Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Neuroreport ; 32(17): 1379-1387, 2021 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-34718250

RESUMO

OBJECTIVES: Paeoniflorin, an active component of Radix Paeoniae Alba, has a neuroprotective effect in Parkinson's animal models. However, its mechanism of action remains to be determined. METHODS: In this study, we hypothesized that the neuroprotective effect of paeoniflorin occurs through the α-synuclein/protein kinase C δ subtype (PKC-δ) signaling pathway. We tested our hypothesis in the 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine (MPTP)-induced mouse model of Parkinson's disease. We evaluated the effects of paeoniflorin on the expression levels of signal components of the α-synuclein/PKC-δ pathway, cellular apoptosis and motor performance. RESULTS: Our results demonstrated that paeoniflorin restored the motor performance impairment caused by MPTP, inhibited apoptosis, and protected the ultrastructure of neurons. Paeoniflorin treatment also resulted in the dose-dependent upregulation of an antiapoptotic protein, B-cell lymphoma-2, at the mRNA and protein levels, similar to the effects of the positive control, selegiline. In contrast, paeoniflorin treatment downregulated the expression of pro-apoptotic proteins BCL2-Associated X2, α-synuclein, and PKC-δ at the mRNA and protein levels, as well as the level of the activated form of nuclear factor kappa B (p-NF-κB p65). CONCLUSIONS: Thus, our results showed that paeoniflorin exerts its neuroprotective effect by regulating the α-synuclein/PKC-δ signaling pathway to reduce neuronal apoptosis.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Apoptose/efeitos dos fármacos , Glucosídeos/farmacologia , Monoterpenos/farmacologia , Transtornos Parkinsonianos/metabolismo , Proteína Quinase C-delta/efeitos dos fármacos , Substância Negra/efeitos dos fármacos , alfa-Sinucleína/efeitos dos fármacos , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina , Animais , Anexina A5/efeitos dos fármacos , Anexina A5/metabolismo , Antiparkinsonianos/farmacologia , Modelos Animais de Doenças , Camundongos , Microscopia Eletrônica de Transmissão , Neurotoxinas , Transtornos Parkinsonianos/patologia , Transtornos Parkinsonianos/fisiopatologia , Proteína Quinase C-delta/metabolismo , Teste de Desempenho do Rota-Rod , Selegilina/farmacologia , Substância Negra/metabolismo , Substância Negra/patologia , alfa-Sinucleína/metabolismo
2.
Neurotox Res ; 38(2): 385-397, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32378056

RESUMO

Tramadol is a synthetic analogue of codeine that is often prescribed for the treatment of mild to moderate pains. It has a number of side effects including emotional instability and anxiety. In this study, we focus on the structural and functional changes of prefrontal cortex under chronic exposure to tramadol. At the cellular level, the amounts of ROS and annexin V in PC12 cells were evidently increased upon exposure to tramadol (at a concentration of 600 µM for 48 h). To this end, the rats were daily treated with tramadol at doses of 50 mg/kg for 3 weeks. Our findings reveal that tramadol provokes atrophy and apoptosis by the induction of apoptotic markers such as Caspase 3 and 8, pro-inflammatory markers, and downregulation of GDNF. Moreover, it triggers microgliosis and astrogliosis along with neuronal death in the prefrontal cortex. Behavioral disturbance and cognitive impairment are other side effects of tramadol. Overall, our results indicate tramadol-induced neurodegeneration in the prefrontal cortex mainly through activation of neuroinflammatory response.


Assuntos
Apoptose/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Córtex Pré-Frontal/efeitos dos fármacos , Tramadol/toxicidade , Animais , Anexina A5/efeitos dos fármacos , Anexina A5/metabolismo , Astrócitos/efeitos dos fármacos , Astrócitos/patologia , Atrofia , Caspase 3/efeitos dos fármacos , Caspase 3/metabolismo , Caspase 8/efeitos dos fármacos , Caspase 8/metabolismo , Morte Celular , Fator Neurotrófico Derivado de Linhagem de Célula Glial/efeitos dos fármacos , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Gliose/induzido quimicamente , Gliose/metabolismo , Gliose/patologia , Masculino , Microglia/efeitos dos fármacos , Microglia/patologia , Neurônios/metabolismo , Neurônios/patologia , Células PC12 , Córtex Pré-Frontal/metabolismo , Córtex Pré-Frontal/patologia , Ratos , Espécies Reativas de Oxigênio/metabolismo
3.
Cancer ; 123(9): 1662-1673, 2017 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-28055106

RESUMO

BACKGROUND: Nucleophosmin 1 (NPM1) is a nucleocytoplasmic shuttling protein mainly localized in the nucleolus. NPM1 is frequently mutated in acute myeloid leukemia (AML). NPM1c oligomerizes with wild-type nucleophosmin 1 (wt-NPM1), and this leads to its continuous cytoplasmic delocalization and contributes to leukemogenesis. Recent studies have shown that Cytoplasmic NPM1 (NPM1c) degradation leads to growth arrest and apoptosis of NPM1c AML cells and corrects wt-NPM1 normal nucleolar localization. METHODS: AML cells expressing wt-NPM1 or NPM1c or transfected with wt-NPM1 or NPM1c as well as wt-NPM1 and NPM1c AML xenograft mice were used. Cell growth was assessed with trypan blue or a CellTiter 96 proliferation kit. The cell cycle was studied with a propidium iodide (PI) assay. Caspase-mediated intrinsic apoptosis was assessed with annexin V/PI, the mitochondrial membrane potential, and poly(adenosine diphosphate ribose) polymerase cleavage. The expression of NPM1, p53, phosphorylated p53, and p21 was analyzed via immunoblotting. Localization was performed with confocal microscopy. The leukemia burden was evaluated by flow cytometry with an anti-human CD45 antibody. RESULTS: The imidazoquinoxaline 1-(3-methoxyphenyl)-N-methylimidazo[1,2-a]quinoxalin-4-amine (EAPB0503) induced selective proteasome-mediated degradation of NPM1c, restored wt-NPM1 nucleolar localization in NPM1c AML cells, and thus yielded selective growth arrest and apoptosis. Introducing NPM1c to cells normally harboring wt-NPM1 sensitized them to EAPB0503 and led to their growth arrest. Moreover, EAPB0503 selectively reduced the leukemia burden in NPM1c AML xenograft mice. CONCLUSIONS: These findings further reinforce the idea of targeting the NPM1c oncoprotein to eradicate leukemic cells and warrant a broader preclinical evaluation and then a clinical evaluation of this promising drug. Cancer 2017;123:1662-1673. © 2017 American Cancer Society.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Leucemia Mieloide Aguda/tratamento farmacológico , Proteínas Mutantes/efeitos dos fármacos , Proteínas Nucleares/efeitos dos fármacos , Quinoxalinas/farmacologia , Animais , Anexina A5/efeitos dos fármacos , Anexina A5/metabolismo , Linhagem Celular Tumoral , Nucléolo Celular/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/efeitos dos fármacos , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Citoplasma/metabolismo , Citometria de Fluxo , Humanos , Immunoblotting , Leucemia Mieloide Aguda/genética , Camundongos , Microscopia Confocal , Proteínas Mutantes/metabolismo , Mutação , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Nucleofosmina , Fosforilação/efeitos dos fármacos , Poli(ADP-Ribose) Polimerases/efeitos dos fármacos , Proteína Supressora de Tumor p53/efeitos dos fármacos , Proteína Supressora de Tumor p53/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Asian Pac J Cancer Prev ; 13(4): 1395-9, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22799338

RESUMO

BACKGROUND: Histone deacetylase (HDAC) inhibitors have been reported to induce cell growth arrest, apoptosis and differentiation of tumor cells. The present study aimed to examine the effects of trichostatin A (TSA), one such inhibitor, on the cell cycle, apoptosis and invasiveness of osteosarcoma cells. METHODS: MG- 63 cells were treated with TSA at various concentrations. Then, cell growth and apoptosis were determined by 3-(4, 5-dimethyl-2-thiazolyl)-2H-tetrazolium bromide (MTT) and TUNEL assays, respectively; cell cycling was assessed by flow cytometry; invasion assays were performed with the transwell Boyden Chamber system. RESULTS: MTT assays revealed that TSA significantly inhibited the growth of MG-63 cells in a concentration and time dependent manner. TSA treated cells demonstrated morphological changes indicative of apoptosis and TUNEL assays revealed increased apoptosis of MG-63 cells after TSA treatment. Flow cytometry showed that TSA arrested the cell cycle in G1/G2 phase and annexin V positive apoptotic cells increased markedly. In addition, the invasiveness of MG-63 cells was inhibited by TSA in a concentration dependent manner. CONCLUSION: Our findings demonstrate that TSA inhibits the proliferation, induces apoptosis and inhibits invasiveness of osteosarcoma cells in vitro. HDAC inhibitors may thus have promise to become new therapeutic agents against osteosarcoma.


Assuntos
Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Pontos de Checagem da Fase G1 do Ciclo Celular/efeitos dos fármacos , Inibidores de Histona Desacetilases/farmacologia , Ácidos Hidroxâmicos/farmacologia , Osteossarcoma/patologia , Anexina A5/efeitos dos fármacos , Anexina A5/metabolismo , Linhagem Celular Tumoral , Humanos , Invasividade Neoplásica , Osteossarcoma/tratamento farmacológico , Osteossarcoma/metabolismo
5.
Oral Dis ; 18(3): 280-4, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22093110

RESUMO

OBJECTIVE: To determine the effects of fluorides on endothelial functioning. MATERIALS AND METHODS: We analyzed expressions of adhesion molecules, ICAM-1 and ICAM-3, and annexin V, on the surface of human umbilical vein endothelial cells (HUVECs) exposed to various concentrations of NaF and SnF(2) . We compared the effects of fluoride-induced changes with those obtained when stimulating HUVECs with TNF-α and verified whether N-acetyl cysteine (NAC), well-known antioxidant, can prevent both fluoride- and TNF-α-induced alterations. RESULTS: The expressions of annexin V and ICAM-1 increased significantly after adding NaF (5.0 or 7.5mM) or Sn(2) F (0.5 or 0.75mM) to the culture medium. Pre-incubating HUVECs with NAC prevented the effects induced by 5.0 mM of NaF and 0.5 mM of Sn(2) F. Only the highest concentration of NaF (7.5mM) triggered the expression of ICAM-3. The expressions of all three molecules increased significantly upon stimulating the cultures with TNF-α (20ng ml(-1) ); these changes were not reversed by pre-incubation with NAC. CONCLUSIONS: Fluorides induce oxidative stress, resulting in apoptosis and activation of HUVECs, manifested by an elevated expression of ICAM-1. The oxidative stress resulting from a stimulation by the highest NaF concentration triggers ICAM-3 expression on the HUVECs' surface.


Assuntos
Apoptose/efeitos dos fármacos , Cariostáticos/farmacologia , Células Endoteliais/efeitos dos fármacos , Endotélio Vascular/efeitos dos fármacos , Fluoretos/farmacologia , Acetilcisteína/farmacologia , Anexina A5/efeitos dos fármacos , Antígenos CD/efeitos dos fármacos , Antioxidantes/farmacologia , Moléculas de Adesão Celular/efeitos dos fármacos , Técnicas de Cultura de Células , Células Cultivadas , Endotélio Vascular/citologia , Inibidores Enzimáticos/farmacologia , Sequestradores de Radicais Livres/farmacologia , Humanos , Molécula 1 de Adesão Intercelular/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Fluoreto de Sódio/farmacologia , Fluoretos de Estanho/farmacologia , Fator de Necrose Tumoral alfa/farmacologia , Veias Umbilicais/citologia
6.
Biochim Biophys Acta ; 1800(4): 439-47, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20064588

RESUMO

BACKGROUND: Plagiochin E (PLE) is an antifungal active macrocyclic bis(bibenzyl) isolated from liverwort Marchantia polymorpha L. To elucidate the mechanism of action, previous studies revealed that the antifungal effect of PLE was associated with the accumulation of ROS, an important regulator of apoptosis in Candida albicans. The present study was designed to find whether PLE caused apoptosis in C. albicans. METHODS: We assayed the cell cycle by flow cytometry using PI staining, observed the ultrastructure by transmission electron microscopy, studied the nuclear fragmentation by DAPI staining, and investigated the exposure of phosphatidylserine at the outer layer of the cytoplasmic membrane by the FITC-annexin V staining. The effect of PLE on expression of CDC28, CLB2, and CLB4 was determined by RT-PCR. Besides, the activity of metacaspase was detected by FITC-VAD-FMK staining, and the release of cytochrome c from mitochondria was also determined. Furthermore, the effect of antioxidant L-cysteine on PLE-induced apoptosis in C. albicans was also investigated. RESULTS: Cells treated with PLE showed typical markers of apoptosis: G(2)/M cell cycle arrest, chromatin condensation, nuclear fragmentation, and phosphatidylserine exposure. The expression of CDC28, CLB2, and CLB4 was down-regulated by PLE, which may contribute to PLE-induced G(2)/M cell cycle arrest. Besides, PLE promoted the cytochrome c release and activated the metacaspase, which resulted in the yeast apoptosis. The addition of L-cysteine prevented PLE-induced nuclear fragmentation, phosphatidylserine exposure, and metacaspase activation, indicating the ROS was an important mediator of PLE-induced apoptosis. CONCLUSIONS: PLE induced apoptosis in C. albicans through a metacaspase-dependent apoptotic pathway. GENERAL SIGNIFICANCE: In this study, we reported for the first time that PLE induced apoptosis in C. albicans through activating the metacaspase. These results would conduce to elucidate its underlying antifungal mechanism.


Assuntos
Antifúngicos/farmacologia , Hidrocarbonetos Aromáticos com Pontes/farmacologia , Candida albicans/efeitos dos fármacos , Compostos Macrocíclicos/farmacologia , Estilbenos/farmacologia , Anexina A5/efeitos dos fármacos , Anexina A5/metabolismo , Antifúngicos/isolamento & purificação , Apoptose/efeitos dos fármacos , Hidrocarbonetos Aromáticos com Pontes/química , Hidrocarbonetos Aromáticos com Pontes/isolamento & purificação , Candida albicans/citologia , Candida albicans/enzimologia , Candida albicans/genética , Inibidores de Caspase , Ciclo Celular/efeitos dos fármacos , Primers do DNA , Citometria de Fluxo , Compostos Macrocíclicos/química , Compostos Macrocíclicos/isolamento & purificação , Marchantia/química , Modelos Moleculares , Conformação Molecular , Inibidores de Proteases/farmacologia , RNA Fúngico/genética , RNA Fúngico/isolamento & purificação , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Estilbenos/química , Estilbenos/isolamento & purificação
7.
J Oral Pathol Med ; 38(10): 792-800, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19656269

RESUMO

BACKGROUND: Although N(1)-guanyl-1,7,-diamineoheptane (GC7), an inhibitor of deoxyhypusine synthase, has been shown to inhibit cell growth, the mechanism of its action is not completely understood. In this study, we investigated the mechanisms of the effects of GC7 on cell growth, differentiation and apoptosis in relation to adenosine monophosphate-activated protein kinase (AMPK) activation, as AMPK is known to be a possible target for cancer treatment. METHODS: The effects of GC7 on the growth of immortalized human oral keratinocytes (IHOK) and primary oral cancer cells (HN4), was investigated using MTT assay, Western blotting, cell cycle analysis, DNA fragmentation and expression of apoptotic pathway proteins. RESULTS: N(1)-guanyl-1,7,-diamineoheptane inhibited cell proliferation in a time- and dose-dependent manner in IHOK and HN4 cells. GC7 treatment decreased the expression of differentiation markers, such as involucrin, CK13 and CK19. The major mechanism of growth inhibition by GC7 treatment was induction of apoptosis, which is supported by sub-G(1) phase arrest, annexin V-FITC staining and DNA fragmentation analysis. GC7 treatment increased the cytosolic level of cytochrome c and resulted in caspase-3 activation. GC7 treatment also resulted in a strong activation of AMPK. Furthermore, specific AMPK activator blocked the GC7-induced growth inhibition effect, as well as apoptosis. CONCLUSION: These results demonstrate that GC7 blocks immortalized and malignant keratinocyte cell proliferation and differentiation by inducing apoptosis through the mitochondrial and AMPK pathways. On the basis of these observations, we propose that a strategy combining GC7 and AMPK inhibition could be developed into a novel chemotherapeutic modality in oral cancer.


Assuntos
Proteínas Quinases Ativadas por AMP/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Carcinoma/patologia , Inibidores Enzimáticos/farmacologia , Guanina/análogos & derivados , Queratinócitos/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Mucosa Bucal/efeitos dos fármacos , Neoplasias Bucais/patologia , Oxirredutases atuantes sobre Doadores de Grupo CH-NH/antagonistas & inibidores , Anexina A5/efeitos dos fármacos , Caspase 3/efeitos dos fármacos , Proteínas de Ciclo Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Citocromos c/efeitos dos fármacos , Fragmentação do DNA , Relação Dose-Resposta a Droga , Ativação Enzimática , Inibidores Enzimáticos/administração & dosagem , Fase G1/efeitos dos fármacos , Guanina/administração & dosagem , Guanina/farmacologia , Humanos , Queratina-13/antagonistas & inibidores , Queratina-19/antagonistas & inibidores , Mucosa Bucal/patologia , Precursores de Proteínas/antagonistas & inibidores
8.
Brain Res ; 1283: 155-66, 2009 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-19505441

RESUMO

Neuroblastoma is a pediatric extracranial tumor and a major cause of death in children under age 2. Conventional therapy shows inefficacy in most cases and thus development of new therapeutic strategies is urgently needed. We explored the efficacy of combination of the small molecule Bcl-2 inhibitor HA14-1 (HA) and the isoflavonoid genistein (GST) in human malignant neuroblastoma SK-N-BE2 and SH-SY5Y cells. Combination of 10 microM HA and 250 microM GST was optimal for SK-N-BE2 cells and combination of 5 microM HA and 100 microM GST was optimal for SH-SY5Y cells for induction of apoptosis. Phase-contrast microscopy and Wright staining showed morphological features of apoptosis. Cell cycle analysis and Annexin V-FITC/PI binding assay showed that combination of HA and GST was more effective in inducing apoptosis in both cell lines than either HA or GST alone. Western blotting showed that combination of HA and GST caused upregulation of Bax and down regulation of Bcl-2 resulting in increased Bax:Bcl-2 ratio and mitochondrial release of cytochrome c, Smac, and AIF. Down regulation of survival factors such as NF-kappaB, N-Myc, and survivin promoted apoptosis. Activation of caspase-8, calpain, and caspase-3 occurred in course of apoptosis. Increased calpain and caspase-3 activities were confirmed in the degradation of alpha-spectrin to 145 kD spectrin break down product (SBDP) and 120 kD SBDP, respectively. Thus, combination of HA and GST could serve as a promising therapeutic strategy for increasing apoptosis in different human malignant neuroblastoma cells.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Proteínas Reguladoras de Apoptose/antagonistas & inibidores , Benzopiranos/farmacologia , Cisteína Endopeptidases/efeitos dos fármacos , Genisteína/farmacologia , Neuroblastoma/tratamento farmacológico , Nitrilas/farmacologia , Anexina A5/efeitos dos fármacos , Anexina A5/metabolismo , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Proteínas Reguladoras de Apoptose/metabolismo , Benzopiranos/uso terapêutico , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/fisiologia , Cisteína Endopeptidases/metabolismo , Relação Dose-Resposta a Droga , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/uso terapêutico , Genisteína/uso terapêutico , Humanos , Neuroblastoma/metabolismo , Neuroblastoma/fisiopatologia , Nitrilas/uso terapêutico , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico
9.
Pediatr Allergy Immunol ; 20(4): 392-8, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18713315

RESUMO

Decreased severity of graft-versus-host disease after mismatched umbilical cord blood (UCB) transplantation may be attributed in part to the increased propensity to apoptosis of UCB T cells following activation. Interleukin (IL)-15, a pleiotropic cytokine that is essential for T-cell proliferation and survival, may serve as promising immunomodulative therapy post-CB transplantation for its anti-apoptotic effect. This study aimed to determine the kinetics of Fas or tumor necrosis factor-alpha receptor (TNFR) mediated caspase-3 expression and apoptosis of anti-CD3/anti-CD28 activated UCB T cells in the influence of IL-15. Activated caspase-3 expression was analyzed by Western blotting and the percentage of apoptotic cells was determined by annexin-V/propidium iodide (PI) flow cytometric staining. Significant expression of Fas and TNFR2 was detected on anti-CD3/anti-CD28 pre-activated UCB T cells. These cells were susceptible to anti-Fas but not TNF-alpha-induced apoptosis. Kinetic study shows that caspase-3 expression became evident at 6th-8th h following anti-Fas stimulation, while early apoptotic cells with annexin-V(+)/PI(-) expression appeared at 12th-16th h. IL-15, though successful in decreasing apoptosis in pre-activated UCB T cells, failed to completely prevent Fas-mediated caspase-3 expression and apoptosis of CB T cells. The pre-activated UCB and adult peripheral blood T cells behaved similarly with regard to death receptor expression, caspase-3 expression and apoptosis upon Fas-engagement. Although IL-15 promotes overall activated UCB T-cell survival, it did not particularly prevent Fas-mediated activation-induced cell death.


Assuntos
Apoptose/imunologia , Sangue Fetal/imunologia , Linfócitos T/imunologia , Receptor fas/imunologia , Anexina A5/efeitos dos fármacos , Anexina A5/imunologia , Anticorpos/imunologia , Antígenos CD28/imunologia , Complexo CD3/imunologia , Caspase 3/efeitos dos fármacos , Caspase 3/metabolismo , Feminino , Humanos , Recém-Nascido , Interleucina-15/farmacologia , Ativação Linfocitária , Masculino , Linfócitos T/efeitos dos fármacos , Fator de Necrose Tumoral alfa/farmacologia
10.
Clin Cancer Res ; 14(10): 3113-23, 2008 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-18483379

RESUMO

PURPOSE: To evaluate the potential utility of histone deacetylase inhibitors (HDACi) for treatment of retinoblastoma (RB). EXPERIMENTAL DESIGN: Growth-inhibitory effects of HDACi [trichostatin A (TSA), suberoylanilide hydroxamic acid (SAHA), or MS-275] were assessed in human and transgenic murine RB cells. Effects of TSA and MS-275 were also assessed in combination with standard therapeutic agents for RB. Proapoptotic effects of MS-275 and TSA were evaluated by caspase-3/7 activity, Annexin V translocation, and/or Bim expression analyses. Effects of MS-275 on cell cycle distribution and reactive oxygen species levels were determined by flow cytometry. Retinal tissue morphology was evaluated in mice after local administration of MS-275. Analysis of retinal acetyl-histone levels was used to assess MS-275 delivery after systemic administration. Therapeutic effects of MS-275 were determined in transgenic mouse and rat ocular xenograft models of RB after i.p. injection of 20 mg/kg every other day for 21 or 13 days, respectively. RESULTS: TSA, SAHA, and MS-275 dose dependently reduced RB cell survival. TSA and MS-275 showed additive growth-inhibitory effects in combination with carboplatin, etoposide, or vincristine. TSA and MS-275 increased caspase-3/7 activity. MS-275 increased Annexin V membrane translocation and induced G1 arrest. Cytotoxicity of MS-275 was dependent on increased reactive oxygen species levels and was reversed by antioxidant pretreatment. Intraocular administration of 1 microL of 10 micromol/L MS-275 did not alter ocular tissue morphology. Increased acetyl-histone levels confirmed MS-275 delivery to retinal tissue after systemic administration. MS-275 significantly reduced tumor burden in both mouse and rat models of RB. CONCLUSIONS: HDACi should be considered for clinical trials in children with RB.


Assuntos
Antineoplásicos/farmacologia , Inibidores Enzimáticos/farmacologia , Inibidores de Histona Desacetilases , Neoplasias da Retina/tratamento farmacológico , Retinoblastoma/tratamento farmacológico , Animais , Anexina A5/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Benzamidas/farmacologia , Western Blotting , Caspase 3/efeitos dos fármacos , Caspase 7/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Citometria de Fluxo , Humanos , Ácidos Hidroxâmicos/farmacologia , Técnicas In Vitro , Camundongos , Camundongos Transgênicos , Reação em Cadeia da Polimerase , Piridinas/farmacologia , Ratos , Espécies Reativas de Oxigênio/metabolismo , Vorinostat , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Blood ; 111(4): 1951-61, 2008 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-18024794

RESUMO

Adhesion of leukemic cells to vascular cells may confer resistance to chemotherapeutic agents. We hypothesized that disruption of leukemic cell cytoskeletal stability and interference with vascular cell interactions would promote leukemic cell death. We demonstrate that low and nontoxic doses of microtubule-destabilizing agent combretastatin-A4-phosphate (CA4P) inhibit leukemic cell proliferation in vitro and induce mitotic arrest and cell death. Treatment of acute myeloid leukemias (AMLs) with CA4P leads to disruption of mitochondrial membrane potential, release of proapoptotic mitochondrial membrane proteins, and DNA fragmentation, resulting in cell death in part through a caspase-dependent manner. Furthermore, CA4P increases intracellular reactive oxygen species (ROS), and antioxidant treatment imparts partial protection from cell death, suggesting that ROS accumulation contributes to CA4P-induced cytotoxicity in AML. In vivo, CA4P inhibited proliferation and circulation of leukemic cells and diminished the extent of perivascular leukemic infiltrates, prolonging survival of mice that underwent xenotransplantation without inducing hematologic toxicity. CA4P decreases the interaction of leukemic cells with neovessels by down-regulating the expression of the adhesion molecule VCAM-1 thereby augmenting leukemic cell death. These data suggest that CA4P targets both circulating and vascular-adherent leukemic cells through mitochondrial damage and down-regulation of VCAM-1 without incurring hematologic toxicities. As such, CA4P provides for an effective means to treat refractory organ-infiltrating leukemias.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Morte Celular/efeitos dos fármacos , Leucemia Mieloide Aguda/tratamento farmacológico , Mitocôndrias/fisiologia , Estilbenos/farmacologia , Anexina A5/efeitos dos fármacos , Anexina A5/metabolismo , Apoptose/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Dano ao DNA/efeitos dos fármacos , Humanos , Leucemia Mieloide Aguda/patologia , Linfoma/tratamento farmacológico , Linfoma/patologia , Mitocôndrias/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Transplante Heterólogo , Células Tumorais Cultivadas
12.
Toxicol Appl Pharmacol ; 225(3): 318-28, 2007 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-17881028

RESUMO

Undecylprodigiosin (UP) is a bacterial bioactive metabolite produced by Streptomyces and Serratia. In this study, we explored the anticancer effect of UP. Human breast carcinoma cell lines BT-20, MCF-7, MDA-MB-231 and T47D and one nonmalignant human breast epithelial cell line, MCF-10A, were tested in this study. We found that UP exerted a potent cytotoxicity against all breast carcinoma cell lines in a dose- and time-dependent manner. In contrast, UP showed limited toxicity to MCF-10A cells, indicating UP's cytotoxic effect is selective for malignant cells. UP's cytotoxic effect was due to apoptosis, as confirmed by positive TUNEL signals, annexin V-binding, caspase 9 activation and PARP cleavage. Notably, UP-induced apoptosis was blocked by the pan-caspase inhibitor z-VAD.fmk, further indicating the involvement of caspase activity. Moreover, UP caused a marked decrease of the levels of antiapoptotic BCL-X(L), Survivin and XIAP while enhancing the levels of proapoptotic BIK, BIM, MCL-1S and NOXA, consequently favoring induction of apoptosis. Additionally, we found that cells with functional p53 (MCF-7, T47D) or mutant p53 (BT-20, MDA-MB-231) were both susceptible to UP's cytotoxicity. Importantly, UP was able to induce apoptosis in MCF-7 cells with p53 knockdown by RNA interference, confirming the dispensability of p53 in UP-induced apoptosis. Overall, our results establish that UP induces p53-independent apoptosis in breast carcinoma cells with no marked toxicity to nonmalignant cells, raising the possibility of its use as a new chemotherapeutic drug for breast cancer irrespective of p53 status.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias da Mama/tratamento farmacológico , Proteína Supressora de Tumor p53/metabolismo , Anexina A5/efeitos dos fármacos , Anexina A5/metabolismo , Antineoplásicos/administração & dosagem , Proteínas Reguladoras de Apoptose/metabolismo , Proteína 11 Semelhante a Bcl-2 , Caspase 9/efeitos dos fármacos , Caspase 9/metabolismo , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Feminino , Humanos , Marcação In Situ das Extremidades Cortadas , Proteínas Inibidoras de Apoptose , Proteínas de Membrana/metabolismo , Proteínas Associadas aos Microtúbulos/efeitos dos fármacos , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas Mitocondriais , Proteína de Sequência 1 de Leucemia de Células Mieloides , Proteínas de Neoplasias/efeitos dos fármacos , Proteínas de Neoplasias/metabolismo , Poli(ADP-Ribose) Polimerases/efeitos dos fármacos , Poli(ADP-Ribose) Polimerases/metabolismo , Prodigiosina/administração & dosagem , Prodigiosina/análogos & derivados , Prodigiosina/farmacologia , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Serratia marcescens/química , Survivina , Fatores de Tempo , Proteína Supressora de Tumor p53/efeitos dos fármacos , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/efeitos dos fármacos , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/metabolismo , Proteína bcl-X/efeitos dos fármacos , Proteína bcl-X/metabolismo
13.
BMC Cancer ; 7: 237, 2007 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-18166137

RESUMO

BACKGROUND: Medicinal plant is a main source of cancer drug development. Some of the cycloartane triterpenoids isolated from the aerial part of Cimicifuga dahurica showed cytotoxicity in several cancer cell lines. It is of great interest to examine the antiproliferative activity and mechanisms of total triterpenoid glycosides of C. dahurica and therefore might eventually be useful in the prevention or treatment of Hepatoma. METHODS: The total glycosides from the aerial part (TGA) was extracted and its cytotoxicity was evaluated in HepG2 cells and primary cultured normal mouse hepatocytes by an MTT assay. Morphology observation, Annexin V-FITC/PI staining, cell cycle analysis and western blot were used to further elucidate the cytotoxic mechanism of TGA. Implanted mouse H22 hepatoma model was used to demonstrate the tumor growth inhibitory activity of TGA in vivo. RESULTS: The IC50 values of TGA in HepG2 and primary cultured normal mouse hepatocytes were 21 and 105 mug/ml, respectively. TGA induced G0/G1 cell cycle arrest at lower concentration (25 mug/ml), and triggered G2/M arrest and apoptosis at higher concentrations (50 and 100 mug/ml respectively). An increase in the ratio of Bax/Bcl-2 was implicated in TGA-induced apoptosis. In addition, TGA inhibited the growth of the implanted mouse H22 tumor in a dose-dependent manner. CONCLUSION: TGA may potentially find use as a new therapy for the treatment of hepatoma.


Assuntos
Cimicifuga/química , Glicosídeos/farmacologia , Hepatoblastoma/tratamento farmacológico , Hepatoblastoma/patologia , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/patologia , Extratos Vegetais/farmacologia , Animais , Anexina A5/efeitos dos fármacos , Anexina A5/metabolismo , Antineoplásicos Fitogênicos/farmacologia , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta a Droga , Glicosídeos/química , Glicosídeos/isolamento & purificação , Hepatoblastoma/metabolismo , Humanos , Concentração Inibidora 50 , Neoplasias Hepáticas/metabolismo , Camundongos , Camundongos Endogâmicos ICR , Transplante de Neoplasias , Extratos Vegetais/química , Extratos Vegetais/isolamento & purificação , Proteínas Proto-Oncogênicas c-bcl-2/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Células Tumorais Cultivadas , Proteína X Associada a bcl-2/efeitos dos fármacos , Proteína X Associada a bcl-2/metabolismo
14.
Cell Physiol Biochem ; 17(5-6): 201-10, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16790996

RESUMO

Prostaglandin-E2 (PGE2) is known to trigger suicidal death of nucleated cells (apoptosis) and enucleated erythrocytes (eryptosis). In erythrocytes PGE2 induced suicidal cell death involves activation of nonselective cation channels leading to Ca2+ entry followed by cell shrinkage and triggering of Ca2+ sensitive cell membrane scrambling with phosphatidylserine (PS) exposure at the cell surface. The present study was performed to explore whether PGE2 induces apoptosis of nucleated cells similarly through cation channel activation and to possibly disclose the molecular identity of the cation channels involved. To this end, Ca2+ activity was estimated from Fluo3 fluorescence, mitochondrial potential from DePsipher fluorescence, phosphatidylserine exposure from annexin binding, caspase activation from caspAce fluorescence, cell volume from FACS forward scatter, and DNA fragmentation utilizing a photometric enzyme immunoassay. Stimulation of K562 human leukaemia cells with PGE2 (50 microM) increased cytosolic Ca2+ activity, decreased forward scatter, depolarized the mitochondrial potential, increased annexin binding, led to caspase activation and resulted in DNA fragmentation. Gene silencing of the Ca2+-permeable transient receptor potential cation channel TRPC7 significantly blunted PGE2-induced triggering of PS exposure and DNA fragmentation. In conclusion, K562 cells express Ca2+-permeable TRPC7 channels, which are activated by PGE2 and participate in the triggering of apoptosis.


Assuntos
Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Dinoprostona/farmacologia , Canais Iônicos Sensíveis a Ácido , Amilorida/análogos & derivados , Amilorida/farmacologia , Anexina A5/efeitos dos fármacos , Anexina A5/metabolismo , Cálcio/metabolismo , Caspases/efeitos dos fármacos , Caspases/metabolismo , Morte Celular/efeitos dos fármacos , Tamanho Celular/efeitos dos fármacos , Citosol/efeitos dos fármacos , Citosol/metabolismo , Fragmentação do DNA , Relação Dose-Resposta a Droga , Ativação Enzimática , Inativação Gênica , Humanos , Canais Iônicos/efeitos dos fármacos , Canais Iônicos/metabolismo , Células K562/efeitos dos fármacos , Potenciais da Membrana/efeitos dos fármacos , Proteínas de Membrana/efeitos dos fármacos , Proteínas de Membrana/genética , Mitocôndrias/efeitos dos fármacos , Proteínas do Tecido Nervoso/efeitos dos fármacos , Proteínas do Tecido Nervoso/genética , Fosfatidilserinas/metabolismo , Canais de Sódio/efeitos dos fármacos , Canais de Sódio/genética , Canais de Cátion TRPC/efeitos dos fármacos , Canais de Cátion TRPC/genética
15.
J Musculoskelet Neuronal Interact ; 4(1): 101-4, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15615083

RESUMO

Osteocyte viability may play a significant role in the maintenance and integrity of bone. Bone loss due to osteoporosis may be due in part to osteocyte cell death. We have identified a factor that will protect both osteoblasts and osteocytes from cell death due to agents known to be responsible for various forms of osteoporosis. Not only does estrogen preserve osteoblast and osteocyte viability, but so does a molecule called CD40Ligand. This molecule is expressed on activated T lymphocytes, human dendritic cells, and human vascular endothelial cells, whereas its receptor CD40 is expressed on normal epithelium, B cells, and dendritic cells. CD40Ligand protects osteoblasts and the MLO-Y4 osteocyte-like cells against apoptosis induced by glucocorticoids, tumor necrosis factor alpha or etoposide. As tumor necrosis factor a has been shown to be responsible for post-menopausal bone loss and glucocorticoids induce dramatic bone loss, this finding has important implications with regards to potential therapy for both post-menopausal and steroid-induced osteoporosis.


Assuntos
Desenvolvimento Ósseo/fisiologia , Ligante de CD40/fisiologia , Substâncias de Crescimento/fisiologia , Osteócitos/metabolismo , Osteoporose/tratamento farmacológico , Osteoporose/metabolismo , Animais , Anexina A5/efeitos dos fármacos , Anexina A5/metabolismo , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Desenvolvimento Ósseo/efeitos dos fármacos , Osso e Ossos/efeitos dos fármacos , Osso e Ossos/metabolismo , Ligante de CD40/metabolismo , Ligante de CD40/uso terapêutico , Morte Celular/efeitos dos fármacos , Morte Celular/fisiologia , Linhagem Celular , Estrogênios/metabolismo , Estrogênios/farmacologia , Estrogênios/uso terapêutico , Etoposídeo/efeitos adversos , Etoposídeo/antagonistas & inibidores , Glucocorticoides/efeitos adversos , Glucocorticoides/antagonistas & inibidores , Glucocorticoides/metabolismo , Substâncias de Crescimento/metabolismo , Substâncias de Crescimento/uso terapêutico , Camundongos , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Osteócitos/efeitos dos fármacos , Osteoporose/fisiopatologia , Azul Tripano , Fator de Necrose Tumoral alfa/efeitos adversos , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/metabolismo
16.
J Neurosci Res ; 76(6): 834-45, 2004 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-15160395

RESUMO

Multiple sclerosis is a chronic inflammatory disease of the central nervous system. Myelin and oligodendrocytes are considered the major targets of injury caused by a cell-mediated immune response. There is circumstantial evidence that proinflammatory cytokines like tumor necrosis factor alpha (TNF-alpha) and interferon gamma (IFN-gamma) could have disease-promoting roles in multiple sclerosis (MS). In the present study, the cytotoxic effects of IFN-gamma and TNF-alpha on the human oligodendroglial cell lines human oligodendroglioma (HOG) and MO3.13 were analyzed. When the oligodendroglial cell lines were cultured in the presence of IFN-gamma or TNF-alpha, apoptotic cell death was observed in both cell lines after >24 hr incubation. Apoptosis was evidenced by a decrease in cell viability, apoptotic changes in cell and nucleus morphology, and disruption of the membrane asymmetry. Our data show that TNF-alpha and IFN-gamma induce apoptosis in a dose-dependent fashion in both oligodendroglial cell lines and that their synergistic effect results in enhanced cell death. Understanding the regulation of cell death pathways in oligodendrocytes is critical for protecting myelin-producing cells and their associated axons during injury in patients with MS.


Assuntos
Apoptose/efeitos dos fármacos , Interferon gama/administração & dosagem , Oligodendroglia/patologia , Fator de Necrose Tumoral alfa/administração & dosagem , Anexina A5/efeitos dos fármacos , Células Cultivadas , Sinergismo Farmacológico , Humanos , Oligodendroglia/metabolismo , Oligodendroglia/ultraestrutura , Fosfatidilserinas/metabolismo
17.
J Neurosci Res ; 76(6): 846-61, 2004 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-15160396

RESUMO

Cytokines, such as interferon-gamma (IFN-gamma) and tumor necrosis factor-alpha (TNF-alpha), can initiate dual effects resulting in either cell growth or cell death. In this study, the human oligodendroglial cell lines HOG and MO3.13 were used as a model to study the molecular mechanisms of cytokine-induced cell death in human oligodendrocytes. We have previously shown that TNF-alpha and IFN-gamma induce apoptosis in both oligodendroglial cell lines within 72 hr. In the present study, the cell death pathways operating within these cells were further investigated at the gene expression level. Both cell lines express a broad repertoire of caspases and apoptosis-related genes. Some of these genes are specifically up-regulated by cytokine treatment; e.g., caspase-1 is up-regulated by IFN-gamma. In addition to direct cytotoxic effects, IFN-gamma and TNF-alpha also enhance the expression of Fas, TNFR1, and MHC class I molecules in both cell lines. This suggests that cytokines can make oligodendrocytes more vulnerable to different cell death pathways in an inflammatory environment. cDNA microarray analysis of the HOG cell line revealed that TNF-alpha induces genes that regulate apoptosis, survival, inflammation, cell metabolism, and cell signaling. The data suggest that oligodendroglial cells activate both death and survival pathways upon cytokine challenges. However, the survival pathways seem to be unable to compete with the death signal after more than 24 hr of cytokine treatment. These results may contribute to the development of therapeutic strategies aimed at interfering with cytokine-induced cell death of oligodendrocytes in patients with multiple sclerosis.


Assuntos
Apoptose/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Interferon gama/administração & dosagem , Peptídeos e Proteínas de Sinalização Intracelular , Oligodendroglia/patologia , Fator de Necrose Tumoral alfa/administração & dosagem , Proteínas Adaptadoras de Transdução de Sinal , Anexina A5/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Proteínas de Transporte/efeitos dos fármacos , Proteínas de Transporte/genética , Caspases/efeitos dos fármacos , Caspases/genética , Células Cultivadas , Proteínas Correpressoras , Sinergismo Farmacológico , Proteína Ligante Fas , Perfilação da Expressão Gênica , Humanos , Glicoproteínas de Membrana/efeitos dos fármacos , Glicoproteínas de Membrana/genética , Chaperonas Moleculares , Proteínas Nucleares/efeitos dos fármacos , Proteínas Nucleares/genética , Oligodendroglia/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro/análise
18.
J Thorac Cardiovasc Surg ; 126(2): 428-35, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12928640

RESUMO

OBJECTIVE: Contact between blood and the synthetic surfaces of a cardiopulmonary bypass circuit leads to platelet activation, and resultant platelet dysfunction contributes to postoperative bleeding. We compared the effects of various platelet inhibitors on preservation of platelet function during simulated cardiopulmonary bypass circulation. METHODS: Fresh human blood was recirculated in an in vitro cardiopulmonary bypass model circuit. We measured various platelet activation markers including expressions of PAC-1 and P-selectin, annexin V binding, and microparticle formations by means of whole-blood flow cytometry. RESULTS: Two types of glycoprotein IIb/IIIa complex antagonists, peptide-mimetic FK633 and abciximab and prostaglandin E(1), significantly prevented platelet loss and the increase in binding of PAC-1, an antibody specific for fibrinogen receptor on activated platelets, during extracorporeal circulation of heparinized blood. These antagonists significantly suppressed but did not abolish P-selectin expression, annexin V binding, and microparticle formation. Anti-von Willebrand factor monoclonal antibody and aurin tricarboxylic acid (an inhibitor of glycoprotein Ib) had no effect on platelet activation during simulated cardiopulmonary bypass circulation. These data suggest that inhibition of fibrinogen binding glycoprotein IIb/IIIa complex is partly effective in attenuating platelet activation in a heparinized cardiopulmonary bypass model circuit. The direct thrombin inhibitor argatroban prevented platelet loss and expression of P-selectin significantly more than did heparin. A combination of FK633 with argatroban as a substitute for heparin further prevented platelet loss and platelet secretion during simulated cardiopulmonary bypass circulation, although the inhibition of microparticle formation was less. CONCLUSION: The inhibition of both platelet adhesion and thrombin may be effective to preserve platelet number and function during cardiopulmonary bypass circulation.


Assuntos
Anestesia , Plaquetas/efeitos dos fármacos , Plaquetas/fisiologia , Circulação Extracorpórea , Ácidos Pipecólicos/farmacologia , Inibidores da Agregação Plaquetária/farmacologia , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/antagonistas & inibidores , Abciximab , Alprostadil/sangue , Alprostadil/farmacologia , Anexina A5/efeitos dos fármacos , Anticorpos Monoclonais/sangue , Anticorpos Monoclonais/farmacologia , Anticoagulantes/farmacologia , Arginina/análogos & derivados , Dipeptídeos/sangue , Dipeptídeos/farmacologia , Relação Dose-Resposta a Droga , Combinação de Medicamentos , Fosfatase 2 de Especificidade Dupla , Heparina/farmacologia , Humanos , Fragmentos Fab das Imunoglobulinas/sangue , Fragmentos Fab das Imunoglobulinas/farmacologia , Selectina-P/efeitos dos fármacos , Ácidos Pipecólicos/sangue , Ativação Plaquetária/efeitos dos fármacos , Ativação Plaquetária/fisiologia , Inibidores da Agregação Plaquetária/sangue , Contagem de Plaquetas , Complexo Glicoproteico GPIb-IX de Plaquetas/efeitos dos fármacos , Glicoproteína IIb da Membrana de Plaquetas/efeitos dos fármacos , Proteína Fosfatase 2 , Proteínas Tirosina Fosfatases/efeitos dos fármacos , Sulfonamidas , Resultado do Tratamento , Fator de von Willebrand/efeitos dos fármacos
19.
Environ Mol Mutagen ; 41(1): 14-27, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12552588

RESUMO

In vitro assays for chromosome aberrations (i.e., in vitro micronucleus and in vitro metaphase analysis tests) frequently produce false-positive or exaggerated-positive results. Our previous work suggested that apoptosis interferes with these tests, producing misleading results. These previous studies were conducted by performing the in vitro micronucleus test in CTLL-2 cells and a CTLL-2 cell derivative stably transfected with the apoptosis inhibitor gene bcl2. In the present study, these previous observations were extended by examining micronucleus induction with a larger number of compounds in both CTLL-2 and CTLL-2 bcl2 cells and measuring apoptosis with annexin V-FITC. Both cell lines were treated with different classes of compounds that were anticipated to be exclusively apoptosis inducers, or compounds known to be clastogens or aneugens, some of which were anticipated to be both genotoxic and apoptotic. We were able to confirm that compounds that are only apoptogenic induced micronuclei in CTLL-2 but not CTLL-2 bcl2 cells, indicating that the positive responses are due to apoptosis in CTLL-2 cells. Some genotoxins (clastogens and aneugens) did not produce apoptosis by the annexin V assay and gave similar responses in CTLL-2 and CTLL-2 bcl2 cells. Finally, higher responses were induced in CTLL-2 cells compared to CTLL-2 bcl2 cells that were treated with aneugens or clastogens that were also apoptosis inducers, suggesting that the greater response in CTLL-2 cells is a consequence of both genotoxicity and apoptosis. Finally, it was demonstrated that just eliminating CTLL-2 cells having three or more micronuclei from scoring was not adequate for correctly evaluating agents that only produce apoptosis. The results indicate that coupling the in vitro micronucleus test in both CTLL-2 and CTLL-2 bcl2 cells with the measurement of apoptosis is able to distinguish the genotoxic effects of a test compound from its apoptotic potential and is able to avoid interference from apoptosis in the in vitro micronucleus test. These observations may provide the basis for a useful genotoxicity assay.


Assuntos
Apoptose/efeitos dos fármacos , Apoptose/genética , Testes para Micronúcleos/métodos , Animais , Anexina A5/efeitos dos fármacos , Anexina A5/metabolismo , Células Cultivadas , Dexametasona/toxicidade , Etoposídeo/toxicidade , Fluoresceína-5-Isotiocianato/análise , Corantes Fluorescentes/análise , Camundongos , Camundongos Endogâmicos C57BL , Testes para Micronúcleos/normas , Mutagênicos/toxicidade , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Sensibilidade e Especificidade , Linfócitos T Citotóxicos/efeitos dos fármacos
20.
Nutr Cancer ; 44(2): 193-201, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12734068

RESUMO

Resveratrol, a phytoalexin found in grapes and wine, has been shown to exhibit a wide range of pharmacological properties and is believed to play a role in the chemoprevention of human cancer. Resveratrol has also been shown to induce antiproliferation and apoptosis of several leukemia cell lines. In the present study, we investigated the effect of resveratrol in adult T cell leukemia. Our present observations showed that resveratrol induced growth inhibition in all five human T cell lymphotrophic virus-1-infected cell lines examined, with 50% effective dose of 10.4-85.6 mM. In the resveratrol-treated cells, induction of apoptosis was confirmed by annexin V-based analyses and morphological changes. The most surprising observation was that resveratrol treatment resulted in a gradual decrease in the expression of survivin, an antiapoptotic protein, during cell apoptosis. These findings indicate that resveratrol inhibits the growth of human T cell lymphotrophic virus-1-infected cell lines, at least in part, by inducing apoptosis mediated by downregulation in survivin expression. In view of the accumulating evidence that survivin may be an important determinant of a clinical response in adult T cell leukemia, our present findings have led to the suggestion that resveratrol, a common constituent of the human diet, merits further investigation as a potential therapeutic agent for this incurable disease.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Apoptose/efeitos dos fármacos , Inibidores de Cisteína Proteinase/genética , Regulação para Baixo/efeitos dos fármacos , Expressão Gênica/efeitos dos fármacos , Infecções por HTLV-I/genética , Infecções por HTLV-I/prevenção & controle , Leucemia-Linfoma de Células T do Adulto/tratamento farmacológico , Proteínas Associadas aos Microtúbulos/efeitos dos fármacos , Proteínas Associadas aos Microtúbulos/genética , Estilbenos/farmacologia , Adulto , Anexina A5/efeitos dos fármacos , Apoptose/genética , Linhagem Celular/efeitos dos fármacos , Regulação para Baixo/genética , Citometria de Fluxo , Expressão Gênica/genética , Humanos , Proteínas Inibidoras de Apoptose , Proteínas de Neoplasias , Resveratrol , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Survivina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA