Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
1.
Mol Cancer ; 16(1): 10, 2017 01 30.
Artigo em Inglês | MEDLINE | ID: mdl-28137309

RESUMO

Radiation therapy is one of the major tools of cancer treatment, and is widely used for a variety of malignant tumours. Radiotherapy causes DNA damage directly by ionization or indirectly via the generation of reactive oxygen species (ROS), thereby destroying cancer cells. However, ionizing radiation (IR) paradoxically promotes metastasis and invasion of cancer cells by inducing the epithelial-mesenchymal transition (EMT). Metastasis is a major obstacle to successful cancer therapy, and is closely linked to the rates of morbidity and mortality of many cancers. ROS have been shown to play important roles in mediating the biological effects of IR. ROS have been implicated in IR-induced EMT, via activation of several EMT transcription factors-including Snail, HIF-1, ZEB1, and STAT3-that are activated by signalling pathways, including those of TGF-ß, Wnt, Hedgehog, Notch, G-CSF, EGFR/PI3K/Akt, and MAPK. Cancer cells that undergo EMT have been shown to acquire stemness and undergo metabolic changes, although these points are debated. IR is known to induce cancer stem cell (CSC) properties, including dedifferentiation and self-renewal, and to promote oncogenic metabolism by activating these EMT-inducing pathways. Much accumulated evidence has shown that metabolic alterations in cancer cells are closely associated with the EMT and CSC phenotypes; specifically, the IR-induced oncogenic metabolism seems to be required for acquisition of the EMT and CSC phenotypes. IR can also elicit various changes in the tumour microenvironment (TME) that may affect invasion and metastasis. EMT, CSC, and oncogenic metabolism are involved in radioresistance; targeting them may improve the efficacy of radiotherapy, preventing tumour recurrence and metastasis. This study focuses on the molecular mechanisms of IR-induced EMT, CSCs, oncogenic metabolism, and alterations in the TME. We discuss how IR-induced EMT/CSC/oncogenic metabolism may promote resistance to radiotherapy; we also review efforts to develop therapeutic approaches to eliminate these IR-induced adverse effects.


Assuntos
Transição Epitelial-Mesenquimal , Células-Tronco Neoplásicas/efeitos da radiação , Tolerância a Radiação , Desdiferenciação Celular , Humanos , Metástase Neoplásica , Neoplasias , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Fenótipo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais
2.
Mol Cancer ; 10: 113, 2011 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-21917150

RESUMO

BACKGROUND: In contrast to tumor-suppressive apoptosis and autophagic cell death, necrosis promotes tumor progression by releasing the pro-inflammatory and tumor-promoting cytokine high mobility group box 1 (HMGB1), and its presence in tumor patients is associated with poor prognosis. Thus, necrosis has important clinical implications in tumor development; however, its molecular mechanism remains poorly understood. RESULTS: In the present study, we show that Distal-less 2 (Dlx-2), a homeobox gene of the Dlx family that is involved in embryonic development, is induced in cancer cell lines dependently of reactive oxygen species (ROS) in response to glucose deprivation (GD), one of the metabolic stresses occurring in solid tumors. Increased Dlx-2 expression was also detected in the inner regions, which experience metabolic stress, of human tumors and of a multicellular tumor spheroid, an in vitro model of solid tumors. Dlx-2 short hairpin RNA (shRNA) inhibited metabolic stress-induced increase in propidium iodide-positive cell population and HMGB1 and lactate dehydrogenase (LDH) release, indicating the important role(s) of Dlx-2 in metabolic stress-induced necrosis. Dlx-2 shRNA appeared to exert its anti-necrotic effects by preventing metabolic stress-induced increases in mitochondrial ROS, which are responsible for triggering necrosis. CONCLUSIONS: These results suggest that Dlx-2 may be involved in tumor progression via the regulation of metabolic stress-induced necrosis.


Assuntos
Antígenos de Superfície/metabolismo , Neoplasias/metabolismo , Estresse Fisiológico , Antígenos de Superfície/genética , Apoptose , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Agregação Celular , Linhagem Celular Tumoral , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Feminino , Perfilação da Expressão Gênica , Técnicas de Silenciamento de Genes , Glucose/deficiência , Humanos , Potencial da Membrana Mitocondrial , Mitocôndrias/metabolismo , Membranas Mitocondriais/metabolismo , Necrose , Neoplasias/patologia , Análise de Sequência com Séries de Oligonucleotídeos , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Permeabilidade , Interferência de RNA , Espécies Reativas de Oxigênio/metabolismo , Acetato de Tetradecanoilforbol/farmacologia
3.
Int J Cancer ; 126(7): 1582-95, 2010 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-19728331

RESUMO

Hepatocellular carcinoma (HCC) is resistant to chemotherapy. Recently, however, several oxaliplatin-based combinatorial treatments have shown a promising anti-tumor activity in patients with HCC. Presently, we demonstrate that oxaliplatin triggers necrosis more than apoptosis in HepG2, SK-Hep1, SNU-423 and Hep3B HCC cells, while mainly inducing apoptosis in HCT116 and HT29 colon cancer cells. Interestingly, ursodeoxycholic acid (UDCA), a less hydrophobic bile acid that can suppress carcinogenesis, shifted oxaliplatin-induced necrosis to apoptosis in HepG2 cells. The same effect was produced by hydrophilic bile acids (tauroursodeoxycholic acid and taurohyodeoxycholic acid), but not by highly hydrophobic bile acids (deoxycholic acid and chenodeoxycholic acid). UDCA also triggered the necrosis-to-apoptosis switch when cotreated with other platinum-based chemotherapeutic drugs including cisplatin and carboplatin, suggesting that the cell death mode switching effect of UDCA is a general phenomenon when combined with platinum drugs. Oxaliplatin produced high level of reactive oxygen species (ROS) in HepG2 cells and UDCA significantly reduced oxaliplatin-induced ROS generation. In addition, N-acetyl-L-cysteine and the superoxide scavengers butylated hydroxyanisole and dihydroxybenzene-3,5-disulfonic acid attenuated necrosis, indicating a critical role(s) of ROS in occurrence of necrotic death. Apoptosis induced by combined treatment appeared to be mediated by p53-caspase 8-caspase 3 pathway. In conclusion, UDCA switches oxaliplatin-induced necrosis to apoptosis via inhibition of ROS production and activation of the p53-caspase 8 pathway in HepG2 cells. As necrosis and subsequent inflammation are implicated in tumor progression and malignancy, our results imply a potential improved efficacy of UDCA-combined chemotherapy in HCC by reducing inflammatory responses that may be triggered by oxaliplatin.


Assuntos
Apoptose/efeitos dos fármacos , Carcinoma Hepatocelular/tratamento farmacológico , Caspase 8/metabolismo , Neoplasias Hepáticas/tratamento farmacológico , Compostos Organoplatínicos/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Ácido Ursodesoxicólico/farmacologia , Antineoplásicos/farmacologia , Western Blotting , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Caspase 8/genética , Colagogos e Coleréticos/farmacologia , Ensaio de Desvio de Mobilidade Eletroforética , Células HT29/efeitos dos fármacos , Células Hep G2/efeitos dos fármacos , Humanos , L-Lactato Desidrogenase/metabolismo , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Necrose , Oxaliplatina , RNA Interferente Pequeno/farmacologia , Proteína Supressora de Tumor p53/antagonistas & inibidores , Proteína Supressora de Tumor p53/genética
4.
Biochemistry ; 48(18): 3795-7, 2009 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-19338268

RESUMO

Heat shock transcription factor 1 (HSF1) is a central regulator in the heat shock response. However, its trimerization mechanism remains unclear. Here, we demonstrate that three conserved aromatic amino acids (Trp37, Tyr60, and Phe104) are essential for HSF1 trimerization. Point mutation and fluorescence spectroscopy experiments show that an intramolecular interaction between Tyr60 and alpha-helix 1 in the DNA-binding domain stabilizes the HSF1 structure upon heat stress. Furthermore, intermolecular aromatic-aromatic interaction between the Trp37 and Phe104 supports the approach with the Cys36 and Cys103. Thus, the existence of two differential interactions facilitates the formation of intermolecular disulfide bonds, leading to the heat-induced HSF1 trimerization.


Assuntos
Biopolímeros/química , Proteínas de Ligação a DNA/química , Dissulfetos/química , Fatores de Transcrição/química , Fatores de Transcrição de Choque Térmico , Humanos , Conformação Proteica , Espectrometria de Fluorescência
5.
Int J Oncol ; 34(4): 1077-83, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19287965

RESUMO

Anthocyanins are a class of flavonoids, widely spread throughout the plant kingdom, that exhibit important anti-oxidant and anti-inflammatory actions as well as chemotherapeutic effects. However, little is known concerning the molecular mechanisms by which these activities are exerted. In this study, we investigated the anthocyanins isolated from Vitis coignetiae Pulliat for their potential anti-proliferative and apoptotic effects on human leukemia U937 cells. It was found that these anthocyanins inhibit cell viability and induce apoptotic cell death of U937 cells in a dose-dependent manner, as measured by hemocytometer counts, by alteration in the mitochondrial membrane potential, by increases in sub-G1 populations and by DNA ladder formation. Apoptosis of U937 cells by anthocyanins was associated with modulation of expression of Bcl-2 and IAP family members. Consequently, anthocyanin treatment induced proteolytic activation of caspase-3, -8 and -9, and a concomitant degradation of poly(ADP-ribose) polymerase. However, anthocyanin-induced growth inhibition and apoptosis were significantly attenuated in Bcl-2 overexpressing U937 cells. Furthermore, z-DEVD-fmk, a caspase-3 specific inhibitor, blocked apoptosis and increased the survival of anthocyanin-treated U937 cells. Taken together, these results show that Bcl-2 and caspases are key regulators of apoptosis in response to anthocyanins in human leukemia U937 cells.


Assuntos
Antocianinas/farmacologia , Apoptose , Caspases/metabolismo , Regulação para Baixo , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Sobrevivência Celular , DNA/metabolismo , Ativação Enzimática , Humanos , Potenciais da Membrana , Membranas Mitocondriais/metabolismo , Células U937 , Vitis/metabolismo
6.
Mol Med Rep ; 20(3): 2339-2346, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31322179

RESUMO

Reactive oxygen species (ROS) are important cellular second messengers involved in various aspects of cell signaling. ROS are elevated in multiple types of cancer cells, and this elevation is known to be involved in pathological processes of cancer. Although high levels of ROS exert cytotoxic effects on cancer cells, low levels of ROS stimulate cell proliferation and survival by inducing several pro­survival signaling pathways. In addition, ROS have been shown to induce epithelial­mesenchymal transition (EMT), which is essential for the initiation of metastasis. However, the precise mechanism of ROS­induced EMT remains to be elucidated. In the present study, it was indicated that ROS induce EMT by activating Snail expression, which then represses E­cadherin expression in MCF­7 cells. It was further indicated that distal­less homeobox­2 (Dlx­2), one of the human Dlx gene family proteins involved in embryonic development, acts as an upstream regulator of ROS­induced Snail expression. It was also revealed that ROS treatment induces the glycolytic switch, a phenomenon whereby cancer cells primarily rely on glycolysis instead of mitochondrial oxidative phosphorylation for ATP production, even in the presence of oxygen. In addition, ROS inhibited oxidative phosphorylation and caused cytochrome c oxidase inhibition via the Dlx­2/Snail cascade. These results suggest that ROS induce EMT, the glycolytic switch and mitochondrial repression by activating the Dlx­2/Snail axis, thereby playing crucial roles in MCF­7 cancer cell progression.


Assuntos
Neoplasias da Mama/metabolismo , Transição Epitelial-Mesenquimal , Proteínas de Homeodomínio/metabolismo , Mitocôndrias/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Fatores de Transcrição da Família Snail/metabolismo , Fatores de Transcrição/metabolismo , Feminino , Glicólise , Humanos , Células MCF-7 , Transdução de Sinais
7.
Biochemistry ; 47(22): 6007-15, 2008 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-18457423

RESUMO

Under circumstances of heat stress, heat shock transcription factor 1 (HSF1) plays important roles in heat shock protein expression. In this study, an increasing concentration of dithiothreitol (DTT) was found to either enhance or inhibit the heat-induced trimerization of HSF1, suggesting the involvement of dual redox-dependent HSF1 activation mechanisms. Our in vitro experiments show that the heat-induced bonding between the cysteine C36 and C103 residues of HSF1 forms an intermolecular disulfide covalent bond (SS-I bond) and that it directly causes HSF1 to trimerize and bond to DNA. Gel filtration assays show that HSF1 can form intermolecular hydrophobic interaction-mediated (iHI-m) noncovalent oligomers. However, the lack of a trimerization domain prevents HSF1 activation, which suggests that iHI-m noncovalent trimerization is a precondition of SS-I bond formation. On the other hand, intramolecular SS-II bond (in which the C153, C373, and C378 residues of HSF1 participate) formation inhibits this iHI-m trimerization, thereby preventing SS-I bond formation and DNA binding. Thus, HSF1 activation is regulated positively by intermolecular SS-I bond formation and negatively by intramolecular SS-II bond formation. Importantly, these two SS bonds confer different DTT sensitivities (the SS-II bond is more sensitive). Therefore, a low concentration of DTT cleaves the SS-II bond but not the SS-I bond and thus improves DNA binding of HSF1, whereas a high concentration DTT cuts both SS bonds and inhibits HSF1 activation. We propose that these interesting effects further explain cellular HSF1 trimerization, DNA binding, and transcription when cells are under stress.


Assuntos
Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/metabolismo , DNA/metabolismo , Fatores de Transcrição/química , Fatores de Transcrição/metabolismo , Sítios de Ligação , Cisteína/genética , Cisteína/metabolismo , Ditiotreitol , Fatores de Transcrição de Choque Térmico , Humanos , Modelos Biológicos , Oxirredução
8.
Biochim Biophys Acta ; 1769(1): 41-8, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17207870

RESUMO

Caudal-related homeobox transcription factors are involved in the definition of the anteroposterior axis and intestinal development. Recent reports indicate that dysregulation of CDX1 and CDX2, the human homologues of Drosophila caudal, are associated with several types of cancer. Very little is known, however, about the regulatory mechanisms that direct the caudal-related homeobox gene expression. In this study, we have identified the binding sites for bHLH-PAS proteins, referred to as CNS midline element (CME), in the 5'-flanking region of the Drosophila caudal gene. Analyses using transgenic flies carrying a caudal-lacZ fusion gene bearing a wild-type or mutant CME indicate that the CME sites are required for caudal gene expression in vivo. We also determined that the caudal promoter activity can be regulated by Trachealess (Trh)/Tango (Tgo) bHLH-PAS proteins, via the CME sites. Our results suggest that the Drosophila caudal gene is a target of the Trh/Tgo bHLH-PAS proteins.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/farmacologia , Proteínas de Drosophila/farmacologia , Drosophila/genética , Regulação da Expressão Gênica no Desenvolvimento , Genes Homeobox , Proteínas de Homeodomínio/genética , Fatores de Transcrição/genética , Animais , Proteínas de Drosophila/genética , Embrião não Mamífero , Transgenes
9.
Cancer Lett ; 261(1): 98-107, 2008 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-18093727

RESUMO

Platycodin D (PD) is a major constituent of triterpene saponins found in the root of Platycodon grandiflorum. Recent studies have demonstrated that PD is a potentially interesting candidate for use in cancer chemotherapy. However, the molecular mechanisms responsible for PD-induced telomerase inhibition remain to be poorly known. In this study, we examined the effects of PD treatment on telomerase activity in different human leukemia cell lines. At concentrations between 10 and 20 microM, PD exerted a dose-dependent direct cytotoxic effect and inhibition of telomerase activity via downregulation of hTERT expression. Because c-Myc and Sp1 are known to directly regulate transcription of hTERT, we also evaluated the expression and DNA binding activity of these proteins. PD treatment reduced c-Myc and Sp1 protein levels and DNA binding activities in a dose-dependent manner. We also observed that PD treatment downregulates the activation of Akt, thereby reducing the phosphorylation and nuclear translocation of hTERT. We conclude that PD has direct cytotoxic effect on human leukemia cells and suppresses telomerase activity through transcriptional and posttranslational suppression of hTERT.


Assuntos
Apoptose/efeitos dos fármacos , Leucemia/enzimologia , Leucemia/patologia , Saponinas/farmacologia , Telomerase/metabolismo , Triterpenos/farmacologia , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Regulação para Baixo , Humanos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo
10.
Int J Oncol ; 32(4): 851-60, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18360712

RESUMO

Both cellular and clinical studies have shown that hyperthermia is one of the most potent sensitizers for the action of ionizing radiation. Although hyperthermic improvement in clinical outcome is suggested to be linked to its ability to induce cell cycle arrest and apoptosis, and to activate the immune system and to cause increases in blood flow and tumor oxygenation, the mechanism behind this is still unclear. Previously, we demonstrated that glucose deprivation (GD), a common characteristic of the tumor microenvironment, induced necrosis, which is implicated in tumor progression and aggressiveness, through the production of reactive oxygen species (ROS) in A549 lung carcinoma cells. We examined the effects of heat shock on ROS production and necrosis in response to GD. Here we show that mild, but not harsh, heat shock prevented GD-induced necrosis and switched the cell death mode to apoptosis in A549 cells through the ERK1/2 pathway that could suppress GD-induced CuZnSOD release and ROS production. These results demonstrate that contrary to severe heat shock, mild heat shock has the ability to decrease oxidative stress in cells, thereby causing the cell death mode switch from tumor promoting necrosis to tumor suppressive apoptosis, which may contribute to its anti-neoplastic activities.


Assuntos
Adenocarcinoma/patologia , Apoptose , Glucose/deficiência , Hipertermia Induzida , Neoplasias Pulmonares/patologia , Linhagem Celular Tumoral , MAP Quinases Reguladas por Sinal Extracelular/fisiologia , Febre de Causa Desconhecida/complicações , Humanos , Necrose , Espécies Reativas de Oxigênio/metabolismo , Superóxido Dismutase/fisiologia
11.
Oncol Rep ; 19(5): 1165-71, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18425372

RESUMO

Sodium salicylate, the active metabolite of aspirin, has been shown to exert anti-inflammatory activities by inhibiting the expression of various pro-inflammatory factors, and has potent anti-cancer effects against a number of human cancers including colon, lung, breast and leukemia. Necrotic cell death is emerging as one of the crucial factors that trigger an inflammatory response since during necrotic death the cell membrane is ruptured and the intracellular constituents including high mobility group box 1 (HMGB1) are released into the extracellular space, thereby activating an inflammatory response. In contrast, autophagic death is regarded as a form of tumour suppressive cell death, as indicated in tumour suppressors such as beclin 1 in autophagic pathways. To better understand the anti-inflammatory properties of sodium salicylate and its effect on necrotic cell death in A549 cells induced by glucose depletion (GD), a common characteristic of the tumour micro-environment, was examined. While GD induced mostly necrotic death in A549 cells, salicylate suppresssed GD-induced necrosis and HMGB1 release. In addition, salicylate shifted the cell death pattern to autophagy by inhibiting GD-induced Cu/Zn superoxide dismutase release and ROS production. These results indicate that the activity of salicylate to prevent necrotic death may contribute to its anti-inflammatory action and suppress tumour development possibly through switching the cell death mode from tumour-promoting necrotic cell death to tumour-suppressive autophagic cell death.


Assuntos
Adenocarcinoma/metabolismo , Autofagia , Inibidores de Ciclo-Oxigenase/farmacologia , Regulação Neoplásica da Expressão Gênica , Glucose/metabolismo , Proteína HMGB1/biossíntese , Neoplasias Pulmonares/embriologia , Necrose , Salicilato de Sódio/farmacologia , Anti-Inflamatórios/farmacologia , Benzimidazóis/farmacologia , Linhagem Celular Tumoral , Proteína HMGB1/metabolismo , Humanos , Superóxido Dismutase/metabolismo , Sais de Tetrazólio/farmacologia , Tiazóis/farmacologia
12.
Oncol Rep ; 19(2): 517-26, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18202802

RESUMO

The tumor suppressor protein p53 restricts proliferation in response to DNA damage or the deregulation of mitogenic oncogenes, by leading to the induction of various cell cycle checkpoints, apoptosis or cellular senescence. Consequently, p53 mutations increase cell proliferation and survival and in some settings promote genomic instability and resistance to certain anti-cancer drugs. It is very important to identify chemotherapeutic agents that activate in a p53-independent manner for the development of treatments for p53-deficient tumors. Pectenotoxin-2 (PTX-2), isolated from marine sponges has been reported to display significant cytotoxicity to p53-deficient cancer cell lines. In this study, we compared the anti-cancer activity of PTX-2 in order to further test the status of p53 using two well-known hepatocarcinoma cell lines, p53-deficient Hep3B and p53-wild-type HepG2. MTT assay indicated that Hep3B cells were highly susceptible, whereas HepG2 cells were more resistant to this compound which was connected with the induction of apoptotic cell death in p53-deficient Hep3B cells, though not in HepG2 cells. The apoptosis induced by PTX-2 in Hep3B cells was associated with the down-regulation of anti-apoptotic Bcl-2 members (Bcl-2 and Bcl-xL) and IAP family proteins, the up-regulation of pro-apoptotic Bax protein and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-receptor 1/receptor 2 (DR4/DR5) and mitochondrial dysfunction. PTX-2 activated caspases (caspase-3, -8 and -9) and the blockade of caspase-3 activity by the caspase-3 inhibitor prevented the PTX-2-induced apoptosis in Hep3B cells. Additionally, the transcription factor early growth response-1 (Egr-1) gene was transcriptionally activated and the levels of non-steroidal anti-inflammatory drugs (NSAID)-activated gene-1 (NAG-1) protein were also elevated in PTX-2-treated Hep3B cells. Although further studies are needed to prove that an increased expression of Egr-1 by PTX-2 directly leads to NAG-1 induction and then apoptosis induction in p53-deficient Hep3B cells, the results of this study suggest that PTX-2 may be a good candidate for the development of a potential anti-tumorigenic agent in p53-deficient tumors.


Assuntos
Antineoplásicos/farmacologia , Apoptose , Carcinoma Hepatocelular/metabolismo , Furanos/farmacologia , Neoplasias Hepáticas/metabolismo , Piranos/farmacologia , Proteína Supressora de Tumor p53/metabolismo , Carcinoma Hepatocelular/genética , Inibidores de Caspase , Caspases/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Citocinas/metabolismo , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Fator 15 de Diferenciação de Crescimento , Humanos , Neoplasias Hepáticas/genética , Macrolídeos , Metaloproteinases da Matriz/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Proteína Supressora de Tumor p53/genética
13.
J Microbiol Biotechnol ; 18(11): 1862-8, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19047834

RESUMO

Streptochlorin is a small molecule isolated from marine Streptomyces sp. that is known to have antiangiogenic and anticancer properties. In this study, we examined the effects of this compound on reactive oxygen species (ROS) production and the association of these effects with apoptotic tumor cell death, using a human hepatocarcinoma Hep3B cell line. The results of this study demonstrated that streptochlorin mediates ROS production, and that this mediation is followed by a decrease in the mitochondrial membrane potential (MMP, m), activation of caspase-3, and downregulation of antiapoptotic Bcl-2 protein. The quenching of ROS generation by N-acetyl-L-cysteine administration, a scavenger of ROS, reversed the streptochlorin-induced apoptosis effects via inhibition of ROS production, MMP collapse, and the subsequent activation of caspase-3. These observations clearly indicate that ROS are involved in the early molecular events in the streptochlorin-induced apoptotic pathway. Taken together, our data imply that streptochlorin-induced ROS is a key mediator of MMP collapse, which leads to the caspase-3 activation, culminating in apoptosis.


Assuntos
Apoptose , Indóis/farmacologia , Potencial da Membrana Mitocondrial , Mitocôndrias/metabolismo , Oxazóis/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Streptomyces/metabolismo , Apoptose/efeitos dos fármacos , Carcinoma Hepatocelular , Linhagem Celular Tumoral , Humanos , Indóis/isolamento & purificação , Indóis/metabolismo , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Potencial da Membrana Mitocondrial/fisiologia , Mitocôndrias/fisiologia , Oxazóis/isolamento & purificação , Oxazóis/metabolismo , Espécies Reativas de Oxigênio/farmacologia
14.
Oxid Med Cell Longev ; 2018: 3537471, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29636841

RESUMO

Rapidly growing malignant tumors frequently encounter hypoxia and nutrient (e.g., glucose) deprivation, which occurs because of insufficient blood supply. This results in necrotic cell death in the core region of solid tumors. Necrotic cells release their cellular cytoplasmic contents into the extracellular space, such as high mobility group box 1 (HMGB1), which is a nonhistone nuclear protein, but acts as a proinflammatory and tumor-promoting cytokine when released by necrotic cells. These released molecules recruit immune and inflammatory cells, which exert tumor-promoting activity by inducing angiogenesis, proliferation, and invasion. Development of a necrotic core in cancer patients is also associated with poor prognosis. Conventionally, necrosis has been thought of as an unregulated process, unlike programmed cell death processes like apoptosis and autophagy. Recently, necrosis has been recognized as a programmed cell death, encompassing processes such as oncosis, necroptosis, and others. Metabolic stress-induced necrosis and its regulatory mechanisms have been poorly investigated until recently. Snail and Dlx-2, EMT-inducing transcription factors, are responsible for metabolic stress-induced necrosis in tumors. Snail and Dlx-2 contribute to tumor progression by promoting necrosis and inducing EMT and oncogenic metabolism. Oncogenic metabolism has been shown to play a role(s) in initiating necrosis. Here, we discuss the molecular mechanisms underlying metabolic stress-induced programmed necrosis that promote tumor progression and aggressiveness.


Assuntos
Autofagia/fisiologia , Morte Celular/fisiologia , Necrose/metabolismo , Neoplasias/patologia , Apoptose , Progressão da Doença , Humanos
15.
Oxid Med Cell Longev ; 2018: 1027453, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30671168

RESUMO

Metastasis is a major obstacle to the efficient and successful treatment of cancer. Initiation of metastasis requires epithelial-mesenchymal transition (EMT) that is regulated by several transcription factors, including Snail and ZEB1/2. EMT is closely linked to the acquisition of cancer stem cell (CSC) properties and chemoresistance, which contribute to tumor malignancy. Tumor suppressor p53 inhibits EMT and metastasis by negatively regulating several EMT-inducing transcription factors and regulatory molecules; thus, its inhibition is crucial in EMT, invasion, metastasis, and stemness. Metabolic alterations are another hallmark of cancer. Most cancer cells are more dependent on glycolysis than on mitochondrial oxidative phosphorylation for their energy production, even in the presence of oxygen. Cancer cells enhance other oncogenic metabolic pathways, such as glutamine metabolism, pentose phosphate pathway, and the synthesis of fatty acids and cholesterol. Metabolic reprogramming in cancer is regulated by the activation of oncogenes or loss of tumor suppressors that contribute to tumor progression. Oncogenic metabolism has been recently linked closely with the induction of EMT or CSC phenotypes by the induction of several metabolic enzyme genes. In addition, several transcription factors and molecules involved in EMT or CSCs, including Snail, Dlx-2, HIF-1α, STAT3, TGF-ß, Wnt, and Akt, regulate oncogenic metabolism. Moreover, p53 induces metabolic change by directly regulating several metabolic enzymes. The collective data indicate the importance of oncogenic metabolism in the regulation of EMT, cell invasion and metastasis, and adoption of the CSC phenotype, which all contribute to malignant transformation and tumor development. In this review, we highlight the oncogenic metabolism as a key regulator of EMT and CSC, which is related with tumor progression involving metastasis and chemoresistance. Targeting oncometabolism might be a promising strategy for the development of effective anticancer therapy.


Assuntos
Transição Epitelial-Mesenquimal , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Células-Tronco Neoplásicas/metabolismo , Fatores de Transcrição/metabolismo , Animais , Humanos , Metástase Neoplásica , Neoplasias/patologia , Células-Tronco Neoplásicas/patologia
16.
Int J Mol Med ; 20(2): 187-92, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17611636

RESUMO

Ethyl pyruvate (EP), a stable lipophilic pyruvate derivative, has been shown to exert anti-inflammatory activities through inhibiting the expression of various pro-inflammatory mediators as well as circulating levels of high mobility group box protein 1 (HMGB1) in a variety of in vitro and in vivo model systems. Necrotic cell death triggers an inflammatory response through release of HMGB1 in the extracellular space due to the membrane rupture. In an effort to better understand the pharmacological action mechanism that could explain the anti-inflammatory properties of EP, we examined the effects of EP on necrotic cell death in A549 lung adenocarcinoma cells in response to glucose deprivation (GD), a common characteristic of the tumor microenvironment. Here we show that EP prevented GD-induced necrosis and HMGB1 release and switched the cell death mode to apoptosis through inhibiting GD-induced CuZn superoxide dismutase release and ROS production. These results suggest that the necrosis-to-apoptosis switch activity of EP may contribute to its anti-inflammatory action and that EP may suppress tumor development possibly through its activity to induce the cell death mode switch from tumor promoting necrotic cell death to tumor suppressive apoptotic cell death.


Assuntos
Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Apoptose/efeitos dos fármacos , Proteína HMGB1/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Piruvatos/farmacologia , Glucose/farmacologia , Humanos , Inflamação/prevenção & controle , Necrose/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Superóxido Dismutase/metabolismo , Células Tumorais Cultivadas
17.
Oncotarget ; 7(7): 7925-39, 2016 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-26771232

RESUMO

Most cancer cells depend on enhanced glucose and glutamine (Gln) metabolism for growth and survival. Oncogenic metabolism provides biosynthetic precursors for nucleotides, lipids, and amino acids; however, its specific roles in tumor progression are largely unknown. We previously showed that distal-less homeobox-2 (Dlx-2), a homeodomain transcription factor involved in embryonic and tumor development, induces glycolytic switch and epithelial-mesenchymal transition (EMT) by inducing Snail expression. Here we show that Dlx-2 also induces the expression of the crucial Gln metabolism enzyme glutaminase (GLS1), which converts Gln to glutamate. TGF-ß and Wnt induced GLS1 expression in a Dlx-2-dependent manner. GLS1 shRNA (shGLS1) suppressed in vivo tumor metastasis and growth. Inhibition of Gln metabolism by shGLS1, Gln deprivation, and Gln metabolism inhibitors (DON, 968 and BPTES) prevented Dlx-2-, TGF-ß-, Wnt-, and Snail-induced EMT and glycolytic switch. Finally, shDlx-2 and Gln metabolism inhibition decreased Snail mRNA levels through p53-dependent upregulation of Snail-targeting microRNAs. These results demonstrate that the Dlx-2/GLS1/Gln metabolism axis is an important regulator of TGF-ß/Wnt-induced, Snail-dependent EMT, metastasis, and glycolytic switch.


Assuntos
Transição Epitelial-Mesenquimal , Glutaminase/metabolismo , Glutamina/metabolismo , Glicólise/fisiologia , Proteínas de Homeodomínio/metabolismo , Neoplasias/patologia , Fatores de Transcrição/metabolismo , Trifosfato de Adenosina/metabolismo , Apoptose , Western Blotting , Proliferação de Células , Imunoprecipitação da Cromatina , Imunofluorescência , Glutaminase/antagonistas & inibidores , Glutaminase/genética , Células HeLa , Células Hep G2 , Proteínas de Homeodomínio/antagonistas & inibidores , Proteínas de Homeodomínio/genética , Humanos , Células MCF-7 , Neoplasias/genética , Neoplasias/metabolismo , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Fatores de Transcrição da Família Snail/antagonistas & inibidores , Fatores de Transcrição da Família Snail/genética , Fatores de Transcrição da Família Snail/metabolismo , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/genética , Fator de Crescimento Transformador beta/antagonistas & inibidores , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo , Células Tumorais Cultivadas
18.
J Neurosci ; 22(18): 7931-40, 2002 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-12223546

RESUMO

Dopamine receptor subtypes D1 and D2, and many other seven-transmembrane receptors including adenosine receptor A2A, are colocalized in striatum of brain. These receptors stimulate or inhibit adenylyl cyclases (ACs) to produce distinct physiological and pharmacological responses and interact with each other synergistically or antagonistically at various levels. The identity of the AC isoform that is coupled to each of these receptors, however, remains unknown. To investigate the in vivo role of the type 5 adenylyl cyclase (AC5), which is preferentially expressed in striatum, mice deficient for the AC5 gene were generated. The genetic ablation of the AC5 gene eliminated >80% of forskolin-induced AC activity and 85-90% of AC activity stimulated by either D1 or A2A receptor agonists in striatum. However, D1- or A2A-specific pharmaco-behaviors were basically preserved, whereas the signal cascade from D2 to AC was completely abolished in AC5(-/-), and motor activity of AC5(-/-) was not suppressed by treatment of cataleptic doses of the antipsychotic drugs haloperidol and sulpiride. Interestingly, both haloperidol and clozapine at low doses remarkably increased the locomotion of AC5(-/-) in the open field test that was produced in part by a common mechanism that involved the increased activation of D1 dopamine receptors. Together, these results suggest that AC5 is the principal AC integrating signals from multiple receptors including D1, D2, and A2A in striatum and the cascade involving AC5 among diverse D2 signaling pathways is essential for neuroleptic effects of antipsychotic drugs.


Assuntos
Adenilil Ciclases/deficiência , Encéfalo/metabolismo , Isoenzimas/deficiência , Receptores de Dopamina D2/metabolismo , Adenilil Ciclases/genética , Animais , Antipsicóticos/farmacologia , Encéfalo/efeitos dos fármacos , Cerebelo/efeitos dos fármacos , Cerebelo/metabolismo , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Colforsina/farmacologia , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/metabolismo , AMP Cíclico , Agonistas de Dopamina/farmacologia , Ativação Enzimática/efeitos dos fármacos , Ativadores de Enzimas/farmacologia , Viabilidade Fetal/genética , Marcação de Genes , Heterozigoto , Homozigoto , Isoenzimas/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Atividade Motora/efeitos dos fármacos , Receptor A2A de Adenosina , Receptores de Dopamina D1/metabolismo , Receptores Purinérgicos P1/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
19.
Int J Mol Med ; 16(5): 841-9, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16211253

RESUMO

Sodium salicylate, one of anti-inflammatory agents, is known to partially induce the heat shock response: it stimulates the DNA-binding of heat shock factor 1 (HSF1) without inducing heat shock gene expression. Here we show that when C6 glioma cells are recovered from sodium salicylate treatment, they highly induce heat shock protein 72 (HSP72), but not HSP73 and HSP90, demonstrating that salicylate-induced inert HSF1 can be fully activated into a transcriptionally competent form by sodium salicylate recovery (SR)-specific mechanism. Fluorescent analysis using 2',7'-dichlorodihydrofluorescein diacetate revealed that sodium salicylate enhanced reactive oxygen species (ROS) production. N-acetyl-L-cysteine (NAC, a ROS scavenger) completely suppressed SR-induced HSP72 synthesis and HSP72 promoter-driven CAT reporter gene transcription as well as salicylate-induced HSF1-DNA binding, indicating a critical role(s) of ROS in the SR-induced HSP72 gene regulation. We also show that treatment of C6 cells with sodium salicylate activated p38MAPK and inactivated ERK1/2 in a ROS-independent manner and activities of these protein kinases returned during recovery period to the control level. Inhibiting p38MAPK and ERK1/2 with the p38MAPK inhibitors (SB203580 and SB202190) and the MEK1/2 inhibitor (PD98059 and U0126) or with expression of dominant negative p38MAPK and ERK1/2 abolished SR-induced HSP72 synthesis and HSP70 promoter-driven CAT activity. However, sodium salicylate-induced HSF1-DNA binding was not affected by the p38MAPK inhibitor or the MEK1/2 inhibitor. These findings suggest that sodium salicylate partially activates HSF1 via ROS production and p38MAPK activation and the salicylate-induced inert HSF1 can be fully activated into a transcriptionally competent form by the ERK1/2 signaling pathways that are activated independently of ROS during SR.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Proteínas de Ligação a DNA/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Salicilato de Sódio/farmacologia , Fatores de Transcrição/metabolismo , Animais , DNA/metabolismo , Glioblastoma , Proteínas de Choque Térmico HSP90/metabolismo , Fatores de Transcrição de Choque Térmico , 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 , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Fosforilação , Regiões Promotoras Genéticas , Ratos , Células Tumorais Cultivadas , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
20.
Int J Mol Med ; 16(5): 833-9, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16211252

RESUMO

Salicylate and jasmonates are two different types of plant hormone that play critical roles in plant defense responses against insect herbivores and microbial pathogens, through activating defense genes. These two natural products have been shown to have similar activities in animal cells: the compounds are able to induce cell cycle arrest or apoptosis in a variety of human cancer cells including those of colon, prostate, breast, and leukemia, suggesting the chemicals may potentially be a novel class of anti-cancer drugs. Since sodium salicylate can induce the heat shock response in animals, we examined the effects of jasmonates on the heat shock response in C6 glioma cells. Here, we show that brief exposure to methyl jasmonate (MeJA), but not to jasmonic acid, induces heat shock protein 72 (HSP72), but not HSP73 and HSP90, via heat shock factor I (HSF1) activation in C6 glioma cells without affecting cell viability. Intracellular H2O2 and O2-, and mitochondrial ROS were prominently increased in response to 5 mM MeJA in C6 cells. MeJA-induced HSP72 expression, HSF1 DNA binding, and human HSP70 promoter-driven CAT activity were prevented by N-acetyl-L-cysteine (a general antioxidant), catalase (a specific antioxidant for H2O2), and sodium formate (an inhibitor of OH.), but not by Rac1 dominant negative mutant Rac1N17 and diphenyleneiodonium (a NADPH oxidase inhibitor), indicating that MeJA induces HSP72 expression though HSF1 that is activated via Rac1-NADPH oxidase-independent ROS production pathway. These results suggest that the plant stress hormones share the ability to induce heat shock response in animal cells.


Assuntos
Acetatos/farmacologia , Antineoplásicos/farmacologia , Neoplasias do Sistema Nervoso Central/metabolismo , Ciclopentanos/farmacologia , Proteínas de Ligação a DNA/metabolismo , Glioblastoma/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Fatores de Transcrição/metabolismo , Animais , Linhagem Celular Tumoral , Neoplasias do Sistema Nervoso Central/genética , Sequestradores de Radicais Livres/farmacologia , Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Glioblastoma/genética , Fatores de Transcrição de Choque Térmico , Humanos , 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 , Oxilipinas , Ratos , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA