Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
J Virol ; 75(15): 7050-8, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11435585

RESUMO

G207 is an oncolytic herpes simplex virus (HSV) which is attenuated by inactivation of viral ribonucleotide reductase (RR) and deletion of both gamma(1)34.5 genes. The cellular counterparts that can functionally substitute for viral RR and the carboxyl-terminal domain of ICP34.5 are cellular RR and the corresponding homologous domain of the growth arrest and DNA damage protein 34 (GADD34), respectively. Because the thymidylate synthetase (TS) inhibitor fluorodeoxyuridine (FUdR) can alter expression of cellular RR and GADD34, we examined the effect of FUdR on G207 bioactivity with the hypothesis that FUdR-induced cellular changes will alter viral proliferation and cytotoxicity. Replication of wild-type HSV-1 was impaired in the presence of 10 nM FUdR, whereas G207 demonstrated increased replication under the same conditions. Combined use of FUdR and G207 resulted in synergistic cytotoxicity. FUdR exposure caused elevation of RR activity at 10 and 100 nM, whereas GADD34 was induced only at 100 nM. The effect of enhanced viral replication by FUdR was suppressed by hydroxyurea, a known inhibitor of RR. These results demonstrate that the growth advantage of G207 in FUdR-treated cells is primarily based on an RR-dependent mechanism. Although our findings show that TS inhibition impairs viral replication, the FUdR-induced RR elevation may overcome this disadvantage, resulting in enhanced replication of G207. These data provide the cellular basis for the combined use of RR-negative HSV mutants and TS inhibitors in the treatment of cancer.


Assuntos
Antivirais/farmacologia , Floxuridina/farmacologia , Herpesvirus Humano 1/enzimologia , Ribonucleotídeo Redutases/metabolismo , Proteínas Virais/metabolismo , Replicação Viral/efeitos dos fármacos , Animais , Antígenos de Diferenciação , Ciclo Celular , Proteínas de Ciclo Celular , Chlorocebus aethiops , Expressão Gênica , Genes Reporter , Herpesvirus Humano 1/efeitos dos fármacos , Herpesvirus Humano 1/fisiologia , Humanos , Mutagênese Insercional , Proteína Fosfatase 1 , Proteínas/genética , Ribonucleotídeo Redutases/genética , Células Tumorais Cultivadas , Células Vero , Proteínas Virais/genética , beta-Galactosidase/genética
2.
Cancer Res ; 61(2): 439-44, 2001 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-11212227

RESUMO

The mechanisms underlying neoplastic epithelial cell killing by ionizing radiation are largely unknown. We discovered a novel response to radiation manifested by autophagy and the development of acidic vesicular organelles (AVO). Acidification of AVO was mediated by the vacuolar H+-ATPase. Staining with the lysosomotropic agent acridine orange enabled us to quantify AVO accumulation and to demonstrate their time- and dose-dependent appearance. The appearance of AVO occurred in the presence of the pan-caspase inhibitor z-Val-Ala-Asp(Ome)-fluoromethyl ketone, but was inhibited by 3-methyladenine, an inhibitor of autophagy. The accretion of AVO in surviving progenies of irradiated cells, and the increased incidence of clonogenic death after inhibition of vacuolar H+-ATPase suggest that formation of acidic organelles represents a novel defense mechanism against radiation damage.


Assuntos
Autofagia/efeitos da radiação , Vesículas Citoplasmáticas/efeitos da radiação , ATPases Vacuolares Próton-Translocadoras , Adenina/análogos & derivados , Adenina/farmacologia , Vesículas Citoplasmáticas/efeitos dos fármacos , Vesículas Citoplasmáticas/ultraestrutura , DNA de Neoplasias/genética , DNA de Neoplasias/efeitos da radiação , Relação Dose-Resposta à Radiação , Humanos , Concentração de Íons de Hidrogênio , Microscopia Eletrônica , ATPases Translocadoras de Prótons/metabolismo , Fatores de Tempo , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/efeitos da radiação , Células Tumorais Cultivadas/ultraestrutura
3.
Cancer Res ; 61(2): 725-31, 2001 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-11212275

RESUMO

Epidemiological studies link increased garlic (Allium sativum) consumption with a reduced incidence of colon cancer in various human populations. Experimental carcinogenesis studies in animal models and in cell culture systems indicate that several allium-derived compounds exhibit inhibitory effects and that the underlying mechanisms may involve both the initiation and promotion phases of carcinogenesis. To provide a better understanding of the effects of allium derivatives on the prevention of colon cancer, we examined two water-soluble derivatives of garlic, S-allylcysteine (SAC) and S-allylmercaptocysteine (SAMC), for their effects on proliferation and cell cycle progression in two human colon cancer cell lines, SW-480 and HT-29. For comparison, we included the compound sulindac sulfide (SS), because sulindac compounds are well-established colon cancer chemopreventive agents. We found that SAMC, but not SAC, inhibited the growth of both cell lines at doses similar to that of SS. SAMC also induced apoptosis, and this was associated with an increase in caspase3-like activity. These affects of SAMC were accompanied by induction of jun kinase activity and a marked increase in endogenous levels of reduced glutathione. Although SS caused inhibition of cell cycle progression from G1 to S, SAMC inhibited progression at G2-M, and a fraction of the SW-480 and HT-29 cells were specifically arrested in mitosis. Coadministration of SS with SAMC enhanced the growth inhibitory and apoptotic effects of SS. These findings suggest that SAMC may be useful in colon cancer prevention when used alone or in combination with SS or other chemopreventive agents.


Assuntos
Antineoplásicos/farmacologia , Divisão Celular/efeitos dos fármacos , Cisteína/análogos & derivados , Cisteína/farmacologia , Sulindaco/farmacologia , Apoptose/efeitos dos fármacos , Western Blotting , Caspase 3 , Caspases/efeitos dos fármacos , Caspases/metabolismo , Ciclo Celular/efeitos dos fármacos , Cisteína/química , Relação Dose-Resposta a Droga , Ativação Enzimática/efeitos dos fármacos , Fase G2/efeitos dos fármacos , Alho/química , Glutationa/efeitos dos fármacos , Glutationa/metabolismo , Células HT29 , Humanos , Hibridização in Situ Fluorescente , Isoenzimas/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno , Cinética , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Mitose/efeitos dos fármacos , Plantas Medicinais , Proteínas Proto-Oncogênicas c-bcl-2/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Sulindaco/análogos & derivados , Células Tumorais Cultivadas/citologia , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/metabolismo
4.
Infect Immun ; 68(9): 5321-8, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10948161

RESUMO

Helicobacter pylori infection is associated with the development of gastric cancer. In short-term coculture with AGS gastric cells, H. pylori inhibits cell cycle progression and induces dose-dependent apoptosis. Based on the concept that an imbalance between proliferation and apoptosis may contribute to the emergence of gastric cancer, we chronically exposed AGS cells to H. pylori as a model of chronic exposure in humans. The AGS derivatives selected by this process were stably resistant not only to H. pylori-induced apoptosis but also to apoptosis induced by other enteric bacteria and by several toxic agents including radiation and cancer chemotherapy. Like the parental AGS cells, the derivatives underwent G(1)/S-phase cell cycle inhibition in response to H. pylori. The AGS derivatives displayed a marked decrease in cellular levels of the cell cycle control protein p27(kip1). We found a similar decrease in epithelial cell p27(kip1) expression in gastric biopsy specimens from H. pylori-infected patients. These findings are consistent with observations that link decreases in the p27(kip1) level to increased susceptibility to cancer in mice with p27(kip1) deleted and to a poor prognosis of gastric cancer in humans. This is the first demonstration that bacterial infection can lead to apoptosis resistance and to cross-resistance to other inducers of apoptosis such as bacteria, chemotherapeutic agents, and radiation. The development of apoptosis resistance and downmodulation of p27(kip1) may contribute to the increased risk for gastric cancer observed in humans chronically exposed to H. pylori.


Assuntos
Apoptose , Proteínas de Ciclo Celular , Mucosa Gástrica/patologia , Infecções por Helicobacter/patologia , Helicobacter pylori , Proteínas Associadas aos Microtúbulos/análise , Proteínas Supressoras de Tumor , Aderência Bacteriana , Ciclo Celular , Divisão Celular , Linhagem Celular , Doença Crônica , Inibidor de Quinase Dependente de Ciclina p27 , Mucosa Gástrica/química , Genes p53 , Infecções por Helicobacter/complicações , Infecções por Helicobacter/metabolismo , Humanos , Fenótipo , Neoplasias Gástricas/etiologia
5.
Mol Med ; 5(9): 606-16, 1999 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10551902

RESUMO

BACKGROUND: Costimulatory and cellular adhesion molecules are thought to be essential components of antigen presentation in the immune response to cancer. The current studies examine gene transfer utilizing herpes viral amplicon vectors (HSV) to direct surface expression of adhesion molecules, and specifically evaluate the potential of a tumor-expressing intercellular adhesion molecule-1 (ICAM-1) to elicit an anti-tumor response. MATERIALS AND METHODS: The human ICAM-1 (hICAM1) gene was inserted into an HSV amplicon vector and tested in a transplantable rat hepatocellular carcinoma and in a human colorectal cancer cell line. Cell surface ICAM-1 expression was assessed by flow cytometry. Lymphocyte binding to HSV-hICAM1-transduced cells was compared with that to cells transduced with HSV not carrying the ICAM gene. Tumorigenicity of HSV-hICAM1-transduced tumor cells were tested in syngeneic Buffalo rats. Additionally, immunization with irradiated (10,000 rads) HSV-hICAM1-transduced tumor cells was performed to determine its effect on tumor growth. RESULTS: A 20-min exposure of tumor cells at a multiplicity of infection (MOI) of 1 resulted in high-level cell surface expression of human ICAM in approximately 25% of tumor cells. Transduced rat or human tumor cells exhibited significantly enhanced binding of lymphocytes (p < 0.05). HSV-hICAM1-transduced cells elicited an increase in infiltration by CD4(+) lymphocytes in vivo and exhibited decreased tumorigenicity. Immunization with irradiated HSV-hICAM1-transduced cells protected against growth of subsequent injected parental tumor cells. CONCLUSIONS: HSV amplicon-mediated gene transfer is an efficient method for modifying the cell surface expression of adhesion molecules. Increased tumor expression of ICAM-1 represents a promising immune anti-cancer strategy.


Assuntos
Carcinoma Hepatocelular/imunologia , Imunidade Inata/genética , Molécula 1 de Adesão Intercelular/genética , Neoplasias Hepáticas/imunologia , Simplexvirus/genética , Animais , Testes de Carcinogenicidade , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patologia , Adesão Celular , Divisão Celular/genética , Técnicas de Transferência de Genes , Humanos , Imuno-Histoquímica/métodos , Molécula 1 de Adesão Intercelular/imunologia , Molécula 1 de Adesão Intercelular/metabolismo , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patologia , Masculino , Ratos , Células Tumorais Cultivadas , Vacinas/administração & dosagem
6.
Biochem Pharmacol ; 58(7): 1097-107, 1999 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-10484067

RESUMO

We examined the activity of two metabolites of sulindac (a nonsteroidal anti-inflammatory drug), sulindac sulfide and sulindac sulfone (exisulind, Prevatec), and a novel highly potent analog of exisulind (CP248) on a series of human prostate epithelial cell lines. Marked growth inhibition was seen with the BPH-1, LNCaP, and PC3 cell lines with IC50 values of about 66 microM, 137 microM, and 64 nM for sulindac sulfide, exisulind, and CP248, respectively. DNA flow cytometry and 4',6'-diamido-2-phenylindole (DAPI) staining indicated that these three compounds also induced apoptosis in all of these cell lines. Similar growth inhibition also was seen with the PrEC normal human prostate epithelial cell line, but these cells were resistant to induction of apoptosis at concentrations up to 300 microM, 1 mM, and 750 nM of sulindac sulfide, exisulind, and CP248, respectively. Derivatives of LNCaP cells that stably overexpress bcl-2 remained sensitive to growth inhibition and induction of apoptosis by these compounds. In vitro enzyme assays indicated that despite its high potency in inhibiting growth and inducing apoptosis, CP248, like exisulind, lacked cyclooxygenase (COX-1 and COX-2) inhibitory activity even at concentrations up to 10 mM. Moreover, despite variations of COX-1 and COX-2 expression, the three benign and malignant prostate cell lines showed similar sensitivity to growth inhibition and induction of apoptosis by these three compounds. Therefore, sulindac derivatives can cause growth inhibition and induce apoptosis in human prostate cancer cells by a COX-1 and -2 independent mechanism, and this occurs irrespective of androgen sensitivity or increased expression of bcl-2. These compounds may be useful in the prevention and treatment of human prostate cancer.


Assuntos
Antineoplásicos/farmacologia , Apoptose , Neoplasias da Próstata/tratamento farmacológico , Sulindaco/farmacologia , Androgênios/metabolismo , Anti-Inflamatórios não Esteroides/farmacologia , Divisão Celular/efeitos dos fármacos , Inibidores de Ciclo-Oxigenase/farmacologia , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Masculino , Prostaglandina-Endoperóxido Sintases/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Sulindaco/análogos & derivados , Células Tumorais Cultivadas
7.
Clin Cancer Res ; 5(7): 1876-83, 1999 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10430095

RESUMO

Although in the past 10 years paclitaxel has emerged as a successful drug in cancer therapy, the overall response rate to this drug in patients with advanced metastatic disease remains low. Therefore, an understanding of the mechanism of the effect of paclitaxel on inducing apoptosis and the discovery of new ways to enhance the effect of paclitaxel will be critical to improving the therapeutic efficiency of this drug. In the present studies, we have determined that the cyclin-dependent kinase inhibitor flavopiridol significantly enhances paclitaxel-induced apoptosis in the human gastric and breast cancer cell lines MKN-74 and MCF-7. Flavopiridol enhances paclitaxel-induced apoptosis only when administered after paclitaxel treatment. The activation of caspases, specifically caspase 3, is enhanced by flavopiridol on paclitaxel-treated cells. In accordance with this, poly(ADP-ribose) polymerase cleavage is enhanced in combination therapy relative to single-agent paclitaxel. The induction of apoptosis, activation of caspase 3, and poly(ADP-ribose) polymerase cleavage in treatment regimens with paclitaxel and paclitaxel followed by flavopiridol were reversed by treatment with the caspase inhibitor benzyloxycarbonyl-Val-Ala-Asp-fluoromethylketone, which supports the notion that caspases are the executioners of apoptosis in these processes. Paclitaxel alone causes transient mitotic arrest with activation of cdc-2 kinase. Cells exit mitosis in a specific time window without cytokinesis, with a decrease in cdc-2 kinase activity and MPM-2 labeling. Flavopiridol accelerates the mitotic exit when administered after paclitaxel treatment in association with a more rapid decrease in MPM-2 labeling. In contrast, pretreatment with flavopiridol prevents cells from entering mitosis by inhibiting cdc-2 kinase activity, thus antagonizing the paclitaxel effect. Therefore, in this study we show that potentiation of paclitaxel-induced apoptosis by flavopiridol is highly sequence dependent, such that mitotic entry and cdc-2 kinase activation by paclitaxel must precede flavopiridol therapy, and the synergistic effect of flavopiridol on paclitaxel-treated cells is due to enhancement in caspase activation.


Assuntos
Antineoplásicos/farmacologia , Apoptose , Neoplasias da Mama/enzimologia , Caspases/metabolismo , Flavonoides/farmacologia , Paclitaxel/farmacologia , Piperidinas/farmacologia , Neoplasias Gástricas/enzimologia , Neoplasias da Mama/metabolismo , Proteína Quinase CDC2/metabolismo , Ciclina B/metabolismo , Ciclina B1 , Interações Medicamentosas , Ativação Enzimática , Humanos , Mitose/efeitos dos fármacos , Poli Adenosina Difosfato Ribose/metabolismo , Proteína do Retinoblastoma/metabolismo , Neoplasias Gástricas/metabolismo , Células Tumorais Cultivadas
8.
Cancer Res ; 59(10): 2277-81, 1999 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-10344728

RESUMO

Infection with the bacterium Helicobacter pylori is associated epidemiologically with development of gastric cancer. To better understand the role of H. pylori in carcinogenesis, we examined the effects of H. pylori on cell cycle-related events in the AGS gastric cancer cell line. During coculture, wild-type, toxigenic, cagA-positive H. pylori induced both apoptosis and inhibition of cell cycle progression at G1-S in AGS cells. These effects were most apparent in AGS cells synchronized by serum-deprivation and then stimulated to progress through the cell cycle by refeeding. An isogenic cagA-negative mutant H. pylori, produced similar effects. In contrast to changes induced by 5-fluorouracil, the inhibition of cell cycle progression from G1 to S caused by H. pylori was not accompanied by sustained changes in p53 or p21cip1, but was associated with reduced expression of p27kip1 and inhibition of transcriptional activation of the serum-response element of c-fos. Our results indicate that H. pylori inhibits cell cycle progression at G1-S and induces apoptosis, associated with reduced expression of p27kip1 in AGS gastric cancer cells. In vivo, similar effects as a result of H. pylori infection may lead to potentially deleterious compensatory hyperproliferation by nonneoplastic gastric epithelial cells.


Assuntos
Antígenos de Bactérias , Proteínas de Ciclo Celular/biossíntese , Fase G1 , Mucosa Gástrica/patologia , Regulação Bacteriana da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Helicobacter pylori/fisiologia , Fase S , Neoplasias Gástricas/patologia , Apoptose/efeitos dos fármacos , Proteínas de Bactérias/genética , Proteínas de Ciclo Celular/genética , Meios de Cultura Livres de Soro/farmacologia , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Fluoruracila/farmacologia , Mucosa Gástrica/microbiologia , Genes fos , Helicobacter pylori/genética , Humanos , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/genética , Células Tumorais Cultivadas
9.
Anticancer Res ; 18(5A): 3139-52, 1998.
Artigo em Inglês | MEDLINE | ID: mdl-9858877

RESUMO

Protein kinase C (PKC) is a central component in signal transduction and growth control and might be an appropriate target for the chemotherapy of human brain tumors. This study demonstrates that the staurosporine derivative Ro 31-8220, a potent PKC inhibitor, inhibited the growth of 7 human brain tumor cell lines with an IC50 of about 2 microM. Calphostin C, a structurally unrelated PKC inhibitor, inhibited the growth of two of these cell lines with an IC50 of about 100 to 300 nM. Drug withdrawal and clonogenicity assays indicated that the growth inhibition by both of these compounds was irreversible. Morphologic studies, DNA fragmentation studies and flow cytometric assays showed that the treated glioblastoma cells underwent apoptosis. Treatment of glioblastoma cells with Ro 31-8220 lead to a rapid decline in the level of the anti-apoptosis protein bcl-2. At least three of the glioblastoma cell lines carried mutant p53 alleles with missense mutations in the DNA binding domain of p53. Therefore, the induction of apoptosis in these cell lines occurred through a p53-independent mechanism. Furthermore treatment of these glioblastoma cell lines with Ro 31-8220 or calphostin C led to an increase of cells in the G2-M phase of the cell cycle. This correlated with a decrease in CDC2-associated histone H1 kinase activity, as well as a decrease in the level of the CDC2 protein as shown by immunoblotting. When added to subcellular assays Ro 31-8220 markedly inhibited CDC2 histone H1 kinase activity with an IC50 of 100 nM, but calphostin C directly inhibited this kinase activity only at very high concentrations (above 100 microM). Thus these compounds inhibit the growth of glioblastoma cells through novel mechanisms. Ro 31-8220, in particular, might be a useful agent for the treatment of human brain tumors.


Assuntos
Anticarcinógenos/uso terapêutico , Apoptose , Astrocitoma/tratamento farmacológico , Neoplasias Encefálicas/tratamento farmacológico , Indóis/uso terapêutico , Naftalenos/uso terapêutico , Proteína Quinase C/antagonistas & inibidores , Apoptose/genética , Proteína Quinase CDC2/metabolismo , Divisão Celular/efeitos dos fármacos , Fragmentação do DNA , DNA de Neoplasias/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Genes p53/efeitos dos fármacos , Glioblastoma/tratamento farmacológico , Humanos , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Células Tumorais Cultivadas/efeitos dos fármacos
10.
Breast Cancer Res Treat ; 48(3): 195-203, 1998 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9598866

RESUMO

Nonsteriodal anti-inflammatory drugs (NSAIDs) are among the most commonly used medications in the United States and elsewhere, mainly for the treatment of arthritis. The NSAID sulindac causes regression and prevents the recurrence of premalignant colonic polyps in patients with familial adenomatous polyposis and inhibits colon carcinogenesis in rodents. Sulindac and sulindac sulfone, a metabolite of sulindac that lacks cyclooxygenase (cox) inhibitory activity, also inhibit mammary carcinogenesis in rats. To obtain insights into the relevance of these findings to human breast cancer, we examined the mechanism of action of sulindac and its sulfide and sulfone metabolites on the normal human mammary epithelial cell line MCF-10F and the human breast cancer cell line MCF-7. Of the three compounds, the sulfide was the most potent inhibitor of cell growth, although the sulfone and sulfoxide were also active at higher concentrations. Treatment of MCF-10F and MCF-7 cells with 100 microM sulindac sulfide resulted in accumulation of cells in the G1 phase of the cell cycle and induction of apoptosis. Apoptosis occurred within 24 h as determined by the TUNEL assay and DNA laddering was observed at 72 h. The accumulation of cells in G1 was associated with decreased levels of expression of cyclin D1 but no effect was seen on the expression of CDK4 or the immediate early response gene c-jun. Treatment with sulindac sulfide caused a striking induction of the CDK inhibitor p21WAF1 in MCF-10F cells. The MCF-7 cell line expressed a high basal level of p21WAF1 which did not change significantly after drug treatment. The pro-apoptotic gene BAX was not induced in either MCF-10F or MCF-7 cells by sulindac sulfide. Stable overexpression of cyclin D1, which frequently occurs in breast cancers, did not protect mammary epithelial cells from inhibition by the sulfide. These studies suggest that this class of compounds warrants further study with respect to breast cancer prevention and treatment.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias da Mama/tratamento farmacológico , Mama/efeitos dos fármacos , Sulindaco/farmacologia , Anti-Inflamatórios não Esteroides/uso terapêutico , Antineoplásicos/uso terapêutico , Mama/citologia , Neoplasias da Mama/patologia , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Ciclina D1/análise , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/análise , Células Epiteliais/efeitos dos fármacos , Feminino , Humanos , Sulindaco/análogos & derivados , Sulindaco/uso terapêutico
11.
Nat Biotechnol ; 16(2): 196-201, 1998 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9487530

RESUMO

Neuronal precursor cells are widespread in the forebrain ventricular/subventricular zone, and may provide a cellular substrate for brain repair. Clonal lines derived from single progenitors can become progressively less representative of their parental precursors with time and passage in vitro. We have developed an alternative strategy for the isolation and enrichment of precursor cells, by fluorescence-activated cell sorting of forebrain cells transfected with the gene for green fluorescent protein, driven by the neuronal T alpha 1 tubulin promoter. Using this approach, neural precursors and young neurons can be identified and selectively harvested from a variety of samples, including both avian and mammalian forebrains at different developmental stages.


Assuntos
Proteínas Luminescentes/genética , Neurônios/citologia , Regiões Promotoras Genéticas , Células-Tronco/citologia , Tubulina (Proteína)/genética , Animais , Biomarcadores , Separação Celular , Células Cultivadas , Embrião de Galinha , Citometria de Fluxo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Fluorescência Verde , Prosencéfalo/citologia , Prosencéfalo/embriologia , Ratos , Transfecção
12.
Gastroenterology ; 113(6): 1892-900, 1997 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9394728

RESUMO

BACKGROUND & AIMS: Mutations of c-K-ras occur commonly in colonic neoplasms. The aim of this study was to determine how c-K-ras mutations alter the responses to the chemopreventive agent sulindac. METHODS: The parental rat intestinal cell line IEC-18 and c-K-ras-transformed derivatives were treated with sulindac sulfide. Cell cycle distribution was determined by flow-cytometric analysis (fluorescence-activated cell sorter), apoptosis by DNA fragmentation (laddering), flow cytometry, and microscopy, and changes in gene expression by immunoblotting. RESULTS: Sulindac sulfide inhibited cell growth and induced apoptosis in a time- and dose-dependent manner more rapidly in and at lower concentrations in parental cells than ras-transformed cells. Expression of the sulindac sulfide arrested cells in G0/G1, but cells entered apoptosis throughout the cell cycle. Proapoptotic protein Bak was relatively high in untreated parental cells and increased markedly after sulindac sulfide but was low in untreated ras-transformed cells and did not increase after sulindac sulfide. Expression of other Bcl-2 family members was unchanged after sulindac sulfide. However, sulindac sulfide reduced levels of cyclin D1 protein and cyclin E- and cyclin D1-associated kinase activity. CONCLUSIONS: c-K-ras-transformed enterocytes are relatively resistant to sulindac sulfide-induced growth inhibition and apoptosis, which may result from specific reduction of bak expression.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Apoptose/fisiologia , Genes ras/fisiologia , Intestinos/efeitos dos fármacos , Intestinos/fisiologia , Sulindaco/farmacologia , Animais , Ciclo Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Linhagem Celular Transformada , Resistência a Medicamentos/fisiologia , Regulação da Expressão Gênica/efeitos dos fármacos , Intestinos/citologia , Ratos
13.
Biochem Biophys Res Commun ; 237(3): 678-84, 1997 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-9299426

RESUMO

Micronuclei (MN) formation was defined as a form of radiation-induced damage in MCF-7 cells. MN appeared post-mitosis and were scored in bi-nucleated cells of cytochalasin B treated cultures. MN were surrounded by an envelope composed of inner and outer membranes, and contained fragmented chromosomes. However, typical features of apoptosis, such as chromatin margination or condensation were not observed. Reducing serum concentration resulted in a decreased MN formation, suggesting that serum factors directly affected MN formation and/or that serum depletion decreased the availability of radiation sensitive MN-forming cells for mitosis. Irradiation of G1 and S phase enriched populations revealed that S phase cells were more prone to MN formation than G1 cells. Radiation-induced chromosomal aberration can therefore be modulated by altering serum level and cell cycle distribution.


Assuntos
Ciclo Celular/fisiologia , Micronúcleos com Defeito Cromossômico/efeitos da radiação , Ciclo Celular/efeitos da radiação , Cromossomos Humanos/efeitos da radiação , Cromossomos Humanos/ultraestrutura , Meios de Cultura , Citocalasina B/farmacologia , Fragmentação do DNA , Relação Dose-Resposta à Radiação , Feminino , Fase G1 , Humanos , Micronúcleos com Defeito Cromossômico/efeitos dos fármacos , Micronúcleos com Defeito Cromossômico/ultraestrutura , Microscopia Eletrônica , Mitose , Fase S , Células Tumorais Cultivadas
14.
Cancer Res ; 57(8): 1569-74, 1997 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-9108461

RESUMO

Cyclin D1 plays an important role in regulating the progression of cells through the G1 phase of the cell cycle. This gene is frequently overexpressed in human colon cancer. To address the role of cyclin D1 in growth control and tumorigenesis in this disease, we have overexpressed an antisense cyclin D1 cDNA construct in the human colon cancer cell line SW480E8, which expresses high levels of cyclin D1. The integration and expression of the antisense construct was verified by Southern and Northern blot analyses, respectively, and resulted in decreased expression of the cyclin D1 protein. This was associated with decreased levels of the Rb and p27Kip1 proteins. In addition, the hypophosphorylated form of Rb was increased in these cells. The SW480E8 antisense cyclin D1 cells displayed an increased doubling time, a decrease in saturation density, decreased plating efficiency and anchorage-independent growth, and a loss of tumorigenicity in nude mice. These findings provide direct evidence that increased expression of cyclin D1 in colon tumor cells contributes to their abnormal growth and tumorigenicity. The ability to revert the transformed phenotype of these cells with antisense cyclin D1 suggests that cyclin D1 or its associated cyclin-dependent kinase 4 may be useful targets in the therapy of colon cancer.


Assuntos
Biomarcadores Tumorais/metabolismo , Proteínas de Ciclo Celular , Neoplasias do Colo/metabolismo , Ciclinas/metabolismo , DNA Antissenso/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas , Proteínas Supressoras de Tumor , Animais , Ciclo Celular , Divisão Celular/efeitos dos fármacos , Neoplasias do Colo/genética , Neoplasias do Colo/patologia , Ciclina D1 , Quinase 4 Dependente de Ciclina , Quinase 6 Dependente de Ciclina , Inibidor de Quinase Dependente de Ciclina p27 , Quinases Ciclina-Dependentes/metabolismo , Ciclinas/genética , Ciclinas/fisiologia , Regulação para Baixo , Vetores Genéticos , Humanos , Camundongos , Camundongos Nus , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas Oncogênicas/genética , Proteínas Oncogênicas/fisiologia , Proteínas Serina-Treonina Quinases/metabolismo , Proteína do Retinoblastoma/metabolismo , Células Tumorais Cultivadas
15.
Lasers Surg Med ; 20(3): 319-31, 1997.
Artigo em Inglês | MEDLINE | ID: mdl-9138261

RESUMO

BACKGROUND AND OBJECTIVE: The objective of this study was to examine the question of whether unique spectral patterns were associated with cell proliferation and could be identified by comparing the fluorescence pattern of slow to rapid growing cells. STUDY DESIGN/MATERIALS AND METHODS: Three in vitro model systems, (A431 cells inhibited by EGF, serum-starved 3T3 fibroblasts, and normal oral epithelial cells exposed to TGF beta), were analyzed using fluorescence spectroscopy. Growth status was monitored by cell number, 3H-thymidine incorporation, and flow cytometry. RESULTS: The excitation spectra (lambda ex 240-430 nm, lambda em 450 nm) effectively distinguished slow and rapid growing cells in all three systems. Statistical analysis of the ratios of the main broad peak (320-350 nm) to a point on the down-slope of the curve at 370 nm was statistically significant. Ratios in the emission scan (lambda ex 340 nm, lambda em 360-660 nm) could separate slow and rapid growing A431 and oral epithelial cells (P = 0.0001 and P = 0.023, respectively), but not slow and fast growing 3T3 cells (P = 0.56). CONCLUSION: Innate cellular fluorescence has the potential to discriminate proliferating and nonproliferating cell populations.


Assuntos
Divisão Celular , Espectrometria de Fluorescência , Animais , Contagem de Células , Linhagem Celular , Células Cultivadas , DNA/metabolismo , Fibroblastos/citologia , Citometria de Fluxo , Humanos , Camundongos , Mucosa Bucal/citologia , Células Tumorais Cultivadas
16.
Pathobiology ; 64(6): 339-46, 1996.
Artigo em Inglês | MEDLINE | ID: mdl-9159029

RESUMO

Human peripheral blood mononuclear cells from healthy donors were treated ex vivo with the proteolytic enzyme bromelain and studied by flow cytometry. Bromelain-treated lymphocytes exhibited 60-90% reduced cell surface staining for CD44 and CD62-L molecules. While the staining for molecules CD16, CD56 and CD49d was unaffected, a moderate increase (10-40%) in expression of the beta(2)-integrins CD11a-c was seen. This selective modulation of cell adhesion molecules (CAM) was seen on T cells and NK cells, as well. The selective modulation of CAM may help explain some of the clinical effects observed after bromelain treatment in patients suffering from chronic inflammatory disease, HIV and cancer.


Assuntos
Bromelaínas/farmacologia , Moléculas de Adesão Celular/metabolismo , Linfócitos/efeitos dos fármacos , Linfócitos/metabolismo , Adulto , Antígenos CD18/metabolismo , Complexo CD3/metabolismo , Antígeno CD56/metabolismo , Humanos , Receptores de Hialuronatos/metabolismo , Técnicas In Vitro , Subpopulações de Linfócitos/efeitos dos fármacos , Subpopulações de Linfócitos/imunologia , Subpopulações de Linfócitos/metabolismo , Receptores de IgG/metabolismo
17.
Cytometry ; 14(3): 265-70, 1993.
Artigo em Inglês | MEDLINE | ID: mdl-7682492

RESUMO

Flow cytometry has been used to evaluate several techniques for introducing macromolecules into large numbers of living cells. One technique is cell fusion with red blood cell ghosts loaded with a fluorescent reporter molecule (RBCF). The second technique, termed osmotic lysis of pinosomes (OLP), involves a brief exposure of cells to a hypertonic solution containing the reporter molecule; subsequently, a hypotonic media is added which lyses the pinosomes formed during the hypertonic treatment. A third technique, scrape loading (SL), involves the creation of transient holes in the cell membrane through the application of mechanical forces, which allows for the passage of reporter molecules into cells. A comparison of these techniques is presented here. OLP appears to offer several advantages: It is a simple procedure, virtually all cells are fluorescently labelled, and it is capable of loading larger amounts of material more uniformly into cells while maintaining excellent viability.


Assuntos
Fusão Celular , Membrana Eritrocítica , Fusão de Membrana , Dextranos , Eritrócitos , Estudos de Avaliação como Assunto , Citometria de Fluxo/métodos , Corantes Fluorescentes , Soluções Hipertônicas , Rodaminas
19.
Radiat Res ; 124(1 Suppl): S80-7, 1990 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-2236515

RESUMO

The effect of polyamine depletion on the survival response of human lung carcinoma cells (A-549) to acute heating at 45 degrees C and its effect on the induction and decay of thermotolerance were investigated in exponential and plateau-phase cells. A 48-h exposure to 1 mM alpha-difluoromethylornithine (DFMO), an irreversible inhibitor of ornithine decarboxylase, was used to deplete intracellular levels of putrescine and spermidine. Inhibition of polyamine synthesis had no effect on the survival of exponential cells to heating at 45 degrees C, but slightly enhanced the killing of slowly proliferating plateau-phase cells. While DFMO treatment did not inhibit the development of thermotolerance, it caused a reduction in the thermotolerance ratio of exponential cells from 2.6 to 1.80, and from 1.66 to 1.59 in plateau-phase cells. DFMO caused thermotolerance to decay more rapidly in polyamine-depleted cells as well. Flow cytometry demonstrated that DFMO did not alter the cell cycle distribution of plateau-phase cells (i.e., greater than 73% in G1/G0), but caused a block and time-dependent accumulation of exponential cells in G1/G0. The cytostatic properties of DFMO in exponential cells which favor its use with phase-specific agents, and its ability to alter the magnitude and decay of thermotolerance in human carcinoma cells suggest a potential role for this nontoxic agent in clinically oriented hyperthermia studies.


Assuntos
Temperatura Alta , Poliaminas/metabolismo , Células Tumorais Cultivadas/fisiologia , Aclimatação/fisiologia , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Eflornitina/farmacologia , Humanos , Técnicas In Vitro , Neoplasias Pulmonares , Inibidores da Ornitina Descarboxilase , Fatores de Tempo
20.
J Mol Cell Immunol ; 4(5): 255-66; discussion 266-8, 1990.
Artigo em Inglês | MEDLINE | ID: mdl-2169757

RESUMO

The antibody response to protein antigens requires specific cooperation between B and T cells. In order to deliver the helper signal, T cells must recognize, in the context of Class II MHC, processed antigen on the membrane of B cells. Processed antigen is in the form of peptides bound in a given site of the Class II MHC molecule; in order to address the question of where, in the B cell, the complex of Class II MHC and processed antigen is formed, we studied the subcellular localization of these two molecules. Since the formation of this complex is the crucial step in antigen processing and presentation, the answer to this question is central to the whole problem of the physiology of antigen handling by B cells. To collect information pertinent to the question, we have compared, in B cells, the intracellular traffic of Class II MHC and of monovalent and divalent anti-immunoglobulin antibodies used as protein ligands of the membrane immunoglobulins. We have done so by two-color immunofluorescence microscopy, and we have detected extensive confluence of Class II MHC molecules with the immunoglobulin ligand, both mono- and bi-valent, in the endosomes of LPS-activated murine B cells. Whereas the ligand clearly reaches the endosomes by internalization from the cell membrane, the Class II MHC molecules could reach the same location either by endocytosis from the membrane or through targeting to the endosomes of newly synthesized Class II MHC molecules. We have collected quantitative evidence for endocytosis of Class II MHC by following, with the fluorescence activated cell sorter, the quenching of the fluorescence of fluoresceinated Fab' anti Class II MHC in LPS-activated murine B cells; this quenching indicates the entry of the label into an acidic intracellular compartment. Together with the results of others, obtained with different methods, our observations support the concept that, at least in mature activated B cells, Class II MHC molecules reach the organelles where they meet processed protein antigens, mainly through the endocytic route. Since activated B cells endocytose their membrane Class II MHC, and not their membrane Class I, our results contribute to the understanding of how B cells present antigens, that have bound to their membrane immunoglobulins, to Class II-restricted helper T cells and not to Class I-restricted cytolytic T cells.


Assuntos
Linfócitos B/imunologia , Endocitose/imunologia , Antígenos de Histocompatibilidade Classe II/imunologia , Ativação Linfocitária/imunologia , Animais , Anticorpos Monoclonais/imunologia , Células Cultivadas , Cloroquina/farmacologia , Citosol/imunologia , Endocitose/efeitos dos fármacos , Citometria de Fluxo , Concentração de Íons de Hidrogênio , Fragmentos Fab das Imunoglobulinas/imunologia , Cinética , Lipopolissacarídeos/farmacologia , Ativação Linfocitária/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos A , Camundongos Endogâmicos BALB C , Microscopia de Fluorescência , Baço/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA