Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Oncol Rep ; 22(1): 161-70, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19513519

RESUMEN

beta-Elemene, a new plant-derived anticancer agent with low toxicity, has been reported to be effective in the treatment of leukemia and solid tumors. In the current study, we explored the therapeutic application of beta-elemene in sensitizing lung cancer cells to cisplatin. beta-Elemene considerably enhanced the inhibitory effect of cisplatin on cell proliferation in a time- and dose-dependent manner in the human non-small cell lung cancer (NSCLC) cell lines H460 and A549. Furthermore, this effect of beta-elemene on cisplatin activity occurred through the induction of apoptosis in NSCLC cells, as assessed by an ELISA-based assay, TUNEL assay and annexin V binding assay. Consistent with these results, the protein levels of Bax and phospho-Bcl-2 increased and those of Bcl-2 and XIAP decreased in cells treated with beta-elemene in combination with cisplatin, compared with the levels in cells treated with either agent alone. Finally, beta-elemene augmented the cisplatin-induced increases in caspase-3, -7, -9 and -10 activities and cleaved caspase-3, -9 and poly(ADP-ribose) polymerase levels in NSCLC cells. These observations suggest that beta-elemene sensitizes NSCLC cells to cisplatin via a mitochondria-mediated intrinsic apoptosis pathway involving Bcl-2 family proteins and IAPs (inhibitor of apoptosis proteins). Our data provide a rationale for developing a combination of beta-elemene and cisplatin as a regimen for the treatment of lung carcinoma and other cisplatin-resistant tumors.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Apoptosis/efectos de los fármacos , Carcinoma de Pulmón de Células no Pequeñas/patología , Neoplasias Pulmonares/patología , Antineoplásicos Fitogénicos/administración & dosificación , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Caspasas/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cisplatino/administración & dosificación , Relación Dosis-Respuesta a Droga , Activación Enzimática , Humanos , Neoplasias Pulmonares/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Poli(ADP-Ribosa) Polimerasas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Sesquiterpenos/administración & dosificación , Factores de Tiempo , Proteína Inhibidora de la Apoptosis Ligada a X/metabolismo , Proteína X Asociada a bcl-2/metabolismo
2.
Int J Mol Med ; 23(3): 357-62, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19212654

RESUMEN

Mammalian sterile 20-like kinase 1 (MST1) is a serine/threonine protein kinase that is activated in response to a variety of apoptotic stimuli and causes apoptosis when over-expressed in mammalian cells. The physiological regulation and cellular targets of MST1 are not well understood. Using a yeast two-hybrid system, we identified human WW45 (hWW45, also called hSav1) as an MST1-binding protein. The association between the two proteins was confirmed by immunofluorescence and co-immunoprecipitation, and hWW45 was present in both the cytoplasm and nucleus. When hWW45 alone was over-expressed, it weakly induced apoptosis. However, hWW45 augmented MST1-induced apoptosis when the two were co-expressed. Conversely, RNA interference-mediated depletion of endogenous hWW45 suppressed MST1-induced apoptosis. These results indicate that hWW45 is required to enhance MST1-mediated apoptosis in vivo and thus is a critical player in an MST1-driven cell death signaling pathway.


Asunto(s)
Apoptosis/fisiología , Proteínas de Ciclo Celular/biosíntesis , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal/fisiología , Animales , Proteínas de Ciclo Celular/genética , Núcleo Celular/genética , Citoplasma/genética , Células HeLa , Humanos , Péptidos y Proteínas de Señalización Intracelular , Ratones , Células 3T3 NIH , Proteínas Serina-Treonina Quinasas/genética , Interferencia de ARN , Técnicas del Sistema de Dos Híbridos
3.
Int J Oncol ; 31(2): 241-52, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17611679

RESUMEN

Beta-elemene has recently raised interest in P.R. China as a novel antitumor plant drug isolated from the Chinese medicinal herb Zedoary. To explore potentially useful combinations of beta-elemene with taxanes in the clinic, we characterized the effects of beta-elemene combined with taxanes in human lung cancer cells using a median effect analysis, micronucleus assay, apoptotic detection, and determination of gene expression in the signaling pathways of apoptosis. The synergistic analysis indicated that the interactions of beta-elemene with paclitaxel or docetaxel ranged from slight synergism to synergism. Combinations of beta-elemene with docetaxel induced much stronger synergistic interactions in p53 mutant H23 cells and p53 null H358 cells than in p53 wild-type H460 and A549 cells. Similar synergistic interactions were observed by micronucleus assay, apoptotic detection, and determination of apoptotic gene expression. Our findings indicate that the synergistic effects achieved with combinations of beta-elemene and taxanes are related to the augmented cytotoxic efficacy of taxanes owing to the action of beta-elemene. In H460 and A549 cells, dose-dependent upregulation of p53 protein expression was observed in cultures treated with docetaxel alone and with docetaxel plus beta-elemene, whereas no significant change in p53 expression was observed in any of the treatment groups in H23 cells. Fas revealed no alteration of expression with any of the treatments in this study. However, the combination treatments induced increased cytochrome c release from mitochondria, significant caspase-8 and -3 cleavage, and downregulation of Bcl-2 and Bcl-XL expression. These results suggest that, although p53 plays an important role in taxane-induced cell death, apoptosis induced by beta-elemene or in combination with docetaxel thereof seems to be initiated through a p53- and Fas-independent pathway via mitochondria in our lung cancer cells. The suppression of specific 'survival' gene expression appears to be the key action leading to the synergistic effect of combination treatments with beta-elemene and taxanes. Finally, the beta-elemene-induced alteration of cell membrane permeability, which has potential to result in enhanced cellular uptake of taxanes, may also contribute to the synergistic interactions of the combination treatments.


Asunto(s)
Antineoplásicos/administración & dosificación , Antineoplásicos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Carcinoma/tratamiento farmacológico , Ensayos de Selección de Medicamentos Antitumorales , Regulación Neoplásica de la Expresión Génica , Neoplasias Pulmonares/tratamiento farmacológico , Sesquiterpenos/administración & dosificación , Taxoides/administración & dosificación , Antineoplásicos/química , Línea Celular Tumoral , Genes p53 , Humanos , Técnicas In Vitro , Concentración 50 Inhibidora , Medicina Tradicional China , Mutación , Sales de Tetrazolio/farmacología , Tiazoles/farmacología
4.
Cell Oncol ; 29(4): 335-49, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17641417

RESUMEN

The E2F family members play a critical role in cell cycle regulation and other biological processes in the cell. To better understand the involvement of E2F-1 in the development and progression of gastric tumors, we investigated the mutation and expression of E2F-1 in human gastric cancer tissues and the effect of E2F-1 overexpression on the proliferation of gastric carcinoma cells. In this study, 80 pairs of gastric cancer specimens and paratumor tissues from different patients and 40 stomach mucosa specimens from healthy individuals were examined. PCR-SSCP analysis demonstrated that mutations were not detected in any of the gastric cancer and normal tissue specimens. In addition, the results of an immunohistochemistry assay revealed higher expression rates of E2F-1 (P<0.01) in gastric cancer tissues (72.5%) than in paratumor tissues (30.0%) of the same individuals and stomach mucosa from healthy individuals (22.5%). However, no correlation was observed between the E2F-1 levels and patients' clinical features, such as sex, age, histological types, lymph node metastasis, and clinical stages (P>0.05). Finally, the influence of E2F-1 overexpression on the growth of human gastric carcinoma MKN-45 cells in vitro was assessed by measuring colony formation, cell survival, and cell cycle progression. Our data clearly showed that cell growth and proliferation were significantly inhibited in MKN-45 tumor cells transfected with the expression vector encoding E2F-1 in comparison with nontransfected cells or cells transfected with empty vector. These findings suggest that E2F-1, a stable and conservative gene during the oncogenesis and progression of stomach cancers, may potentially serve as a biomarker for clinical diagnosis of gastric carcinomas and as a target for the development of novel therapeutic interventions to treat this disease.


Asunto(s)
Factor de Transcripción E2F1/genética , Regulación Neoplásica de la Expresión Génica , Expresión Génica , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Regulación hacia Arriba/genética , Adulto , Anciano , Animales , Células COS , Ciclo Celular , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Chlorocebus aethiops , Femenino , Mucosa Gástrica/metabolismo , Vectores Genéticos , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Mutación/genética , Polimorfismo Conformacional Retorcido-Simple , Transfección
5.
Oncol Rep ; 17(6): 1437-44, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17487402

RESUMEN

Apoptosis results in cell death within 10 min after initiation by Bcl-2 family proteins and mitochondria; however, cells enter the apoptotic pathway at different elapsed times after being triggered. Intrinsic factors related to chemical or physical cell damage can initiate apoptosis at a specific cell cycle phase; it is not clear whether cells insulted via an extrinsic pathway also die at a specific cell cycle phase, or how apoptosis is related to cell cycle progression in cells. To illustrate the kinetic changes of apoptosis during cell cycle progression, we examined both intrinsically and extrinsically induced apoptosis in MOLT-4 and Jurkat lymphocytic leukemia cells and in cultured peripheral blood lymphocytes (PBLs) using a recently modified annexin V and propidium iodide method, which detects cell cycle-specific apoptosis. Apoptosis predominantly occurred at a specific cell cycle phase. Leukemia cells were sensitive to induction by both intrinsic (X-rays, UV light, camptothecin, arsenic trioxide, and the traditional Chinese medicine Jinke, which is an extract of Auricularia auricula) and extrinsic factors (via Fas and TNF receptor pathways). The phase at which leukemia cells entered apoptosis depended on the nature of the insult (X-ray or UV, G1-phase; camptothecin, S-phase; arsenic, G1/S phases; Jinke, G1/S phases; and TNF or Fas ligand, G1/S phases), whereas PBLs did not exhibit such insult-dependent differences. PHA-stimulated PBLs entered apoptosis, and additional cells were recruited following additional insults. Unstimulated PBLs remained unresponsive to apoptosis, and proliferating cells became insensitive to the insults after the cell cycle checkpoint was abolished by caffeine. Confluent or starving PBLs were also unresponsive to apoptotic triggers. Thus, apoptotic cell death is a cell cycle event with most, if not all, apoptosis being initiated during a particular cell cycle phase, and changes in the cell cycle result in changes in the apoptotic pattern and schedule. The coordination of apoptosis and proliferation in cells offers a mechanism for the integration of both cell cycle and apoptotic signals.


Asunto(s)
Apoptosis , Ciclo Celular , Leucemia Linfoide , Linfocitos/fisiología , Proliferación Celular , Humanos , Células Jurkat , Linfocitos/efectos de los fármacos , Linfocitos/efectos de la radiación , Factores de Tiempo , Células Tumorales Cultivadas
6.
Int J Oncol ; 28(5): 1225-32, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16596239

RESUMEN

Telomerase is implicated in the development of cellular immortality and oncogenesis. It has been shown that telomerase activity is considerably higher in the tissue of many different cancers than in normal tissue, and that the inhibition or downregulation of telomerase activity can prevent the malignant proliferation of tumor cells. Antisense oligonucleotides have been widely used in suppressing the expression of genes and, therefore, in the present research, we evaluated the effect of antisense human telomerase RNA (hTR) on glioma cell growth in vitro and in vivo. We showed that antisense hTR cDNA significantly inhibited TJ905 human glioma cell proliferation in vitro and tumor growth in vivo, as determined by MTT assay and by measuring the volume of glioma in nude mice. Consistent with these results, we found that telomerase activity and the mRNA levels of hTR and hTERT (human telomerase reverse transcriptase) expression were markedly decreased in tumor cells treated with antisense hTR cDNA, as assessed by TRAP (telomeric repeat amplification protocol) assay and RT-PCR (reverse transcription-polymerase chain reaction) analysis. Our study conclusively demonstrates that antisense hTR effectively inhibits the growth of human glioma cells in vitro and in vivo and, thus, may be potentially used for gene therapy of malignant gliomas and other cancers.


Asunto(s)
Neoplasias Encefálicas/patología , Glioma/patología , ARN sin Sentido/farmacología , ARN sin Sentido/uso terapéutico , Telomerasa/genética , Actinas/genética , Secuencia de Bases , División Celular/efectos de los fármacos , Línea Celular Tumoral , Cartilla de ADN , Humanos , Cinética , ARN Mensajero/genética
7.
Int J Mol Med ; 17(2): 309-17, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16391831

RESUMEN

The emergence of antibiotic-resistant bacterial strains still remains a significant problem for antimicrobial chemotherapy in the clinic. Bacterial viruses (bacteriophages or phages) have been suggested to be used as alternative therapeutic agents for bacterial infections. However, the efficacy of phage therapy in treating drug-resistant infections in humans is uncertain. Therefore in the present study, we examined the effectiveness of phages in the treatment of imipenem-resistant Pseudomonas aeruginosa (IMPR-Pa) infection in an experimental mouse model. Twenty-nine strains of phage were isolated from our hospital sewage, and phage ØA392 was representatively used for all testing because it has lytic activity against a wide range of clinical isolates of IMPR-Pa. We found that intraperitoneal (i.p.) injections of one IMPR-Pa strain (3 x 10(7) CFU) caused bacteremia and all mice died within 24 h. A single i.p. inoculation of the phage strain (MOI > or =0.01) at up to 60 min after the bacterial challenge was sufficient to rescue 100% of the animals. This lifesaving effect coincided with the rapid appearance of ØA392 in the circulation (within 2 h after i.p. injection), which remained at substantially higher levels for up to 48 h until the bacteria were eradicated. However, the survival rates of the mice dropped to approximately 50% and 20% when the same dose of this purified phage preparation was administered at 180 min and 360 min, respectively, after IMPR-Pa infections. In addition, we demonstrated that the ability of this phage to rescue bacteremic animals was due to the functional capabilities of the phage and not to a non-specific immune effect. The protection from death occurred only in animals inoculated with bacteria-specific virulent phage strains. When the heat-inactivated phages were used, the survival rate of the infected mice was dramatically reduced to 20% or lower. Moreover, the levels of the antibody against the phage were not significantly changed at the time when the bacteremic animals were protected by the active phages. Finally, our observations revealed that the inoculation of the mice with high-doses of ØA392 alone produced no adverse effects attributable to the phage. These data indicate that phages can save animals from pernicious P. aeruginosa infections and suggest that phage therapy may be potentially used as a stand-alone therapy for patients with IMPR-Pa infections.


Asunto(s)
Bacteriemia/genética , Bacteriemia/terapia , Bacteriófagos/genética , Terapia Genética/métodos , Imipenem/farmacología , Pseudomonas aeruginosa/efectos de los fármacos , Pseudomonas aeruginosa/virología , Animales , Bacteriemia/microbiología , Bacteriófagos/aislamiento & purificación , Bacteriófagos/fisiología , Bacteriófagos/ultraestructura , Modelos Animales de Enfermedad , Farmacorresistencia Bacteriana , Femenino , Ratones , Ratones Endogámicos BALB C , Microscopía Electrónica , Pseudomonas aeruginosa/fisiología , Temperatura
8.
Int J Mol Med ; 17(2): 347-55, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16391836

RESUMEN

The emergence of multidrug-resistant bacteria has become a global crisis. Accumulating evidence shows that bacteriophages (phages) can rescue animals from a variety of lethal infections and be effective in treating drug-resistant infections in humans. Enterobacteriaceae, producing extended spectrum beta-lactamase enzymes (ESBLs), are resistant to a broad range of beta-lactamase antibiotics. One of the most common ESBL-producing gram-negative bacilli in Enterobacteriaceae is Escherichia coli. Since ESBL-producing E. coli poses a formidable challenge in the management of critically ill patients with bacterial infections, we undertook this study to explore the possible therapeutic utility of phages to control ESBL-producing E. coli infections. The phage Ø9882 used in this study was isolated from our hospital sewage and has lytic activity against a broad range of clinical isolates of ESBL-producing E. coli. ESBL-producing E. coli strains (n=30) were isolated in the clinic, and one of them was used to induce bacteremia in a murine model. Bacteremia was established by intraperitoneal (i.p.) injection of 3 x 10(7) CFU/ml, the minimum lethal dose (MLD) of bacterium in this animal model. Mice infected with the MLD of this strain alone died within 14 h, whereas a single i.p. inoculation of Ø9882 (MOI > or =10(-4)) given 40 min after the bacterial challenge led to 100% survival at 24-168 h, compared to 0% survival of saline-treated controls. Protection was obtained even when administration of the phage was delayed up to 60 min after the bacterial infection and the survival rate of infected animals was 60% at 168 h. Furthermore, it was shown that the therapeutic efficacy of Ø9882 in lethal systemic infection in our model is due to the functional capability of the phage and not the nonspecific immune effects. Our data both in vitro and in vivo revealed that: i) the protection of mice from death occurred only in animals infected with selected bacterial strains and the virulent phage specific to them; ii) when the phages were heat-inactivated, survival of the infected mice was strikingly decreased to 0; and iii) the level of antibody against the phage was not substantially elevated when the bacteremic animals were protected by the phage. The present findings indicate that phages can effectively rescue our mouse model from bacteremia and death, and thus provide the rationale and framework to evaluate the therapeutical efficacy of lytic phages against fatal ESBL-producing E. coli infections in humans.


Asunto(s)
Bacteriemia/microbiología , Bacteriemia/terapia , Bacteriófagos/genética , Escherichia coli/enzimología , Escherichia coli/genética , Terapia Genética/métodos , beta-Lactamasas/biosíntesis , Animales , Bacteriemia/genética , Bacteriemia/metabolismo , Bacteriófagos/aislamiento & purificación , Bacteriófagos/ultraestructura , Modelos Animales de Enfermedad , Femenino , Calor , Ratones , Ratones Endogámicos BALB C , Microscopía Electrónica
9.
Int J Mol Med ; 17(5): 911-6, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16596280

RESUMEN

Graves' ophthalmopathy (GO) is considered to be an organ-specific autoimmune disease. However, the pathogenesis of GO is incompletely understood at the present time. To clarify the immunological differences between newly diagnosed GO and Graves' disease (GD) without ophthalmopathy or healthy controls (HC), we examined T-cell profile and the Th1/Th2 profile cell balance in GO (n=20), GD (n=20) and HC (n=20) using flow cytometry. We also assessed the influence of methimazole on the immunocyte profiles in patients with GO and GD and analyzed the relationship of the immunologic changes with CAS, FT3, FT4, TRAb, TMA and TGA among the three investigated groups. We report in this study that: 1) The percentage of CD4+ T cells and the ratio of CD4+/CD8+ cells were higher, but the population of CD8+ T cells was lower in both GO and GD than those of HC (P<0.05); 2) The percentage of CD8-/IFNgamma+ T cells (Th1) and the ratio of CD8-/IFNgamma+ to CD8-/IL-4+ T cells (Th1/Th2) in GO were considerably higher as compared to those in GD and HC (P<0.05). On the contrary, the population of Th1 cells, as well as the ratio of Th1/Th2 cells, was lower in GD than that of GO and HC (P<0.05); 3) There were no significant differences in T-cell profile and the Th1/Th2 cell balance in either GO or GD patients before and after methimazole treatment; 4) There was a positive correlation of Th1 cell percentage and the Th1/Th2 cell ratio with the clinical activity score (CAS) in GO (P<0.05), whereas CAS in GO had no correlation with the T-cell profile, the percentage of Th2 cells, and TRAb (P>0.05); 5) T-cell subset and the ratio of Th1/Th2 cells did not correlate significantly with FT3, FT4, TRAb, TMA, or TGA in GO and GD (P>0.05). Finally, 6) there were no statistical differences in TRAb, TMA, and TGA between early GO and GD without ophthalmopathy (P>0.05). Collectively, these results indicate that the balance of Th1/Th2 in GO shifts to Th1 dominance and that the cellular immune responses mediated by the Th1-type CD4+ cells might play a dominant role in the pathogenesis of GO, and thus suggest that the Th1 cell percentage and the ratio of Th1/Th2 cell subsets may be potentially utilized as clinical parameters for disease activity, for monitoring the effectiveness of immunosuppressive treatment, or for developing immunospecific forms of therapy for Graves' ophthalmopathy.


Asunto(s)
Linfocitos T CD4-Positivos/citología , Enfermedad de Graves/patología , Oftalmopatía de Graves/patología , Células TH1/citología , Células Th2/citología , Adolescente , Adulto , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/metabolismo , Células Cultivadas , Femenino , Citometría de Flujo , Enfermedad de Graves/inmunología , Enfermedad de Graves/metabolismo , Oftalmopatía de Graves/inmunología , Oftalmopatía de Graves/metabolismo , Humanos , Inmunohistoquímica , Interferón gamma/análisis , Interleucina-4/análisis , Recuento de Linfocitos , Masculino , Receptores de Tirotropina/análisis , Células TH1/metabolismo , Células Th2/metabolismo , Tirotropina/análisis , Tiroxina/análisis , Triyodotironina/análisis
10.
Int J Mol Med ; 18(5): 885-92, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17016618

RESUMEN

Transmembrane tumor necrosis factor-alpha (mTNF-alpha) is known to be the precursor of soluble TNF-alpha (sTNF-alpha). mTNF-alpha can act as a ligand on the TNF receptor- (TNFR)- bearing cell through 'forward signaling' or as a receptor on the TNF producing cell through 'reverse signaling'. In the current study, we investigated the role of mTNF-alpha-mediated reverse signaling in regulating sTNF-alpha-induced activation of human monocytic U937 cells. We demonstrated that pretreatment with sTNFRI, for inducing reverse signaling through mTNF-alpha, sensitized U937 cells to sTNF-alpha stimulation, as evidenced by an increase in reactive oxygen production and mRNA levels of proinflammatory cytokines (TNF-alpha, IL-1beta, and IL-8) in these cells. Further experiments revealed that IkappaB-alpha degradation was increased in the monocytic cells primed with sTNFRI, implying that reverse signaling of mTNF-alpha sensitizes U937 cells via an NF-kappaB-dependent mechanism. On the other hand, binding of sTNFRI to mTNF-alpha after sTNF-alpha-induced activation of U937 cells reduced mRNA stability (half-life) of IL-1beta and IL-8. The involvement of reverse signaling in the process was verified by using a mutated form of mTNF-alpha lacking the majority of the cytoplasmic domain. Our results clearly showed that enhanced mRNA degradation of the cytokines occurred only in U937 cells transfected with a wild-type mTNF-alpha, but not in those cells transfected with the mutant mTNF-alpha. Taken together, these data suggest that reverse signaling through mTNF-alpha may exert a double role in modulating sTNF-alpha bioactivity. It is positive when reverse signaling occurs prior to sTNF-alpha stimulation, while it is negative when reverse signaling occurs after the sTNF-alpha signal. Thus, our findings strengthen a role of mTNF-alpha-mediated reverse signaling in the regulation of immune-inflammatory response and control of inflammatory reaction.


Asunto(s)
Activación de Macrófagos , Monocitos/inmunología , Receptores del Factor de Necrosis Tumoral/fisiología , Factor de Necrosis Tumoral alfa/fisiología , Línea Celular Tumoral , Membrana Celular/metabolismo , Humanos , Proteínas I-kappa B/metabolismo , Interleucina-1/genética , Interleucina-1/metabolismo , Interleucina-8/genética , Interleucina-8/metabolismo , Monocitos/efectos de los fármacos , Inhibidor NF-kappaB alfa , ARN Mensajero/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/farmacología , Células U937
11.
Cancer Res ; 64(5): 1607-10, 2004 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-14996718

RESUMEN

Protein complex of cyclin B1 and cyclin-dependent protein kinase 1 induces phosphorylation of key substrates that mediate cell cycle transition during the G(2)-M phase. It is believed that cyclin B1 accumulates in the S phase of the cell cycle and reaches the maximal level at mitosis but is absent in G(1)-phase cells. In the present study, we demonstrated that cyclin B1 was expressed in the arrested G(1)-phase MOLT-4 lymphocyte leukemia cells and in G(1) phase T-7 transitional tumor cells, as determined by flow cytometry. In addition, we showed that cyclin B1 was detected in the G(1) phase in breast cancer cells from patient tissues and in lymphocytes from patients with leukemia. These findings were confirmed for the first time by postsorting Western blot analysis and by confocal microscopy. Furthermore, by using postsorting Western blotting, we found that cyclin B1 was expressed in different time-window sections of the G(1) phase under different conditions. For the asynchronously growing T-7 cells, cyclin B1 was detected in early G(1) phase, whereas in MOLT-4 cells arrested in G(1)-S phase, cyclin B1 was mainly detected in late G(1) phase. We propose that the cyclin B1 expressed in the G(1) phase may differ from that expressed in the G(2)-M phase, and that this unscheduled type of cyclin B1 may play an important role in tumorigenesis and apoptosis.


Asunto(s)
Ciclina B/análisis , Fase G1 , Neoplasias/química , Western Blotting , Línea Celular Tumoral , Ciclina B/fisiología , Ciclina B1 , Femenino , Humanos , Microscopía Confocal , Neoplasias/patología
12.
Int J Oncol ; 27(1): 247-56, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15942666

RESUMEN

Lung cancer is the most common cause of cancer mortality in both male and female patients in the United States of America, as well as in the rest of the world. Over one million people are diagnosed with lung cancer every year worldwide. The taxane is one of the most powerful classes of novel antitumor agents and has become an integral part of several commonly used chemotherapy regimens in lung cancer management over the past few years. Although the ability of taxanes to disrupt microtubule dynamics is well documented, the molecular basis by which taxanes suppress cancer cell growth and induce apoptotic cell death is not clearly defined. In this review, we focus on the molecular mechanisms of the antitumor activity of taxanes (paclitaxel and docetaxel) in lung cancer, and discuss the interactions of taxanes with microtubules, the roles of cell cycle control and cell death induction in the anticancer action of taxanes, as well as the signal transduction pathways involved in the processes. In addition, we discuss the possible mechanisms of taxane resistance, because drug resistance to these anti-neoplastic agents affects therapy efficacy and is also a major obstacle in the clinic for the successful treatment of lung cancer. Understanding the molecular mechanisms underlying the antitumor effect of taxanes and the drug resistance to taxanes may lead to the design of biologically and pharmacologically targeted therapeutic strategies for taxane resistant tumors, and to the improvement of chemotherapy effect and cancer patient survival.


Asunto(s)
Antineoplásicos/farmacología , Resistencia a Antineoplásicos , Neoplasias Pulmonares/tratamiento farmacológico , Taxoides/farmacología , Antineoplásicos Fitogénicos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica , Apoptosis , Ciclo Celular , Femenino , Humanos , Neoplasias Pulmonares/mortalidad , Masculino , Microtúbulos/metabolismo , Microtúbulos/ultraestructura , Transducción de Señal
13.
Int J Oncol ; 26(3): 703-11, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15703827

RESUMEN

Electrochemical treatment is among the most effective therapies in the management of cervical malignancy. However, the mechanism of action of this treatment remains largely unknown. Therefore, the purpose of the current project was to establish a suitable eletrochemotherapy regimen for cervical cancer and to investigate the mechanism of the therapy in an in vitro model for human cervical carcinoma. HeLa cells were used as a model for cervical cancer in this study, and the effect of electrochemical treatment on these cells was examined in four different dosage groups (5 V + 5 C, 10 V + 5 C, 5 V + 10 C and 10 V + 10 C). Our results showed that the combinations of lower voltage and higher current (5 V/10 V + 10 C) had a greater anticancer effect in this model as compared to other groups. In addition, we compared the cytotoxic effect between electrochemical treatment and different pH condition treatments in this system, and found that the efficacy of electrochemical treatment in cell killing was better than that of acidic or basic medium treatment. Moreover, we demonstrated that the efficacy of electrochemical treatment was correlated with the degree of ionization and alteration in pH scale. The electrodes were basic on the cathode side which elevated the cations K+, Ca2+ and Mg2+, while the electrodes were acidic on the anode side which reduced the anion Cl-. We also assessed the effect of electrochemical treatment on cell cycle distribution in HeLa cells and showed that the percentage of cells in the G1 phase of the cell cycle was increased (G1 arrest), while the cell population in the S phase was decreased. Furthermore, we demonstrated that the levels of the cell cycle regulator cyclin D1 expression were dramatically reduced when 5 V/10 V + 10 C treatments were applied to these cells, as determined by RT-PCR analysis. By contrast, no significant changes in the levels of cyclin B1, CDK1 or CDK4 were detected. Based on these observations, we conclude that the combination of lower voltage and higher current may be a potentially effective eletrochemotherapy regimen for cervical cancer in the clinic, and that the antitumor effect of electrochemical treatment on cervical carcinoma cells is mediated partly via regulating ionization degree, pH state and cell cycle control.


Asunto(s)
Carcinoma/patología , Carcinoma/terapia , Ciclo Celular/fisiología , Terapia por Estimulación Eléctrica , Neoplasias del Cuello Uterino/patología , Neoplasias del Cuello Uterino/terapia , Cationes , Muerte Celular , Ciclina D1/biosíntesis , Electroquímica , Electrodos , Femenino , Citometría de Flujo , Perfilación de la Expresión Génica , Células HeLa , Humanos , Concentración de Iones de Hidrógeno , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
14.
Int J Oncol ; 26(5): 1321-7, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15809724

RESUMEN

pRb and its family members p130 and p107 regulate cell cycle progression and direct G1/S transition in mammalian cells through interaction with the transcription factor E2F. Phosphatidylinositol 3-kinase (PI3K) is an essential component of growth factor-regulated pathways and plays a crucial role in the regulation of cellular proliferation and differentiation. It has been demonstrated that PI3K can regulate cell cycle progression via Akt-mediated pathway. However, the possible interactions between PI3K and Rb pathways remain to be defined. It was reported that the unique 24-amino-acid N-terminal end of the p55 regulatory subunits of PI3K is an Rb-binding domain and affects Rb action or Rb-E2F interaction. The 24 N-terminal amino acids of p55gamma encoded by a cDNA construct could compete with the endogenous p55gamma for binding to Rb, which influences Rb-mediated signaling and blocks cell cycle progression. In the current study, we investigated the effects of this 24-peptide on cell proliferation in human gastric carcinoma MKN-28 cells by means of cell cycle analysis, BrdU incorporation, and determining the levels of cell cycle regulatory molecule expression. Our results showed that p55gammaPI3K and the Rb family members p130 and p107 exist in MKN-28 cells, while the p110PI3K was not detected. Moreover, p55gammaPI3K was found binding to p130/p107 in these cells. We demonstrated that the introduction of the plasmid N24p55-GFP (harboring the cDNA for the 24 N-terminal amino acids of p55gamma) into MKN-28 cells caused cell cycle arrest at G1. Furthermore, we showed that the over-expression of the 24-peptide in MKN-28 cells decreased the population of cells incorporating BrdU and reduced the levels of cyclin D1 and cyclin A. These observations suggest that PI3K can regulate cell cycle progression and cell proliferation in human gastric tumor cells via Rb-mediated pathway, and that this effect of PI3K is mediated through a direct association with Rb via the N-terminal end of its p55 kDa regulatory subunits and modulating Rb-E2F interactions. Taken together with previous studies, our data provide a new therapeutic target in human stomach cancer. Strategies targeting PI3K signal transduction or the association of PI3K with Rb, or regulating PI3K-Rb interactions could be employed for gene therapy or chemotherapy of gastric cancer and other tumors.


Asunto(s)
Carcinoma/genética , Carcinoma/patología , Ciclo Celular/genética , Ciclo Celular/fisiología , Fosfatidilinositol 3-Quinasas/biosíntesis , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Proliferación Celular , ADN Complementario/genética , Humanos , Fragmentos de Péptidos , Proteína de Retinoblastoma/metabolismo , Transducción de Señal , Células Tumorales Cultivadas , Regulación hacia Arriba
15.
Int J Oncol ; 27(3): 699-704, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16077919

RESUMEN

Caspase-3 is a critical effector caspase for apoptosis, which cleaves proteins, including cytoskeletal and associated proteins, kinases, and members of the Bcl-2 family of apoptosis-related proteins. This leads to changes in apoptotic morphology, such as membrane externalization and cytoplasm and nuclear condensation. It has been reported that pro-caspase-3 is activated in the cytosol. However, it remains obscure how caspase-3 activation correlates to serial changes in cell morphology during apoptosis. The current study was therefore undertaken to assess the relationship between caspase-3 activation and its subcellular localization and alterations in apoptotic morphology in MOLT-4 human leukemia cells exposed to X-ray irradiation. Fluorescence labeled inhibitor of caspases (FLICA) was used to detect caspase-3 activity in apoptotic cells in this project; cell morphology and caspase-3 sub-localization were determined by confocal microscopy. Our data showed that MOLT-4 cells presented typical morphological changes in apoptosis, such as membrane reversion, DNA fragmentation, and formation of apoptotic cell bodies following 10 Gray (Gy) of X-ray irradiation. Caspase-3 was activated 2 h after X-ray irradiation, and its activity increased markedly after 4-6-h exposure. Membrane reversion in MOLT-4 leukemia cells was detected by Annexin V assay at 4 h following X-ray irradiation, 2 h after the elevated caspase-3 activity was measured. Cytologically, activation of caspase-3 was first observed close to the inside surface of the cellular membrane, then transferred to the cytoplasm, and finally translocated to the nuclear region. We conclude that caspase-3 is activated in MOLT-4 cells following exposure to X-rays, and that the enhanced caspase-3 activity and its sub-localization shifting is correlated to changes in apoptotic morphology. The spatial shift of activated caspase-3 in X-ray-induced apoptotic MOLT-4 leukemia cells is a process of crucial importance for apoptosis.


Asunto(s)
Apoptosis/efectos de la radiación , Caspasas/metabolismo , Caspasa 3 , Caspasas/química , Línea Celular Tumoral , Membrana Celular/metabolismo , Membrana Celular/efectos de la radiación , Forma de la Célula/efectos de la radiación , Fragmentación del ADN/efectos de la radiación , Activación Enzimática/efectos de la radiación , Precursores Enzimáticos/metabolismo , Citometría de Flujo/métodos , Colorantes Fluorescentes/química , Humanos , Leucemia/metabolismo , Leucemia/patología , Microscopía Confocal , Factores de Tiempo , Rayos X
16.
Oncol Rep ; 13(5): 793-9, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15809740

RESUMEN

Chemotherapy plays a major role in cancer management; however, acquired drug resistance remains a significant problem for ovarian cancer treatment. Chemoresistance is regulated by the coordinated expression of a set of genes. Thus, the identification of genes specifically modulated in the process provides an important step toward the discovery of underlying molecular mechanisms in drug resistance events. We recently developed five drug-resistant human ovarian carcinoma cell lines, including two cisplatin (cis) resistant cell lines, two carboplatin (car) resistant cell lines and one taxol (tax) resistant cell line. In this study, we investigated differential gene expression between these resistant cell lines and their parental cell lines by the fluorescence differential display-polymerase chain reaction (FDD-PCR) technique. We first screened and identified differentially expressed genes in the resistant ovarian cancer cell lines, and we then sequenced and analyzed these genes by bioinformatics software. A total of 33 fragments were displayed in the two resistant cell lines (S-cis and S-car) derived from the sensitive SKOV-3 cell line, and 36 fragments were displayed in the three resistant cell lines (A-cis, A-car and A-tax) derived from the sensitive A2780 cell line. After purification, cloning, sequencing, and homology analysis on the NCBI BLAST GenBank, 12 gene fragments were identified from the resistant S-cis and S-car cells, and 23 gene fragments were identified from the resistant A-cis, A-car and A-tax cells. Although a homolog search of the NIH GenBank revealed that most of the gene fragments were not significantly associated with the known drug resistance-related genes, our study conclusively demonstrates that FDD-PCR is a useful tool for analyzing the differential gene expression between resistant and sensitive tumor cells and for identifying novel chemoresistance-associated genes and potential biological markers or genetic markers of drug resistance.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Neoplasias Ováricas/genética , Reacción en Cadena de la Polimerasa , Secuencia de Bases , Línea Celular Tumoral , Clonación Molecular , Cartilla de ADN , Resistencia a Antineoplásicos , Femenino , Perfilación de la Expresión Génica , Humanos , Datos de Secuencia Molecular , Proteínas de Neoplasias/genética , Reacción en Cadena de la Polimerasa/métodos , Regiones Promotoras Genéticas
17.
Int J Mol Med ; 16(4): 645-51, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16142399

RESUMEN

The use of 'double-thymidine block' was the first widely accepted method for inducing cell synchrony and remains one of the most effective and frequently used techniques for analyzing the cell cycle today. While thymidine is in itself an inhibitor of DNA replication, thymidine blocks are typically used to generate cell synchrony at the G1/S boundary. We have previously presented the first evidence that shows the growth imbalance and altered expression levels of cyclins A, B1, D3 and E in MOLT-4 cells synchronized in the cell cycle by thymidine. The major objective of the present study was to compare the levels of cyclins A, B1, D3 and E in G1 phase of the cell cycle between synchronized and unperturbed asynchronously growing human lymphocyte leukemia MOLT-4 cells. Here, we demonstrate that the sorted, asynchronously growing MOLT-4 cells had considerably lower levels of cyclins A, B1, D3 and E than their counterparts of the cells arrested in G1/S phase, as assessed by flow cytometry. In addition, we confirmed these results by using post-sorting Western blotting, a new method we recently developed for examining protein expression in specific phases of sorted, synchronized or asynchronously growing cells. Our findings revealed that the levels of cyclins D3 and E in the asynchronously growing MOLT-4 cells were significantly lower than those in synchronized cultures. Interestingly, protein expression levels of cyclins A and B1 in the asynchronously growing MOLT-4 cells were barely measurable, suggesting that these proteins were either not expressed or under detectable levels. These studies indicate that our synchronization protocol may have disturbed cell proliferation and metabolism as evidenced by significant differences in the expression of cyclins between asynchronously growing and synchronized cells, and further suggest that the levels of cyclins A, B1, D3 and E in synchronized cultures cannot represent those in unperturbed, asynchronously growing cells. Thus, it appears that thymidine-treated, synchronized cells may not be suitable experimental models for the study of normal cell cycle.


Asunto(s)
División Celular/fisiología , Ciclinas/análisis , Fase G1 , Western Blotting , Ciclo Celular/efectos de los fármacos , División Celular/efectos de los fármacos , División Celular/genética , Línea Celular Tumoral , Ciclina A/análisis , Ciclina B/análisis , Ciclina B1 , Ciclina D3 , Ciclina E/análisis , ADN de Neoplasias/análisis , ADN de Neoplasias/genética , Citometría de Flujo , Humanos , Timidina/farmacología
18.
Int J Oncol ; 25(2): 445-51, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15254743

RESUMEN

SU5416 is a selective inhibitor of vascular endothelial growth factor (VEGF) receptors with anti-angiogenesis activity for human cancers. We have previously reported that SU5416 sensitizes ovarian cancer cells to cisplatin via suppression of nucleotide excision repair activity. This study sought to gain further insights into the mechanisms underlying the synergistic effect of SU5416 and cisplatin on cytotoxicity in human ovarian tumor cells. Here, we show that SU5416 inhibited the expression of G1 cell cycle checkpoint regulators, p53, p21, p27 and MDM2 in ovarian carcinoma cells. We also demonstrate that SU5416 triggered the apoptosis of these cells, in addition to augmenting the apoptosis induced by cisplatin, as determined by a Sub-G1 profile analysis using a flow cytometer. Furthermore, we show that SU5416-induced apoptosis is associated with a decrease in the expression of the apoptosis inhibitors, MDM2 and Bcl-2, and an increase in the level of NF-kappaB inhibitor, IkappaBalpha. NF-kappaB is an anti-apoptotic transcription factor, which induces the apoptosis inhibitors, Bcl-XL and IAPs (inhibitor of apoptosis proteins), and IkappaBalpha is an inhibitor of NF-kappaB, which binds to the NF-kappaB and retains it in the cytoplasm. Finally, the compound was found to block cisplatin-induced increases in AP-1 expression and JNK activity, as well as Raf-1 protein level in these cells. Together, these results suggest that the chemosensitizing effect of SU5416 on ovarian tumor cells may be mediated, at least in part, through inhibiting G1 checkpoint control and up-regulating the apoptotic response to cisplatin.


Asunto(s)
Inhibidores de la Angiogénesis/toxicidad , Antineoplásicos/toxicidad , Cisplatino/toxicidad , Indoles/toxicidad , Neoplasias Ováricas/metabolismo , Pirroles/toxicidad , Inhibidores de la Angiogénesis/farmacología , Inhibidores de la Angiogénesis/uso terapéutico , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Apoptosis , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Cisplatino/farmacología , Cisplatino/uso terapéutico , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Sinergismo Farmacológico , Femenino , Humanos , Proteínas I-kappa B/metabolismo , Indoles/farmacología , Indoles/uso terapéutico , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , MAP Quinasa Quinasa 4 , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Inhibidor NF-kappaB alfa , Neoplasias Ováricas/tratamiento farmacológico , Proteínas Proto-Oncogénicas/metabolismo , Pirroles/farmacología , Pirroles/uso terapéutico , Factor de Transcripción AP-1/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Regulación hacia Arriba
19.
Oncol Rep ; 12(5): 955-65, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15492778

RESUMEN

Cisplatin is one of the most potent anticancer agents, displaying significant clinical activity against a variety of solid tumors. For more than two decades, the most effective systemic chemotherapy for non-small cell lung cancer (NSCLC), the leading cause of cancer morbidity and mortality among men and women in the western world, was cisplatin-based combination treatment. Unfortunately, the outcome of cisplatin therapy on NSCLC seems to have reached a plateau. Therefore, the biological mechanisms of cisplatin action need to be understood in order to overcome the treatment plateau on NSCLC. Moreover, the development of resistance is a hurdle in the use of this drug. The molecular mechanisms that underlie this chemoresistance are largely unknown. Possible mechanisms of acquired resistance to cisplatin include reduced intracellular accumulation of cisplatin, enhanced drug inactivation by metallothionine and glutathione, increased repair activity of DNA damage, and altered expression of oncogenes and regulatory proteins. In addition, it is generally accepted that cytotoxicity of cisplatin is mediated through induction of apoptosis and arrest of cell cycle resulting from its interaction with DNA, such as the formation of cisplatin-DNA adducts, which activates multiple signaling pathways, including those involving p53, Bcl-2 family, caspases, cyclins, CDKs, pRb, PKC, MAPK and PI3K/Akt. Increased expression of anti-apoptotic genes and mutations in the intrinsic apoptotic pathway may contribute to the inability of cells to detect DNA damage or to induce apoptosis. Towards an understanding of the molecular basis of the cellular response to cisplatin-based chemotherapy in NSCLC, in this review we provide some insights into the pathways involved in cisplatin damage from entering the cells to execution of apoptosis or survival of NSCLC cells. We believe that as more and more molecular mechanisms of response to cisplatin-based therapy are unraveled, this knowledge should provide a basis for further studies to improve our understanding of molecular events associated with lung NSCLC as well as to devise novel and effective therapeutic approaches to overcome the treatment plateau or reverse drug resistance in this disease.


Asunto(s)
Antineoplásicos/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Cisplatino/uso terapéutico , Neoplasias Pulmonares/tratamiento farmacológico , Animales , Humanos
20.
Oncol Rep ; 9(5): 1041-5, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12168070

RESUMEN

Cell cycle analysis has become increasingly important in verifying the effect of anti-tumor drugs and cytokinetic research. In the early methods of cell cycle analysis, the flow cytometry relied on DNA content, and therefore, the cell cycle could be only broken into three stages: G(0)/G(1), S, and G(2)/M phase. It could not distinguish the G(0), G(1), G(2), and M phase cells, let alone the sub-phases in G(1) phase. In cell cycle, expression of cyclin E living up to the maximal level in the cells undergoing transition from G(1) to S phase, and G(2) + M cells are cyclin E negative. Expression of cyclin A is progressively increasing during S phase and is maximal in G(2) phase cells. Therefore, in the current study we established a cyclin E + A/DNA multiparameter flow cytometric technique by using a mixture of cyclin E and cyclin A antibodies, which can identify six stages in the whole cell cycle: G(0), early G(1), late G(1), S, G(2), and M phase. Furthermore, we found that cyclin E + A/DNA multiparameter flow cytometry could also be used for stathmokinetic analysis of lymphocyte leukemia MOLT-4 cells after addition of the stathmokinetic agent vinblastine to cultures of exponentially growing MOLT-4 cells. We believe that this new technique will provide a much better tool for molecular cell biology research and especially for cell proliferation kinetics investigations.


Asunto(s)
Separación Celular/métodos , Ciclina A/metabolismo , Ciclina E/metabolismo , Antineoplásicos Fitogénicos/farmacología , Ciclo Celular , División Celular , Ciclina A/inmunología , Ciclina E/inmunología , Fase G1 , Fase G2 , Humanos , Inmunohistoquímica , Cinética , Leucemia Linfoide/metabolismo , Mitosis , Fase S , Factores de Tiempo , Células Tumorales Cultivadas , Vinblastina/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA