Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Sci Rep ; 13(1): 3759, 2023 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-36882567

RESUMO

This study aimed to develop a nomogram for predicting the progression-free survival (PFS) of testicular germ cell tumors (TGCT) patients based on DNA methylation signature and clinicopathological characteristics. The DNA methylation profiles, transcriptome data, and clinical information of TGCT patients were obtained from the Cancer Genome Atlas (TCGA) database. Univariate Cox, lasso Cox, and stepwise multivariate Cox regression were applied to identify a prognostic CpG sites-derived risk signature. Differential expression analysis, functional enrichment analysis, immunoinfiltration analysis, chemotherapy sensitivity analysis, and clinical feature correlation analysis were performed to elucidate the differences among risk groups. A prognostic nomogram integrating CpG sites-derived risk signature and clinicopathological features was further established and evaluated likewise. A risk score model based on 7 CpG sites was developed and found to exhibit significant differences among different survival, staging, radiotherapy, and chemotherapy subgroups. There were 1452 differentially expressed genes between the high- and low-risk groups, with 666 being higher expressed and 786 being lower expressed. Genes highly expressed were significantly enriched in immune-related biological processes and related to T-cell differentiation pathways; meanwhile, down-regulated genes were significantly enriched in extracellular matrix tissue organization-related biological processes and involved in multiple signaling pathways such as PI3K-AKT. As compared with the low-risk group, patients in the high-risk group had decreased lymphocyte infiltration (including T-cell and B-cell) and increased macrophage infiltration (M2 macrophages). They also showed decreased sensitivity to etoposide and bleomycin chemotherapy. Three clusters were obtained by consensus clustering analysis based on the 7 CpG sites and showed distinct prognostic features, and the risk scores in each cluster were significantly different. Multivariate Cox regression analysis found that the risk scores, age, chemotherapy, and staging were independent prognostic factors of PFS of TGCT, and the results were used to formulate a nomogram model that was validated to have a C-index of 0.812. Decision curve analysis showed that the nomogram model was superior to other strategies in the prediction of PFS of TGCT. In this study, we successfully established CpG sites-derived risk signature, which might serve as a useful tool in the prediction of PFS, immunoinfiltration, and chemotherapy sensitivity for TGCT patients.


Assuntos
Metilação de DNA , Neoplasias Embrionárias de Células Germinativas , Humanos , Metilação de DNA/genética , Intervalo Livre de Progressão , Fosfatidilinositol 3-Quinases/genética , Neoplasias Embrionárias de Células Germinativas/genética
2.
Front Oncol ; 11: 734587, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34568070

RESUMO

PURPOSE: Exosomes could be released directly into the urine by the urological tumoral cells, so testing urinary exosomes has great potential for non-invasive diagnosis and monitor of urological tumors. The objective of this study is to systematically review and meta-analysis of urinary exosome for urological tumors diagnosis. MATERIALS AND METHODS: A systematic review of the recent English-language literature was conducted according to the PRISMA statement recommendations (CRD42021250613) using PubMed, Embase, Cochrane Library, Web of Science, and Scopus databases up to April 30, 2021. Risk-of-bias assessment was performed according to the QUADAS 2 tool. The true diagnostic value of urinary exosomes by calculating the number of true positive, false positive, true negative, and false negative, diagnoses by extracting specificity and sensitivity data from the selected literature. RESULTS: Sixteen eligible studies enrolling 3224 patients were identified. The pooled sensitivity and specificity of urinary exosomes as a diagnostic tool in urological tumors were 83% and 88%, respectively. The area under the summary receiver operating characteristic curve was 0.92 (95% CI: 0.89-0.94). Further subgroup analyses showed that our results were stable irrespective of the urinary exosome content type and tumor type. CONCLUSION: Urinary exosomes may serve as novel non-invasive biomarkers for urological cancer detection. Future clinical trial designs must validate and explore their utility in treatment decision-making. SYSTEMATIC REVIEW REGISTRATION: [ https://www.crd.york.ac.uk/prospero/], identifier [CRD42021250613].

3.
Front Mol Biosci ; 8: 683844, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34458320

RESUMO

Cell-free DNA (cf-DNA) has been reported to represent a suitable material for liquid biopsy in the diagnosis and prognosis of various cancers. We performed a meta-analysis of published data to investigate the diagnostic value of cf-DNA for renal cancer (RCa). Systematic searches were conducted using Pubmed, Embase databases, Web of Science, Medline and Cochrane Library to identify relevant publications until the 31st March 2021. For all patients, we evaluated the true diagnostic value of cf-DNA by calculating the number of true positive, false positive, true negative, and false negative, diagnoses by extracting specificity and sensitivity data from the selected literature. In total, 8 studies, featuring 754 RCa patients, and 355 healthy controls, met our inclusion criteria. The overall diagnostic sensitivity and specificity for cf-DNA was 0.71 (95% confidence interval (CI), 0.55-0.83) and 0.79 (95% CI, 0.66-0.88), respectively. The pooled positive likelihood ratio and pooled negative likelihood ratio were 3.42 (95% CI, 2.04-5.72) and 0.36 (95% CI, 0.23-0.58), respectively. The area under the summary receiver operating characteristic curve was 0.82 (95% CI, 0.79-0.85), and the diagnostic odds ratio was 7.80 (95% CI, 4.40-13.85). Collectively, our data demonstrate that cf-DNA has high specificity and sensitivity for diagnosing RCa. Therefore, cf-DNA is a useful biomarker for the diagnosis of RCa.

4.
Bioorg Chem ; 107: 104536, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33342565

RESUMO

Caffeic acid phenethyl ester (CAPE), a bioactive component extracted from propolis of honeybee hives, can inhibit hepatocellular carcinoma (HCC). In order to explore more stable CAPE derivatives, 25 compounds were designed, synthesized, and pharmacologically assessed in vitro and in vivo as anti-tumor agents in HCC. Compounds 8d, 8f, 8l, 8j, and 8k showed favorable antiproliferative activity than other compounds including CAPE in the HCC cell lines. Based on the result of QTRP (Quantitative Thiol Reactivity Profiling), epidermal growth factor receptor (EGFR) and C-terminal Src kinase (CSK) were supposed to the targets of 8f, which was confirmed by binding mode analysis. Furthermore, compounds 8f, 8l, 8j, 8k, 8g, and 8h showed potent inhibitory effects against both CSK and EGFR than other derivatives in an ADP-Glo™ kinase assay. The representative compound, 8f, potently inhibited various tumor growth in murine model including murine hepatocellular carcinoma H22, meanwhile downregulating the EGFR/AKT pathway and enhancing T cell proliferation through inhibition of CSK. Metabolic stability in vitro suggested 8f and 8k were more stable in mouse plasma than CAPE and susceptible to metabolism in liver microsomes. The overall excellent profile of compound 8f makes it a potential candidate for further preclinical investigation.


Assuntos
Ácidos Cafeicos/uso terapêutico , Carcinoma Hepatocelular/tratamento farmacológico , Receptores ErbB/antagonistas & inibidores , Neoplasias Hepáticas/tratamento farmacológico , Álcool Feniletílico/análogos & derivados , Quinases da Família src/antagonistas & inibidores , Animais , Antineoplásicos/química , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Sítios de Ligação , Ácidos Cafeicos/química , Ácidos Cafeicos/farmacologia , Carcinoma Hepatocelular/patologia , Domínio Catalítico , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Regulação para Baixo/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Receptores ErbB/metabolismo , Feminino , Neoplasias Hepáticas/patologia , Masculino , Camundongos , Simulação de Acoplamento Molecular , Álcool Feniletílico/química , Álcool Feniletílico/farmacologia , Álcool Feniletílico/uso terapêutico , Relação Estrutura-Atividade , Quinases da Família src/metabolismo
5.
Oncol Rep ; 44(6): 2455-2464, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33125130

RESUMO

Zinc finger protein 403 (ZFP403), located on human chromosome 17q12­21, is closely associated with the development of cancer. However, to date, there are a limited number of studies on the biological functions of this gene, particularly in prostate cancer (PCa). The results of the present study demonstrated that compared with normal tissues, the expression of ZFP403 was markedly lower in PCa tissues, as shown by the evaluation of the Gene Expression Profiling Interactive Analysis 2 database. The decreased expression of ZFP403 in PCa clinical tissues and cell lines was confirmed by immunohistochemistry, reverse transcription­quantitative PCR and western blot analysis. Using short harpin (sh)RNA inhibition, stably­silenced ZFP403 cell lines were then constructed by lentiviral transfection (LV­PC3­shRNA­1 and 2; LV­DU145­shRNA­1 and 2). The results revealed that the knockdown of ZFP403 in PCa cells promoted cellular proliferation, colony formation, migration and invasiveness in vitro. Moreover, the levels of tumor growth­ and motility­related proteins were significantly altered after ZFP403­knockdown. A xenograft tumor model using nude mice was established to elucidate the role of ZFP403 in tumorigenesis in vivo. Tumor growth was significantly increased in mice injected with ZFP403­knockdown cells compared with the control mice. Overall, the findings of the present study demonstrate that ZFP403 functions as a tumor suppressor gene in PCa by affecting the proliferation, migration and invasiveness of PCa cells, suggesting its potential use as a clinical diagnostic marker.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Biomarcadores Tumorais/genética , Carcinogênese/patologia , Regulação Neoplásica da Expressão Gênica , Neoplasias da Próstata/genética , Proteínas Adaptadoras de Transdução de Sinal/análise , Idoso , Animais , Apoptose/genética , Biomarcadores Tumorais/análise , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Regulação para Baixo , Técnicas de Silenciamento de Genes , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Invasividade Neoplásica/genética , Próstata/patologia , Neoplasias da Próstata/diagnóstico , Neoplasias da Próstata/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Food Sci Nutr ; 8(6): 2882-2894, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32566206

RESUMO

Metabolomic studies were carried out using gas chromatography and mass spectrometry (GC-MS) on Daohuaxiang variety rice (Oryza sativa L.) from the Wuchang Geographical Indication Rice Protection Area in Heilongjiang Province, to investigate the effects of storage on brown rice metabolism. The data were subjected to principal component analysis (PCA), orthogonal partial least squares-discriminant analysis (OPLS-DA), and cluster analysis using software such as SIMCA. Analysis of the samples led to the identification of a total of 160 metabolites. No significant differences were found in the amount of metabolites before and after storage. A total of 31 differential metabolites were screened, and the changes in metabolite content showed a "reverse change" overall. Storage significantly changed the content of various metabolites in rice, with fatty acids impacted most significantly. Metabolic pathway analysis revealed that fatty acid biosynthesis is a key metabolic pathway in rice storage. The degradation of brown rice quality caused by storage is closely related to the composition and content of its metabolites, and that change in lipid content significantly affects brown rice quality during storage.

7.
Cancer Chemother Pharmacol ; 85(4): 661-672, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32072218

RESUMO

PURPOSE: We postulate that the deoxyguanosine analogue CNDAG [9-(2-C-cyano-2-deoxy-1-ß-D-arabino-pentofuranosyl)guanine] likely causes a single-strand break after incorporation into DNA, similar to the action of its cytosine congener CNDAC, and that subsequent DNA replication across the unrepaired nick would generate a double-strand break. This study aimed at identifying cellular responses and repair mechanisms for CNDAG prodrugs, 2-amino-9-(2-C-cyano-2-deoxy-1-ß-D-arabino-pentofuranosyl)-6-methoxy purine (6-OMe) and 9-(2-C-cyano-2-deoxy-1-ß-D-arabino-pentofuranosyl)-2,6-diaminopurine (6-NH2). Each compound is a substrate for adenosine deaminase, the action of which generates CNDAG. METHODS: Growth inhibition assay, clonogenic survival assay, immunoblotting, and cytogenetic analyses (chromosomal aberrations and sister chromatid exchanges) were used to investigate the impact of CNDAG on cell lines. RESULTS: The 6-NH2 derivative was selectively potent in T cell malignant cell lines. Both prodrugs caused increased phosphorylation of ATM and its downstream substrates Chk1, Chk2, SMC1, NBS1, and H2AX, indicating activation of ATM-dependent DNA damage response pathways. In contrast, there was no increase in phosphorylation of DNA-PKcs, which participates in repair of double-strand breaks by non-homologous end-joining. Deficiency in ATM, RAD51D, XRCC3, BRCA2, and XPF, but not DNA-PK or p53, conferred significant clonogenic sensitivity to CNDAG or the prodrugs. Moreover, hamster cells lacking XPF acquired remarkably more chromosomal aberrations after incubation for two cell cycle times with CNDAG 6-NH2, compared to the wild type. Furthermore, CNDAG 6-NH2 induced greater levels of sister chromatid exchanges in wild-type cells exposed for two cycles than those for one cycle, consistent with increased double-strand breaks after a second S phase. CONCLUSION: CNDAG-induced double-strand breaks are repaired mainly through homologous recombination.


Assuntos
Citarabina/análogos & derivados , Dano ao DNA/efeitos dos fármacos , Reparo do DNA/efeitos dos fármacos , Recombinação Homóloga/efeitos dos fármacos , Leucemia de Células T/tratamento farmacológico , Sobrevivência Celular , Citarabina/química , Citarabina/farmacologia , Humanos , Leucemia de Células T/patologia , Fosforilação , Células Tumorais Cultivadas
8.
Oncol Rep ; 42(6): 2806-2813, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31578589

RESUMO

Gastric cancer is an aggressive disease and a common cause of cancer­associated mortality worldwide. Recent studies have indicated that follistatin­like protein 1 (FSTL­1) is expressed and serves essential roles in tumorigenesis; however, the specific functional mechanism of FSTL­1 in gastric cancer progression remains ambiguous. CellTiter­Glo Luminescent Cell Viability and lactate dehydrogenase assays were used to measure cell survival and cell cytotoxicity, respectively. Cell apoptosis was ascertained using the Cell Death Detection ELISA assay and caspase­3/9 activity kits. Reverse transcription­quantitative polymerase chain reaction and western blotting were used to detect the expression levels of FSTL­1. The present study confirmed that FSTL­1 was highly expressed in gastric cancer cells compared with in control cells. Subsequently, FSTL­1 inhibition by small interfering RNA significantly reduced cancer cell survival and induced cytotoxic effects. In addition, knockdown of FSTL­1 in gastric cancer cells promoted apoptosis by increasing caspase­3 and caspase­9 expression. A decrease in signal transducer and activator of transcription 6 (STAT6) phosphorylation was observed in FSTL­1 knockdown cells, and the results confirmed that STAT6 phosphorylation was essential for FSTL­1 knockdown­induced cell apoptosis of cancer cells. Taken together, these results demonstrated that FSTL­1 knockdown may promote cell apoptosis via the STAT6 signaling pathway; therefore, FSTL1 may be considered a novel diagnostic and therapeutic target for gastric cancer.


Assuntos
Apoptose/genética , Proteínas Relacionadas à Folistatina/genética , Fator de Transcrição STAT6/genética , Neoplasias Gástricas/genética , Carcinogênese/genética , Caspase 3/genética , Caspase 9/genética , Linhagem Celular Tumoral , Proliferação de Células/genética , Sobrevivência Celular/genética , Ensaio de Imunoadsorção Enzimática , Proteínas Relacionadas à Folistatina/antagonistas & inibidores , Regulação Neoplásica da Expressão Gênica/genética , Técnicas de Silenciamento de Genes , Humanos , RNA Interferente Pequeno/genética , Transdução de Sinais/genética , Neoplasias Gástricas/patologia
9.
Mol Cancer Ther ; 18(12): 2283-2295, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31501277

