topoisomerases Tipo I , Proteína Supressora de Tumor p53 , Animais , Cricetinae , DNA/genética , Humanos , Recombinação Genética , Células Tumorais Cultivadas
18.
Biochim Biophys Acta ; 1400(1-3): 257-74, 1998 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-9748618

RESUMO

DNA topoisomerase inhibitors are important antineoplastic agents used in the treatment of both leukemias and solid tumors, such as breast, lung and colon cancers. Their clinical usefulness is limited by both natural and acquired tumor cell resistance, which almost always is multifactorial in nature. The resistance can be due to pretarget events, such as drug accumulation, metabolism and intracellular drug distribution, or due to reduced drug-target interaction. More recently, post-target events, such as macromolecular synthesis, cell cycle progression, DNA repair/recombination and regulation of cell death, have been shown to play an important role in the sensitivity toward topoisomerase inhibitors. The different mechanisms involved in the cellular resistance toward clinically used topoisomerase inhibitors will be reviewed in this article with particular emphasis on post-target events.


Assuntos
Antineoplásicos/metabolismo , Inibidores Enzimáticos/metabolismo , Inibidores da Topoisomerase I , Ciclo Celular/fisiologia , Morte Celular/fisiologia , Divisão Celular/efeitos dos fármacos , Reparo do DNA/genética , DNA Topoisomerases Tipo I/genética , Resistência a Medicamentos/fisiologia , Humanos , Mutação/genética , Neoplasias/terapia , Recombinação Genética/genética
19.
Biochimie ; 80(3): 247-54, 1998 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-9615864

RESUMO

We describe in this review the mechanisms of resistance to topoisomerase II inhibitors that have been identified in cell lines rendered resistant to drugs. They concern especially both quantitative and qualitative alterations of topoisomerase II, leading to drug insensitivity of the cells. Expression and activity of topoisomerase II have also been studied in a number of tumor specimens originating from patients, but the role of topoisomerase II alterations in drug resistance in the clinical setting has not yet been firmly established. It would be worthwhile, however, to develop predictive assays for drug activity in human cancers, based upon the topoisomerase II status of tumor samples.


Assuntos
Resistencia a Medicamentos Antineoplásicos , Inibidores Enzimáticos/farmacologia , Inibidores da Topoisomerase II , Animais , Resistência a Múltiplos Medicamentos , Humanos , Células Tumorais Cultivadas
20.
Blood ; 91(10): 3711-23, 1998 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-9573008

RESUMO

During megakaryocyte differentiation, the promegakaryoblast (immature megakaryocyte) increases its ploidy to a 2(x) DNA content by a poorly understood process called endomitosis. This leads to the formation of a giant cell, the megakaryocyte (MK), which subsequently gives rise to platelets. In this report, we show that endomitosis of human MKs is due to abortive mitosis. Human MKs were obtained by a two-step purification of CD34(+) blood or marrow precursors followed by in vitro culture in the presence of MK growth factors. Microscopic examination shows that a large number of centrosomes (up to 32) and centrioles are present in polyploid MKs. After nocodazole treatment, more than 20% of the MK are blocked in a typical pseudo-metaphase. Both spontaneous and nocodazole-induced endomitosis are associated with a breakdown of the nuclear envelope and possess a complex mitotic spindle composed of several asters. Spindle microtubules radiate from each aster, creating a spherical structure. At metaphase, expression of the kinetochore phosphoepitope recognized by the 3F3/2 antibody is lost, and the sister chromatids segregate moving toward the spindle poles. After limited segregation, the chromosomes decondense and the nuclear envelope reforms in the absence of cytokinesis, isolating all chromosomes in a single nucleus. It has been proposed that endomitosis could be due to an abnormal CDK1 activity or an absence of cyclin B1. Our results show that cyclin B1 can be detected in all MKs, including those with a ploidy of 8N or more. The cyclin B1 staining colocalizes with the mitotic spindle. Using flow cytometry, the level of cyclin B1 increased until 8N, but remained identical in 16N and 32N MKs. Cell sorting was used to separate the MKs into a 2N/4N and >4N population. Both cyclin B1 and CDK1 could be detected in the endomitotic polyploid MKs using Western blot analysis, and a histone H1 kinase activity was associated with immunoprecipitated cyclin B1. We conclude that endomitosis of human MKs is due to abortive mitosis, possibly due to alterations in the regulation of mitotic exit.


Assuntos
Megacariócitos/citologia , Mitose , Antígenos CD34/análise , Proteína Quinase CDC2/fisiologia , Diferenciação Celular , Separação Celular , Células Cultivadas , Centríolos/ultraestrutura , Centrossomo/ultraestrutura , Ciclina B/fisiologia , Ciclina B1 , Citometria de Fluxo , Humanos , Megacariócitos/efeitos dos fármacos , Nocodazol/farmacologia , Ploidias , Polietilenoglicóis/farmacologia , Proteínas Recombinantes/farmacologia , Fuso Acromático/efeitos dos fármacos , Fuso Acromático/ultraestrutura , Trombopoetina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...