Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mater Horiz ; 11(2): 499-509, 2024 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-37966888

RESUMO

In-sensor reservoir computing (RC) is a promising technology to reduce power consumption and training costs of machine vision systems by processing optical signals temporally. This study demonstrates a high-dimensional in-sensor RC system with optoelectronic memristors to enhance the performance of the in-sensor RC system. Because optoelectronic memristors can respond to both optical and electrical stimuli, optical and electrical masks are proposed to improve the dimensionality and performance of the in-sensor RC system. An optical mask is employed to regulate the wavelength of light, while an electrical mask is used to control the initial conductance of zinc oxide optoelectronic memristors. The distinct characteristics of these two masks contribute to the representation of various distinguishable reservoir states, making it possible to implement diverse reservoir configurations with minimal correlation and to increase the dimensionality of the in-sensor RC system. Using the high-dimensional in-sensor RC system, handwritten digits are successfully classified with an accuracy of 94.1%. Furthermore, human action pattern recognition is achieved with a high accuracy of 99.4%. These high accuracies are achieved with the use of a single-layer readout network, which can significantly reduce the network size and training costs.

2.
Pharmazie ; 66(12): 982-7, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22312706

RESUMO

We investigated the mechanism of indole-3-carbinol (13C)/ultraviolet B (UVB)-induced apoptosis using SK-MEL-2 and SK-MEL-5 human melanoma cells. 13C/UVB significantly reduced the viability of SK-MEL-2 cells, whereas it had little influence on SK-MEL-5 cells. Correspondingly, cell cycle analysis showed that 13C/UVB induced a clear increase in the sub-G0/G1 phase in SK-MEL-2 cells. Furthermore, 13C/UVB activated caspase-9, caspase-8, caspase-3, and Bid and caused the cleavage of poly(ADP-ribose) polymerase (PARP) in SK-MEL-2 cells. In contrast, 13C/UVB showed no effects on the apoptotic signaling pathways in SK-MEL-5 cells. Moreover, we found that 13C down-regulated the microphthalmia-associated transcription factor (MITF) in SK-MEL-2 cells, but not in SK-MEL-5 cells. Next, to investigate the involvement of MITF in 13C/UVB-induced apoptosis, MITF silencing was conducted using small interfering RNA (siRNA) for MITF in SK-MEL-5 cells. Interestingly, 13C/UVB dramatically decreased the viability of MITF-down-regulated SK-MEL-5 cells. These results indicate that MITF plays a critical role in melanoma cell survival.


Assuntos
Anticarcinógenos/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Indóis/farmacologia , Melanoma/tratamento farmacológico , Melanoma/radioterapia , Fator de Transcrição Associado à Microftalmia/biossíntese , Raios Ultravioleta , Actinas/biossíntese , Western Blotting , Caspases/metabolismo , Ciclo Celular/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Morte Celular/efeitos da radiação , Linhagem Celular Tumoral , Regulação para Baixo/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Humanos , Fator de Transcrição Associado à Microftalmia/genética , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , RNA Interferente Pequeno/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transfecção
3.
Bioorg Med Chem Lett ; 19(3): 742-4, 2009 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-19119006

RESUMO

This study was aimed to evaluate detailed mechanisms on the apoptotic induction of benzyldihydroxyoctenone, a novel compound isolated from Streptomyces sp. KACC91015, in androgen-sensitive LNCaP prostate cancer cells. Benzyldihydroxyoctenone, designated as F3-2-5 in the current study, caused accumulation of apoptotic sub-G(1) phase in the flow cytometric analysis using propidium iodide staining. Moreover, the typical apoptotic DNA fragmentation of the LNCaP cells treated with 30 microM of F3-2-5 was confirmed using the TUNEL assay. This apoptotic induction of F3-2-5 in the LNCaP cells was associated with the cytochrome c release from mitochondria to cytosol, and the activation of procaspase-8, -9, and -3, as well as the specific proteolytic cleavage of poly(ADP-ribose) polymerase (PARP). In addition, F3-2-5 treatment caused the down-regulation of the antiapoptotic protein, such as Bcl-2 and Bcl-X(L), but the proapoptotic protein, such as Bax, was not influenced. To investigate whether apoptotic induction by F3-2-5 is also due to the down-regulation of androgen receptor (AR), Western blot analysis and quantitative RT-PCR were conducted in F3-2-5-treated LNCaP prostate cancer cells. We found that F3-2-5 significantly inhibited the expression levels of AR and prostate-specific antigen (PSA) proteins in a time-dependent manner, as well as F3-2-5 abrogated the up-regulation of AR and PSA genes with and without DHT. Therefore, F3-2-5 has been shown to be an androgen antagonist, suggesting that F3-2-5 could be a potent agent for the treatment of both androgen-dependent and hormone-refractory prostate cancer.


Assuntos
Antineoplásicos/síntese química , Antineoplásicos/farmacologia , Apoptose , Mitocôndrias/metabolismo , Octanóis/síntese química , Octanóis/farmacologia , Neoplasias da Próstata/tratamento farmacológico , Androgênios/metabolismo , Linhagem Celular Tumoral , Química Farmacêutica/métodos , Fragmentação do DNA , Regulação Neoplásica da Expressão Gênica , Humanos , Marcação In Situ das Extremidades Cortadas , Masculino , Modelos Químicos , Antígeno Prostático Específico/biossíntese
4.
Cancer Sci ; 98(6): 795-802, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17433036

RESUMO

In the course of screening for anticancer agents, a novel active compound, F3-2-5, was isolated from culture broth of Streptomyces sp., KACC91015. Its structure was identified using nuclear magnetic resonance, mass spectrometry, and molecular modeling experiments, and confirmed by total synthesis. The growth of various human cancer cell lines was inhibited in a dose-dependent manner by 0.06-0.48 mM F3-2-5 over 24 h. Its IC(50) values were estimated at 37 microM on HeLa, 72 microM on A549, and 190 microM on HT-29 cells. However, F3-2-5 had no antiproliferative effect on normal lymphocytes and normal fibroblasts used as controls. Moreover, it affected cell cycle regulation and caused apoptosis of the HeLa cells; chromatin condensation and DNA fragmentation were observed in cells exposed to 80 microM F3-2-5. Western blot analysis revealed that F3-2-5 inhibited phosphorylation of retinoblastoma protein (pRb) and reduced expression of cyclin-dependent kinase-4 and -6, and cyclin D1 and E, while levels of p53 and p21(WAF1/CIP1) increased. Taken together, these findings show that F3-2-5 inhibits proliferation of HeLa cells by inducing G(1) phase arrest as a consequence of inhibition of pRb phosphorylation following up-regulation of p21(WAF1/CIP1) and p53. Furthermore, apoptosis in HeLa cells treated with F3-2-5 was associated with an increase in Bax and p53, leading to release of cytochrome c, activation of caspase-3, and -8, and cleavage of poly (ADP-ribose) polymerase.


Assuntos
Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Fase G1/efeitos dos fármacos , Octanóis/uso terapêutico , Streptomyces/química , Antineoplásicos/farmacologia , Proliferação de Células/efeitos dos fármacos , Células HeLa , Humanos , Octanóis/isolamento & purificação , Fosforilação , Proteína do Retinoblastoma/metabolismo , Proteína Supressora de Tumor p53/metabolismo
5.
Bioorg Med Chem Lett ; 16(21): 5643-5, 2006 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-16908144

RESUMO

A novel compound showing antiproliferative effect was isolated from Streptomyces sp. Its structure was determined based on the interpretation of the NMR spectra, and its conformation was elucidated using molecular modeling and 2D NOESY. It was determined to be (E)-4-phenyl-3-(pyridine-2-yl)but-2-en-1-ol.


Assuntos
Butanóis/farmacologia , Proliferação de Células/efeitos dos fármacos , Piridinas/farmacologia , Streptomyces , Antineoplásicos/química , Antineoplásicos/farmacologia , Butanóis/química , Células HeLa , Humanos , Piridinas/química
6.
Cancer Sci ; 97(5): 430-6, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16630142

RESUMO

The purpose of the present study was to investigate the mechanisms involved in the antiproliferative and apoptotic effects of MCS-C2, a novel analog of the pyrrolo[2,3-d]pyrimidine nucleoside toyocamycin and sangivamycin, in human prostate cancer LNCaP cells. MCS-C2, a selective inhibitor of cyclin-dependent kinase, was found to inhibit cell growth in a time- and dose-dependent manner, and inhibit cell cycle progression by inducing the arrest of the G1 phase and apoptosis in LNCaP cells. When treated with 3 microM MCS-C2, inhibited proliferation associated with apoptotic induction was found in the LNCaP cells in a concentration and time-dependent manner, and nuclear DAPI staining revealed the typical nuclear features of apoptosis. Furthermore, MCS-C2 induced cell cycle arrest in the G1 phase through the upregulated phosphorylation of the p53 protein at Ser-15 and activation of its downstream target gene p21WAF1/CIP1. Accordingly, these results suggest that MCS-C2 inhibits the proliferation of LNCaP cells by way of G1-phase arrest and apoptosis in association with the regulation of multiple molecules in the cell cycle progression.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Quinases Ciclina-Dependentes/antagonistas & inibidores , Neoplasias da Próstata/tratamento farmacológico , Transdução de Sinais/efeitos dos fármacos , Toiocamicina/análogos & derivados , Antineoplásicos/metabolismo , Antineoplásicos/uso terapêutico , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Quinases Ciclina-Dependentes/metabolismo , Relação Dose-Resposta a Droga , Humanos , Masculino , Fosforilação , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Serina/metabolismo , Toiocamicina/metabolismo , Toiocamicina/farmacologia , Toiocamicina/uso terapêutico , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/metabolismo , Regulação para Cima
7.
Mol Cancer Res ; 4(3): 209-20, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16547158

RESUMO

The net balance of matrix metalloproteinases (MMP) and tissue inhibitor of metalloproteinases (TIMP) system has been known to be a key factor in tumor cell invasion. In the present study, we investigated the molecular mechanisms of anti-invasive and antimigrative activity of transforming growth factor (TGF)-beta1 on HT1080 human fibrosarcoma cells. In in vitro Matrigel invasion and Transwell migration assays, TGF-beta1 dose-dependently inhibited the invasion and migration of HT1080 cells, respectively. Gelatin zymography, Western blot, and real-time PCR analysis showed that TGF-beta1 enhanced the expression and secretion of MMP-2, TIMP-1, and, to a lesser degree, MMP-9 but not membrane type 1-MMP and TIMP-2. The addition of recombinant TIMP-1 protein reduced the Matrigel invasion and Transwell migration of HT1080 cells, similar to TGF-beta1. Because augmentation of TIMP-1 might be the major factor for the anti-invasive and antimigrative activity of TGF-beta1, we investigated possible molecular mechanisms responsible for the expression of TIMP-1 induced by TGF-beta1. Treatment of HT1080 cells with TGF-beta1 rapidly phosphorylated three mitogen-activated protein kinases [MAPK; extracellular signal-regulated kinase 1/2 (ERK1/2), p38, and c-Jun NH2-terminal kinase] and Akt. Among these kinases, the inhibition of only ERK1/2 pathway by PD98059, a specific inhibitor of MAPK/ERK kinase(MEK)-1, and transfection of dominant-negative MEK 1 effectively blocked the TIMP-1 induction by TGF-beta1. Mithramycin, a specific inhibitor of Sp1 transcription factor, but not curcumin, an inhibitor of activator protein-1, and transfection of Sp1 small interfering RNA significantly inhibited the TGF-beta1-induced expression of TIMP-1. In addition, electrophoretic mobility shift assay showed that TGF-beta1 up-regulated Sp1 DNA-binding activity, and PD98059 and mithramycin effectively inhibited these events. Finally, pretreatment of HT1080 cells with PD98059 and mithramycin, but not curcumin, restored the invasive activity of these cells. Taken together, these data suggest that TGF-beta1 modulates the net balance of the MMPs/TIMPs the systems in HT1080 cells for anti-invasion and antimigration by augmenting TIMP-1 through ERK1/2 pathway and Sp1 transcription factor.


Assuntos
MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fibrossarcoma/metabolismo , Fibrossarcoma/patologia , Fator de Transcrição Sp1/metabolismo , Inibidor Tecidual de Metaloproteinase-1/metabolismo , Fator de Crescimento Transformador beta/fisiologia , Movimento Celular/efeitos dos fármacos , Ativação Enzimática , Flavonoides/farmacologia , Humanos , Metaloproteinase 1 da Matriz/genética , Metaloproteinase 1 da Matriz/metabolismo , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Invasividade Neoplásica , Fosforilação , Inibidor Tecidual de Metaloproteinase-1/genética , Fator de Crescimento Transformador beta/farmacologia , Fator de Crescimento Transformador beta1
8.
Int J Cancer ; 118(11): 2711-20, 2006 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-16388516

RESUMO

Emodin (1,3,8-trihydroxy-6-methylanthraquinone), an active component in the root and rhizome of Rheum palmatum, is a tyrosine kinase inhibitor with a number of biological activities, including antitumor effects. Here, we examine the effects of emodin on vascular endothelial growth factor (VEGF)-A-induced angiogenesis, both in vitro and in vivo. In vitro, emodin dose-dependently inhibits proliferation, migration into the denuded area, invasion through a layer of Matrigel and tube formation of human umbilical vein endothelial cells (HUVECs) stimulated with VEGF-A. Emodin also inhibits basic fibroblast growth factor-induced proliferation and migration of HUVECs and VEGF-A-induced tube formation of human dermal microvascular endothelial cells. Specifically, emodin induces the cell cycle arrest of HUVECs in the G0/G1 phase by suppressing cyclin D1 and E expression and retinoblastoma protein phosphorylation, and suppresses Matrigel invasion by inhibiting the basal secretion of matrix metalloproteinase-2 and VEGF-A-stimulated urokinase plasminogen activator receptor expression. Additionally, emodin effectively inhibits phosphorylation of VEGF-A receptor-2 (KDR/Flk-1) and downstream effector molecules, including focal adhesion kinase, extracellular signal-regulated kinase 1/2, p38 mitogen-activated protein kinase, Akt and endothelial nitric oxide synthase. In vivo, emodin strongly suppresses neovessel formation in the chorioallantoic membrane of chick and VEGF-A-induced angiogenesis of the Matrigel plug in mice. Our data collectively demonstrate that emodin effectively inhibits VEGF-A-induced angiogenesis in vitro and in vivo. Moreover, inhibition of phosphorylation of KDR/Flk-1 and downstream effector molecules is a possible underlying mechanism of the anti-angiogenic activity of emodin. Based on these data, we propose that an interaction of emodin with KDR/Flk-1 may be involved in the inhibitory function of emodin toward VEGF-A-induced angiogenesis in vitro and responsible for its potent anti-angiogenic in vivo.


Assuntos
Emodina/farmacologia , Neovascularização Patológica/fisiopatologia , Fator A de Crescimento do Endotélio Vascular/fisiologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Materiais Biocompatíveis , Ciclo Celular , Movimento Celular , Proliferação de Células , Colágeno , Relação Dose-Resposta a Droga , Combinação de Medicamentos , Células Endoteliais , Humanos , Laminina , Camundongos , Invasividade Neoplásica/fisiopatologia , Fosforilação , Proteoglicanas , Cordão Umbilical/irrigação sanguínea , Fator A de Crescimento do Endotélio Vascular/efeitos dos fármacos
9.
Int J Oncol ; 28(1): 119-25, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16327987

RESUMO

Cisplatin is a DNA-damaging chemotherapeutic drug that may have a role in the adjuvant chemotherapy of several solid tumors, such as malignant glioblastoma, and the status of p53 tumor suppressor protein is a critical determinant of cisplatin chemosensitivity. In the present study, we showed the relationship of p53 status and chemosensitivity of cisplatin between two human malignant glioblastoma cell lines, A172 and T98G, harboring wild-type and mutant-type p53, respectively. Cisplatin was found to be more cytotoxic to A172 than T98G cells in a time- and concentration-dependent manner. Cisplatin-induced cytotoxicity manifested as apoptosis, characterized by genomic DNA fragmentation, nuclear condensation and an increase in sub-G1 population. Cisplatin induced the accumulation of p53 and p21 proteins in A172 cells, but not in T98G cells. The introduction of the adenovirus-mediated wild-type p53 gene into T98G cells resulted in the decrease of viability as well as the increase in sub-G1 population with p53 accumulation, activation of caspase-3 protease and release of cytochrome c from the mitochondria. These data strongly suggest that the expression of p53 is essential for the cytotoxic effect of cisplatin in human malignant glioblastoma cells, A172 and T98G, and the introduction of apoptotic signal molecules, such as p53, will be beneficial to achieve chemosensitivity in malignant glioma.


Assuntos
Neoplasias Encefálicas/patologia , Cisplatino/farmacologia , Glioblastoma/patologia , Proteína Supressora de Tumor p53/biossíntese , Apoptose , Caspase 3 , Caspase 9 , Caspases/metabolismo , Citocromos c/metabolismo , Dano ao DNA , Ativação Enzimática , Genes p53 , Humanos , Transdução Genética , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...