RESUMO

Incorporation of the clinically active deoxycytidine analogue 2'-C-cyano-2'-deoxy-1-ß-D-arabino-pentofuranosyl-cytosine (CNDAC) into DNA generates single-strand breaks that are subsequently converted to double-strand breaks (DSB). Here, we investigated the cellular manifestations of these breaks that link these mechanisms to cell death, and we further tested the relevance of DNA repair pathways in protection of cells against CNDAC damage. The present investigations demonstrate that following exposure to CNDAC and a wash into drug-free medium, chromosomal aberrations, DNA strand breaks, and multinucleate cells arose. These portended loss of viability and were dependent upon exposure time, CNDAC concentration, and passage through mitosis. Following a pulse incubation with CNDAC, live cell imaging using GFP-tagged histone H2B as a marker demonstrated a normal rate of progression to mitosis, but a concentration-dependent delay in passage to a second mitosis. Progression through mitosis was also delayed and accompanied by formation of multinucleate cells. CNDAC-treated cells lacking XPF-ERCC1 nuclease function showed a 16-fold increase in chromosome aberrations. Chromosomal damage in Rad51D-mutant cells (homologous recombination repair deficient) were even more severely affected with extensive aberrations. Rodent or human Polq (POLQ) mutant cells, defective in Pol θ-mediated alternative end joining, did not show enhanced cellular sensitivity to CNDAC. These findings are consistent with formation of DSBs in the second S-phase following exposure, resulting in chromosome aberrations, aberrant mitoses, and subsequent apoptosis.


Assuntos
Morte Celular/genética , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , Mitose/genética , Humanos
10.
Cancer Chemother Pharmacol ; 81(2): 255-267, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29189915

RESUMO

PURPOSE: The mechanism of action of CNDAC (2'-C-cyano-2'-deoxy-1-ß-D-arabino-pentofuranosyl-cytosine) is unique among deoxycytidine analogs because upon incorporation into DNA it causes a single strand break which is converted to a double strand break after DNA replication. This lesion requires homologous recombination (HR) for repair. CNDAC, as the parent nucleoside, DFP10917, and as an oral prodrug, sapacitabine, are undergoing clinical trials for hematological malignancies and solid tumors. The purpose of this study is to investigate the potential of CNDAC for the therapy of ovarian cancer (OC). METHODS: Drug sensitivity was evaluated using a clonogenic survival assay. Drug combination effects were quantified by median effect analysis. RESULTS: OC cells lacking function of the key HR genes, BRCA1 or BRCA2, were more sensitive to CNDAC than corresponding HR proficient cells. The sensitization was associated with greater levels of DNA damage in response to CNDAC at clinically achievable concentrations, manifested as chromosomal aberrations. Three classes of CNDAC-based drug combinations were investigated. First, the PARP1 inhibitors, rucaparib and talazoparib, were selectively synergistic with CNDAC in BRCA1/2 deficient OC cells (combination index < 1) at a relatively low concentration range. Second, cisplatin and oxaliplatin had additive combination effects with CNDAC (combination index ~ 1). Finally, paclitaxel and docetaxel achieved additive cell-killing effects with CNDAC at concentration ranges of the taxanes similar for both BRCA1/2 deficient and proficient OC cells. CONCLUSIONS: This study provides mechanistic rationales for combining CNDAC with PARP inhibitors, platinum compounds and taxanes in ovarian cancer lacking BRCA1/2 function.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Proteína BRCA1/genética , Proteína BRCA2/genética , Citarabina/análogos & derivados , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/genética , Antineoplásicos/uso terapêutico , Proteína BRCA1/efeitos dos fármacos , Proteína BRCA2/efeitos dos fármacos , Linhagem Celular Tumoral , Aberrações Cromossômicas/efeitos dos fármacos , Citarabina/uso terapêutico , Dano ao DNA , Sinergismo Farmacológico , Feminino , Humanos , Compostos Organoplatínicos/uso terapêutico , Poli(ADP-Ribose) Polimerase-1 , Ensaio Tumoral de Célula-Tronco
11.
Mol Cancer Ther ; 15(10): 2302-2313, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27474148

RESUMO

CNDAC (2'-C-cyano-2'-deoxy-1-ß-d-arabino-pentofuranosyl-cytosine, DFP10917) and its orally bioavailable prodrug, sapacitabine, are undergoing clinical trials for hematologic malignancies and solid tumors. The unique action mechanism of inducing DNA strand breaks distinguishes CNDAC from other deoxycytidine analogs. To optimize the clinical potentials of CNDAC, we explored multiple strategies combining CNDAC with chemotherapeutic agents targeting distinct DNA damage repair pathways that are currently in clinical use. The ability of each agent to decrease proliferative potential, determined by clonogenic assays, was determined in paired cell lines proficient and deficient in certain DNA repair proteins. Subsequently, each agent was used in combination with CNDAC at fixed concentration ratios. The clonogenicity was quantitated by median effect analysis, and a combination index was calculated. The c-Abl kinase inhibitor imatinib had synergy with CNDAC in HCT116 cells, regardless of p53 status. Inhibitors of PARP1 that interfere with homologous recombination (HR) repair or base excision repair (BER) and agents such as temozolomide that cause DNA damage repaired by the BER pathway were also synergistic with CNDAC. The toxicity of the nitrogen mustards bendamustine and cytoxan, or of platinum compounds, which generate DNA adducts repaired by nucleotide excision repair and HR, was additive with CNDAC. An additive cell killing was also achieved by the combination of CNDAC with taxane mitotic inhibitors (paclitaxel and docetaxel). At concentrations that allow survival of the majority of wild-type cells, the synergistic or additive combination effects were selective in HR-deficient cells. This study provides mechanistic rationales for combining CNDAC with other active drugs. Mol Cancer Ther; 15(10); 2302-13. ©2016 AACR.


Assuntos
Antineoplásicos/farmacologia , Citarabina/análogos & derivados , Quebras de DNA/efeitos dos fármacos , Animais , Linhagem Celular Tumoral , Cricetinae , Citarabina/farmacologia , Dacarbazina/análogos & derivados , Dacarbazina/farmacologia , Relação Dose-Resposta a Droga , Interações Medicamentosas , Sinergismo Farmacológico , Recombinação Homóloga/efeitos dos fármacos , Recombinação Homóloga/genética , Humanos , Platina/farmacologia , Poli(ADP-Ribose) Polimerase-1/antagonistas & inibidores , Temozolomida , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
12.
Zhonghua Wei Chang Wai Ke Za Zhi ; 14(4): 257-60, 2011 Apr.
Artigo em Chinês | MEDLINE | ID: mdl-21538266

RESUMO

OBJECTIVE: To evaluate the value of modified acute physiologic and chronic health score (APACHE II score) in predicting postoperative complications in patients with acute obstructing colorectal carcinoma. METHODS: Postoperative complications in 92 patients with acute obstructing colorectal carcinoma were evaluated by APACHE II score and modified APACHE II score (severe organ dysfunction and immune damage in chronic health indicators were replaced by the duration and degree of obstruction, which were considered as the severity of intestinal obstruction). The sensitivity, specificity, and Youden index were compared with regard to complication prediction. Receiver operating characteristic curves were plotted to calculate area under the curve(AUC). RESULTS: Twenty-five patients developed postoperative complications including 3 deaths. The APACHE-II score(13.72±4.24), modified APACHE II score (19.28±4.92), intestinal obstruction severity score (5.56±2.20) were significantly higher in patients with complications than those in patients without complications (10.58±3.44, 14.69±3.73, 4.10±1.52, all P<0.01). The sensitivity, specificity, accuracy, Youden index, and AUC were 0.640, 0.940, 0.859, 0.580, and 0.839 for the modified APACHE-II score with 20 being the optimal cut-off point, respectively, and were 0.560, 0.896, 0.804, 0.456, and 0.784 for APACHE-II (14 was the optimal cut-off point), respectively. CONCLUSION: The modified APACHE-II score system with the intestinal obstruction severity score is a better prediction method for the occurrence of postoperative complications in patients with acute obstructing colorectal carcinoma.


Assuntos
Neoplasias Colorretais/cirurgia , Obstrução Intestinal/cirurgia , Complicações Pós-Operatórias , APACHE , Adulto , Idoso , Idoso de 80 Anos ou mais , Área Sob a Curva , Neoplasias Colorretais/complicações , Feminino , Humanos , Obstrução Intestinal/etiologia , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos , Sensibilidade e Especificidade
13.
Mol Pharmacol ; 80(2): 321-7, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21546556

RESUMO

4-Hydroxy-5-methoxy-2,3-dihydro-1H-[1,3]benzodioxolo[5,6-c]pyrrolo[1,2-f]-phenanthridium chloride (NK314) is a benzo[c] phenanthridine alkaloid that inhibits topoisomerase IIα, leading to the generation of DNA double-strand breaks (DSBs) and activating the G(2) checkpoint pathway. The purpose of the present studies was to investigate the DNA intercalating properties of NK314, to evaluate the DNA repair mechanisms activated in cells that may lead to resistance to NK314, and to develop mechanism-based combination strategies to maximize the antitumor effect of the compound. A DNA unwinding assay indicated that NK314 intercalates in DNA, a property that likely cooperates with its ability to trap topoisomerase IIα in its cleavage complex form. The consequence of this is the formation of DNA DSBs, as demonstrated by pulsed-field gel electrophoresis and H2AX phosphorylation. Clonogenic assays demonstrated a significant sensitization in NK314-treated cells deficient in DNA-dependent protein kinase (DNA-PK) catalytic subunit, Ku80, ataxia telangiectasia mutated (ATM), BRCA2, or XRCC3 compared with wild-type cells, indicating that both nonhomologous end-joining and homologous recombination DNA repair pathways contribute to cell survival. Furthermore, both the DNA-PK inhibitor 8-(4-dibenzothienyl)-2-(4-morpholinyl)-4H-1-benzopyran-4-one (NU7441) and the ATM inhibitor 2-(4-morpholinyl)-6-(1-thianthrenyl)-4H-pyran-4-one (KU55933) significantly sensitized cells to NK314. We conclude that DNA-PK and ATM contribute to cell survival in response to NK314 and could be potential targets for abrogating resistance and maximizing the antitumor effect of NK314.


Assuntos
Antígenos de Neoplasias/metabolismo , Ataxia Telangiectasia/enzimologia , Ataxia Telangiectasia/patologia , Proteínas de Ciclo Celular/fisiologia , DNA Topoisomerases Tipo II/metabolismo , Proteína Quinase Ativada por DNA/fisiologia , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/fisiologia , Fenantrenos/farmacologia , Proteínas Serina-Treonina Quinases/fisiologia , Inibidores da Topoisomerase/farmacologia , Proteínas Supressoras de Tumor/fisiologia , Antineoplásicos/farmacologia , Ataxia Telangiectasia/tratamento farmacológico , Proteínas Mutadas de Ataxia Telangiectasia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Humanos
14.
Blood ; 117(1): 156-64, 2011 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-20971952

RESUMO

Homoharringtonine (HHT) is a plant alkaloid that inhibits the elongation phase of translation that is currently in clinical trials. Because the intrinsically short-lived antiapoptotic protein myeloid cell leukemia-1 (Mcl-1) has been reported to support the survival of chronic lymphocytic leukemia (CLL) cells, we hypothesized that inhibition of protein synthesis by HHT would decrease Mcl-1 expression and induce apoptosis in CLL. In primary CLL cells, HHT induced significant apoptosis independent of the prognostic characteristics of the patients. This was associated with inhibition of translation and decreased Mcl-1 levels in CLL cells. Mcl-1 reduction was evident as early as 2 hours and continued to decrease in the next 6-8 hours, whereas cell death started in 2 hours and continued to increase for 24 hours. Reduction of the Mcl-1 level was due to translation inhibition and proteasome degradation rather than to transcription inhibition or caspase cleavage. HHT and the transcription inhibitor SNS-032 induced synergistic cell killing. Although stromal cells induced Mcl-1 expression and protected CLL cells from the toxicity of fludarabine, this induction was reversed by HHT, which overcame stromal cell-mediated protection. Thus, these results provide a rationale for clinical development of HHT in CLL as single agent or in combinations.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Apoptose/efeitos dos fármacos , Harringtoninas/farmacologia , Leucemia Linfocítica Crônica de Células B/patologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Western Blotting , Proliferação de Células/efeitos dos fármacos , Sinergismo Farmacológico , Quimioterapia Combinada , Mepesuccinato de Omacetaxina , Humanos , Leucemia Linfocítica Crônica de Células B/tratamento farmacológico , Leucemia Linfocítica Crônica de Células B/metabolismo , Linfócitos/efeitos dos fármacos , Pessoa de Meia-Idade , Membranas Mitocondriais/efeitos dos fármacos , Proteína de Sequência 1 de Leucemia de Células Mieloides , Oxazóis/farmacologia , Proteínas Proto-Oncogênicas c-bcl-2/genética , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Estromais/efeitos dos fármacos , Tiazóis/farmacologia , Células Tumorais Cultivadas
16.
Cancer Res ; 65(15): 6874-81, 2005 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-16061671

RESUMO

2'-C-cyano-2'-deoxy-1-beta-D-arabino-pentofuranosylcytosine (CNDAC) is a nucleoside analogue with a novel mechanism of action that is currently being evaluated in clinical trials. Incorporation of CNDAC triphosphate into DNA and extension during replication leads to single-strand breaks directly caused by beta-elimination. These breaks, or the lesions that arise from further processing, cause cells to arrest in G2. The purpose of this investigation was to define the molecular basis for G2 checkpoint activation and to delineate the sequelae of its abrogation. Cell lines derived from diverse human tissues underwent G2 arrest after CNDAC treatment, suggesting a common mechanism of response to the damage created. CNDAC-induced G2 arrest was instituted by activation of the Chk1-Cdc25C-Cdk1/cyclin B checkpoint pathway. Neither Chk2, p38, nor p53 was required for checkpoint activation. Inhibition of Chk1 kinase with 7-hydroxystaurosporine (UCN-01) abrogated the checkpoint pathway as indicated by dephosphorylation of checkpoint proteins and progression of cells through mitosis and into G1. Cell death was first evident in hematologic cell lines after G1 entry. As indicated by histone H2AX phosphorylation, DNA damage initiated by CNDAC incorporation was transformed into double-strand breaks when ML-1 cells arrested in G2. Some breaks were manifested as chromosomal aberrations when the G2 checkpoint of CNDAC-arrested cells was abrogated by UCN-01 but also in a minor population of cells that escaped to mitosis during treatment with CNDAC alone. These findings provide a mechanistic rationale for the design of new strategies, combining CNDAC with inhibitors of cell cycle checkpoint regulation in the therapy of hematologic malignancies.


Assuntos
Citarabina/análogos & derivados , Fase G2/efeitos dos fármacos , Neoplasias Hematológicas/tratamento farmacológico , Neoplasias Hematológicas/patologia , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Proteína Quinase CDC2/metabolismo , Linhagem Celular Tumoral , Quinase 1 do Ponto de Checagem , Aberrações Cromossômicas , Citarabina/antagonistas & inibidores , Citarabina/farmacologia , Dano ao DNA , Interações Medicamentosas , Fase G2/genética , Neoplasias Hematológicas/genética , Humanos , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patologia , Mitose/efeitos dos fármacos , Proteínas Quinases/metabolismo , Transdução de Sinais/efeitos dos fármacos , Estaurosporina/análogos & derivados , Estaurosporina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA