Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Genes Cells ; 28(4): 288-306, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36788710

RESUMO

Ionizing radiation damages DNA and may lead to the development of cancer. Irradiation also generates reactive oxygen species (ROS) which cause damage to various biological molecules. Relatively low dose-rate irradiation causes less damage. However, the damage and its effects on cell fate are difficult to evaluate. To develop a method to analyze the damage and accompanying changes in physiology in cells irradiated by γ-rays at a relatively low dose-rate, we used the protein array technique to quantify marker proteins involved in the stress response and the regulation of cell growth and death. This method enabled efficient analyses of many replicates of experimental data on cell lysate samples. We detected relatively small changes in the levels of these proteins in the irradiated cells. Changes in protein levels suggested ROS production and DNA damage as well as cell cycle retardation and the progression of cellular senescence. Thus, our approach shows promise for analyzing the biological effects of relatively low dose-rate irradiation.


Assuntos
Senescência Celular , Dano ao DNA , Espécies Reativas de Oxigênio/metabolismo , Raios gama , Senescência Celular/genética , Diferenciação Celular
2.
Int J Mol Sci ; 23(23)2022 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-36499465

RESUMO

4-O-methylascochlorin (MAC) is a 4-fourth carbon-substituted derivative of ascochlorin, a compound extracted from a phytopathogenic fungus Ascochyta viciae. MAC induces apoptosis and autophagy in various cancer cells, but the effects of MAC on apoptosis and autophagy in cervical cancer cells, as well as how the interaction between apoptosis and autophagy mediates the cellular anticancer effects are not known. Here, we investigated that MAC induced apoptotic cell death of cervical cancer cells without regulating the cell cycle and promoted autophagy by inhibiting the phosphorylation of serine-threonine kinase B (Akt), mammalian target of rapamycin (mTOR), and 70-kDa ribosomal protein S6 kinase (p70S6K). Additional investigations suggested that Bcl-2/adenovirus E1B 19 kDa protein-interacting protein 3 (BNIP-3), but not Hypoxia-inducible factor 1 alpha (HIF-1α), is a key regulator of MAC-induced apoptosis and autophagy. BNIP-3 siRNA suppressed MAC-induced increases in cleaved- poly (ADP-ribose) polymerase (PARP) and LC3II expression. The pan-caspase inhibitor Z-VAD-FMK suppressed MAC-induced cell death and enhanced MAC-induced autophagy. The autophagy inhibitor chloroquine (CQ) enhanced MAC-mediated cell death by increasing BNIP-3 expression. These results indicate that MAC induces apoptosis to promote cell death and stimulates autophagy to promote cell survival by increasing BNIP-3 expression. This study also showed that co-treatment of cells with MAC and CQ further enhanced the death of cervical cancer cells.


Assuntos
Neoplasias do Colo do Útero , Feminino , Humanos , Neoplasias do Colo do Útero/genética , Linhagem Celular Tumoral , Autofagia , Apoptose , Cloroquina/farmacologia
3.
J Biochem Mol Toxicol ; 34(10): e22552, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32562591

RESUMO

A prior study identified that 4-O-methylascochlorin (MAC), a methylated derivative of ascochlorin (ASC) from the fungus Ascochyta viciae, activates autophagy in leukemia cells by suppressing c-Myc phosphorylation. However, the effects of MAC on autophagy in other cancer cells remain unknown. In the present study, we demonstrated that MAC activated autophagy in human glioblastoma. MAC increased expression of autophagy-related proteins, such as LC3-II and Beclin-1. Moreover, MAC stimulated AMP-activated protein kinase (AMPK) phosphorylation and suppressed phosphorylation of the mTOR, p70S6K, and 4EBP1. The well-known AMPK activator metformin increased LC3-II levels, which were augmented by MAC cotreatment. AMPK knockdown decreased LC3-II levels and inhibited MAC activation of autophagy. Furthermore, MAC suppression of c-Myc expression activated autophagy. Treatment with the c-MYC inhibitor, 10058-FA, induced autophagy, as did c-Myc small interfering RNA knockdown. These effects were augmented by MAC cotreatment. Taken together, these findings indicated that MAC induces autophagy in human glioblastoma by activating AMPK signaling and inhibiting c-Myc protein expression in human glioblastoma.


Assuntos
Adenilato Quinase/metabolismo , Autofagia/efeitos dos fármacos , Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , Proteínas Proto-Oncogênicas c-myc/antagonistas & inibidores , Terpenos/farmacologia , Animais , Proteína Beclina-1/metabolismo , Neoplasias Encefálicas/enzimologia , Linhagem Celular Tumoral , Regulação para Baixo , Ativação Enzimática , Glioblastoma/enzimologia , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo
4.
Biosci Biotechnol Biochem ; 83(12): 2244-2248, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31392931

RESUMO

Hypoxia-inducible factor-1 (HIF-1) is a transcription factor that plays essential roles in human diseases including cancer. The synthetic ascochlorin derivative 4-O-methylascochlorin stabilizes HIF-1α protein, and activates its transcriptional activity, resulting to induce gene expression of its downstream targets such as VEGF and GLUT-1. Here, we quantified protein level of HIF-1α in human osteosarcoma U2OS cells treated with ascochlorin-related compounds and typical HIF-1α stabilizers to characterize properties of HIF-1α stabilization by 4-O-methylascochlorin. Structure-activity relationship studies suggested that the aromatic moiety and hydrophobic substitution of the 4'-hydroxyl group are important for HIF-1α stabilization by ascochlorin-related compounds. 4-O-Methylascochlorin-induced HIF-1α stabilization was suppressed by ascorbic acid and compound C, but not by Fe(II), whereas ascorbic acid only suppressed HIF-1α stabilization by dimethyloxaloylglycine, an analog of the HIF-1 hydroxylase substrate. Fe(II) completely suppressed iron chelator-induced stabilization. These results suggest that ascochlorin-related compounds stabilize HIF-1α in a manner distinct from iron chelating or substrate competition.


Assuntos
Subunidade alfa do Fator 1 Induzível por Hipóxia/efeitos dos fármacos , Quelantes de Ferro/farmacologia , Oxigenases de Função Mista/metabolismo , Terpenos/farmacologia , Ligação Competitiva , Linhagem Celular Tumoral , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Relação Estrutura-Atividade , Especificidade por Substrato , Terpenos/química
5.
Gan To Kagaku Ryoho ; 46(3): 447-451, 2019 Mar.
Artigo em Japonês | MEDLINE | ID: mdl-30914582

RESUMO

Compared to other types of breast cancers, triple-negative breast cancer(TNBC)has poor prognosis. However, much work has been done towards establishing an effective therapeutic strategy. Vinorelbine(VNB)is an effective therapeutic agent for TNBC, however, the mechanism for its efficacy remains to be elucidated. We found that MX-1, a TNBC cell line, exhibits apoptosis and polyploidy upon VNB treatment. Neither apoptosis nor polyploidy were observed in other types of breast cancer cells upon VNB treatment. Furthermore, inhibitors of respiration, protein synthesis, and DNA synthesis suppressed apoptosis and polyploidy induced by VNB in MX-1 cells. Among microtubule toxins, clinically effective paclitaxel(PTX)and VNB had a greater effect than colchicine and nocodazole on polyploidy induction in MX-1 cells. These results suggest that VNB induces apoptosis in some types of TNBCs through the induction of polyploidy, which is, at least in part, the likely mechanism of its clinical efficacy.


Assuntos
Antineoplásicos Fitogênicos , Apoptose , Neoplasias de Mama Triplo Negativas , Vinorelbina , Antineoplásicos Fitogênicos/farmacologia , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Humanos , Microtúbulos , Poliploidia , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Vinorelbina/farmacologia
6.
J Cell Mol Med ; 22(12): 6345-6356, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30338933

RESUMO

4-O-methylascochlorin (MAC) is a derivative of ascochlorin, a prenyl-phenol compound antibiotic isolated from the fungus Ascochyta viciae. MAC induces caspase/poly (ADP-ribose) polymerase-mediated apoptosis in leukemia cells. However, the effects of MAC on autophagy in cancer cells and the underlying molecular mechanisms remain unknown. Here, we show that MAC induces autophagy in lung cancer cells. MAC significantly induced the expression of autophagy marker proteins including LC3-II, Beclin1, and ATG7. MAC promoted AMP-activated protein kinase (AMPK) phosphorylation and inhibited the phosphorylation of mammalian target of rapamycin (mTOR) and its downstream signalling proteins P70S6K and 4EBP1. The AMPK activator AICAR upregulated LC3-II expression through the AMPK/mTOR pathway similar to the effects of MAC. MAC-induced LC3-II protein expression was slightly reduced in AMPK siRNA transfected cells. MAC upregulated hypoxia-inducible factor-1α (HIF-1α) and BNIP3, which are HIF-1α-dependent autophagic proteins. Treatment with CoCl2 , which mimics hypoxia, induced autophagy similar to the effect of MAC. The HIF-1α inhibitor YC-1 and HIF-1α siRNA inhibited the MAC-induced upregulation of LC3-II and BNIP3. These results suggest that MAC induces autophagy via the AMPK/mTOR signalling pathway and by upregulating HIF-1α and BNIP3 protein expression in lung cancer cells.


Assuntos
Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Neoplasias Pulmonares/tratamento farmacológico , Proteínas de Membrana/genética , Proteínas Proto-Oncogênicas/genética , Terpenos/farmacologia , Antibacterianos/química , Antibacterianos/farmacologia , Apoptose/efeitos dos fármacos , Ascomicetos/química , Autofagia/efeitos dos fármacos , Hipóxia Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Proteínas Associadas aos Microtúbulos/genética , Fosforilação/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR/genética , Terpenos/química , Ativação Transcricional/efeitos dos fármacos
7.
J Cell Biochem ; 119(2): 2036-2047, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28833404

RESUMO

Numerous anti-cancer agents inhibit cell cycle progression via a p53-dependent mechanism; however, other genes such as the proto-oncogene c-Myc are promising targets for anticancer therapy. In the present study, we provide evidence that ascochlorin, an isoprenoid antibiotic, is a non-toxic anti-cancer agent that induces G1 cell cycle arrest and p21WAF1/CIP1 expression by downregulating of c-Myc protein expression. Ascochlorin promoted the G1 arrest, upregulated p53 and p21WAF1/CIP1 , and downregulated c-Myc in HCT116 cells. In p53-deficient cells, ascochlorin enhanced the expression of G1 arrest-related genes except p53. Small interfering RNA (siRNA) mediated c-Myc silencing indicated that the transcriptional repression of c-Myc was related to ascochlorin-mediated modulation of p21WAF1/CIP1 expression. Ascochlorin suppressed the stabilization of the c-Myc protein by inhibiting ERK and P70S6K/4EBP1 phosphorylation, whereas it had no effect on c-Myc degradation mediated by PI3K/Akt/GSK3ß. The ERK inhibitor PD98059 and siRNA-mediated ERK silencing induced G1 arrest and p21WAF1/CIP1 expression by downregulating c-Myc in p53-deficient cells. These results indicated that ascochlorin-induced G1 arrest is associated with the repression of ERK phosphorylation and c-Myc expression. Thus, we reveal a role for ascochlorin in inhibiting tumor growth via G1 arrest, and identify a novel regulatory mechanism for ERK/c-Myc.


Assuntos
Alcenos/farmacologia , Antibióticos Antineoplásicos/farmacologia , Neoplasias Colorretais/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Fenóis/farmacologia , Proteínas Proto-Oncogênicas c-myc/metabolismo , Pontos de Checagem do Ciclo Celular , Neoplasias Colorretais/tratamento farmacológico , Regulação para Baixo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células HCT116 , Humanos , Fosforilação/efeitos dos fármacos , Proto-Oncogene Mas
8.
J Cell Biochem ; 117(4): 978-87, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26399466

RESUMO

A natural compound C23 H32 O4 Cl, ascochlorin (ASC) isolated from an incomplete fungus, Ascochyta viciae has been known to have several biological activities as an antibiotic, antifungal, anti-cancer, anti-hypolipidemic, and anti-hypertension agent. In this study, anti-inflammatory activity has been investigated in lipopolysaccharide (LPS)-induced murine macrophage RAW 264.7 cells, since ASC has not been observed on the inflammatory events. The present study has clearly shown that ASC (1-50 µM) significantly suppressed the production of nitric oxide (NO) and prostaglandin E2 (PGE2 ) and decreased the gene expression of inducible NO synthase (iNOS) and cyclooxygenase-2 (COX-2) in a dose-dependent manner. Moreover, ASC inhibited the mRNA expression and the protein secretion of interleukin (IL)-1ß and IL-6 but not tumor necrosis factor (TNF)-α in LPS-stimulated RAW 264.7 macrophage cells. In addition, ASC suppressed nuclear translocation and DNA binding affinity of nuclear factor-κB (NF-κB). Furthermore, ASC down-regulated phospho-extracellular signal-regulated kinase 1/2 (p-ERK1/2) and p-p38. These results demonstrate that ASC exhibits anti-inflammatory effects in RAW 264.7 macrophage cells.


Assuntos
Alcenos/farmacologia , Anti-Inflamatórios não Esteroides/farmacologia , Ciclo-Oxigenase 2/genética , Lipopolissacarídeos/antagonistas & inibidores , Macrófagos/efeitos dos fármacos , Óxido Nítrico Sintase Tipo II/antagonistas & inibidores , Fenóis/farmacologia , Transdução de Sinais/efeitos dos fármacos , Alcenos/isolamento & purificação , Animais , Anti-Inflamatórios não Esteroides/isolamento & purificação , Linhagem Celular , Ciclo-Oxigenase 2/metabolismo , Dinoprostona/antagonistas & inibidores , Dinoprostona/biossíntese , Regulação da Expressão Gênica , Lipopolissacarídeos/farmacologia , Ativação de Macrófagos/efeitos dos fármacos , Macrófagos/citologia , Macrófagos/metabolismo , Camundongos , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , NF-kappa B/antagonistas & inibidores , NF-kappa B/genética , NF-kappa B/metabolismo , Óxido Nítrico/antagonistas & inibidores , Óxido Nítrico/biossíntese , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/metabolismo , Fenóis/isolamento & purificação , Transporte Proteico , Saccharomycetales/química , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
9.
Apoptosis ; 21(5): 657-68, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26922069

RESUMO

4-O-Methyl-ascochlorin (MAC) is a methylated derivative of the prenyl-phenol antibiotic ascochlorin, which was isolated from an incomplete fungus, Ascochyta viciae. Although the effects of MAC on apoptosis have been reported, the underlying mechanisms remain unknown. Here, we show that MAC promoted apoptotic cell death and downregulated c-Myc expression in K562 human leukemia cells. The effect of MAC on apoptosis was similar to that of 10058-F4 (a c-Myc inhibitor) or c-Myc siRNA, suggesting that the downregulation of c-Myc expression plays a role in the apoptotic effect of MAC. Further investigation showed that MAC downregulated c-Myc by inhibiting protein synthesis. MAC promoted the phosphorylation of AMP-activated protein kinase (AMPK) and inhibited the phosphorylation of mammalian target of rapamycin (mTOR) and its target proteins, including p70S6 K and 4E-BP-1. Treatment of cells with AICAR (an AMPK activator), rapamycin (an mTOR inhibitor), or mTOR siRNA downregulated c-Myc expression and induced apoptosis to a similar extent to that of MAC. These results suggest that the effect of MAC on apoptosis induction in human leukemia cells is mediated by the suppression of c-Myc protein synthesis via an AMPK/mTOR-dependent mechanism.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Antineoplásicos/farmacologia , Apoptose , Proteínas Proto-Oncogênicas c-myc/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR/antagonistas & inibidores , Terpenos/farmacologia , Linhagem Celular , Linhagem Celular Tumoral , Pontos de Checagem da Fase G1 do Ciclo Celular , Humanos , Células K562 , Biossíntese de Proteínas/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-myc/biossíntese , Proteínas Proto-Oncogênicas c-myc/metabolismo , Serina-Treonina Quinases TOR/metabolismo
10.
Arch Biochem Biophys ; 583: 79-86, 2015 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-26271443

RESUMO

Obesity increases the risk of developing many chronic diseases, including type 2 diabetes and certain cancers, and is thereby associated with premature death. The present study was conducted to identify the inhibitory effect of the ascochlorin derivative 4-O-methylascochlorin (MAC) on the differentiation of 3T3-L1 preadipocytes. MAC suppressed the differentiation of 3T3-L1 preadipocytes and inhibited the expression of adipocyte differentiation marker genes, FABP4, PPARγ and C/EBPα. In addition, we found that the inhibitory effects of MAC on differentiation of 3T3-L1 preadipocytes were caused by suppression of mTORC1 via inhibition of mTOR/p70S6K/4E-BP1 phosphorylation and activation of Raptor phosphorylation. MAC also regulated the PPARγ expression and the mTORC1 activation by increasing AMPK phosphorylation and inhibiting PI3K/Akt, which suggest that MAC suppresses the differentiation of 3T3-L1 adipocytes by regulating the AMPK- and PI3K-mTOR-PPARγ signaling pathways. Furthermore, animal model results showed that the phosphorylation of AMPK was enhanced in the liver of C57BL/6 mice intraperitoneally injected with MAC. These results indicate that MAC could be a therapeutic agent for obesity involving PPARγ and AMPK.


Assuntos
Adenilato Quinase/metabolismo , Adipócitos/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , PPAR gama/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo , Terpenos/farmacologia , Células 3T3-L1 , Adipócitos/citologia , Adipócitos/metabolismo , Animais , Camundongos , Fosforilação
11.
Toxicol Appl Pharmacol ; 273(3): 542-50, 2013 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-24096035

RESUMO

Hypoxia-inducible factor (HIF)-1 plays an important role in tumor progression, angiogenesis and metastasis. In this study, we investigated the potential molecular mechanisms underlying the anti-angiogenic effect of ascofuranone, an isoprenoid antibiotic from Ascochyta viciae, in epidermal growth factor (EGF)-1 responsive human breast cancer cells. Ascofuranone significantly and selectively suppressed EGF-induced HIF-1α protein accumulation, whereas it did not affect the expression of HIF-1ß. Furthermore, ascofuranone inhibited the transcriptional activation of vascular endothelial growth factor (VEGF) by reducing protein HIF-1α. Mechanistically, we found that the inhibitory effects of ascofuranone on HIF-1α protein expression are associated with the inhibition of synthesis HIF-1α through an EGF-dependent mechanism. In addition, ascofuranone suppressed EGF-induced phosphorylation of Akt/mTOR/p70S6 kinase, but the phosphorylation of ERK/JNK/p38 kinase was not affected by ascofuranone. These results suggest that ascofuranone suppresses EGF-induced HIF-1α protein translation through the inhibition of Akt/mTOR/p70S6 kinase signaling pathways and plays a novel role in the anti-angiogenic action.


Assuntos
Inibidores da Angiogênese/farmacologia , Fator de Crescimento Epidérmico/farmacologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/biossíntese , Sesquiterpenos/farmacologia , Transdução de Sinais/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Sobrevivência Celular , Imunoprecipitação da Cromatina , Feminino , Regulação da Expressão Gênica , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/antagonistas & inibidores , Células MCF-7 , Camundongos , Camundongos Endogâmicos C57BL , Neovascularização Patológica/tratamento farmacológico , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Ativação Transcricional , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores
12.
J Cell Biochem ; 113(4): 1302-13, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22109717

RESUMO

Ascochlorin, a non-toxic prenylphenol compound derived from the fungus Ascochyta viciae, has been shown recently to have anti-cancer effects on various human cancer cells. However, the precise molecular mechanism of this anti-cancer activity remains to be elucidated. Here, we investigated the effects of ascochlorin on hypoxia-inducible factor-1α (HIF-1α) and vascular endothelial growth factor (VEGF) expression in human epidermoid cervical carcinoma CaSki cells. Ascochlorin inhibited epidermal growth factor (EGF)-induced HIF-1α and VEGF expression through multiple potential mechanisms. First, ascochlorin selectively inhibited HIF-1α expression in response to EGF stimulation, but not in response to hypoxia (1% O(2)) or treatment with a transition metal (CoCl(2)). Second, ascochlorin inhibited EGF-induced ERK-1/2 activation but not AKT activation, both of which play essential roles in EGF-induced HIF-1α protein synthesis. Targeted inhibition of epidermal growth factor receptor (EGFR) expression using an EGFR-specific small interfering RNA (siRNA) diminished HIF-1α expression, which suggested that ascochlorin inhibits HIF-1α expression through suppression of EGFR activation. Finally, we showed that ascochlorin functionally abrogates in vivo tumor angiogenesis induced by EGF in a Matrigel plug assay. Our data suggest that ascochlorin inhibits EGF-mediated induction of HIF-1α expression in CaSki cells, providing a potentially new avenue of development of anti-cancer drugs that target tumor angiogenesis.


Assuntos
Alcenos/farmacologia , Receptores ErbB/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Fenóis/farmacologia , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Transdução de Sinais/efeitos dos fármacos , Neoplasias do Colo do Útero/metabolismo , Fator A de Crescimento do Endotélio Vascular/fisiologia , Sequência de Bases , Western Blotting , Linhagem Celular Tumoral , Primers do DNA , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Neovascularização Patológica/prevenção & controle , RNA Interferente Pequeno , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Neoplasias do Colo do Útero/irrigação sanguínea , Neoplasias do Colo do Útero/patologia
13.
Toxicol In Vitro ; 81: 105342, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35248696

RESUMO

4-O-Methyl-ascochlorin (MAC), a derivative of the prenyl-phenol antibiotic ascochlorin, promotes accumulation of HIF-1α. In this study, we investigated the molecular mechanisms of the effect of MAC on cell migration and mesenchymal epithelial transition (EMT) processes in breast cancer cells. MAC upregulated cell motility and migration regardless of cell viability, and promoted EMT features by regulating EMT-related proteins and transcription. In addition, the MAC-induced increase in the EMT was closely related to activation of HIF-1α expression. However, the MAC-induced EMT was not associated with AMPK phosphorylation or intracellular ROS, which stimulate HIF-1α expression. Similarly, HIF-1α-mediated autophagy induced by MAC was not related to EMT-related proteins. Inhibition of HIF-1α activity inhibited MAC-stimulated cell migration and increased MAC-induced cell death, indicating that HIF-1α activation is important for MAC-mediated cell migration and survival in breast cancer cells. Together, these results suggest that MAC can be used to investigate the link between HIF-1α activation and other oncogenes or tumor suppressors in breast cancer cells.


Assuntos
Neoplasias da Mama , Transição Epitelial-Mesenquimal , Linhagem Celular Tumoral , Movimento Celular , Sobrevivência Celular , Feminino , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Terpenos
15.
Biochem Biophys Res Commun ; 406(3): 353-8, 2011 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-21329651

RESUMO

Chemopreventive or anticancer agents induce cancer cells to apoptosis through the activation of adenosine AMP-activated protein kinase (AMPK), which plays a major role as energy sensors under ATP-deprived condition or ROS generation. In this study, we compared the effects of ascochlorin (ASC), from the fungus Ascochyta viciae, and its derivatives on AMPK activity. We also examined a regulatory mechanism for hypoxia-inducible factor-1α (HIF-1α) stabilization in response to 4-O-methylascochlorin (MAC). We found that AMPK activation was mainly involved with MAC, but not ASC and 4-O-carboxymethylascochlorin (AS-6), indicating that the substitution of 4-O-methyl group from 4-O-hydroxyl group of ASC is important in the activation of AMPK and the expression of HIF-1α. MAC-stabilized HIF-1α via AMPK activation triggered by lowering the intracellular ATP level, not by ROS generation, increases glucose uptake and the expression of vascular endothelial growth factor (VEGF) and glucose transporter 1 (GLUT-1), major target genes of HIF-1α. Moreover, MAC-induced AMPK activity suppressed survival factors, including mTOR and ERK1/2 or translational regulators, including p70S6K and 4E-BP1. Our data suggest that AMPK is a key determinant of MAC-induced HIF-1α expression in response to energy stress, further implying its involvement in MAC-induced apoptosis.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Alcenos/farmacologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Fenóis/farmacologia , Terpenos/farmacologia , Proteínas Quinases Ativadas por AMP/genética , Trifosfato de Adenosina/metabolismo , Alcenos/química , Apoptose , Linhagem Celular Tumoral , Técnicas de Silenciamento de Genes , Humanos , Metilação , Fenóis/química , Regiões Promotoras Genéticas/efeitos dos fármacos , Estabilidade Proteica , RNA Interferente Pequeno/genética , Espécies Reativas de Oxigênio/antagonistas & inibidores , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Serina-Treonina Quinases TOR/metabolismo
16.
Biol Pharm Bull ; 34(7): 1001-4, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21720004

RESUMO

Worldwide, lung cancer is the most common form of cancer and often has a poor prognosis. Establishment of effective therapies for lung cancer is a major concern in clinical cancer research. We compared the cytotoxic effects of chemotherapeutic agents including cisplatin, 5-fluorouracil, vinorelbine and cladribine, on a human lung cancer cell line, A549, and its derivative transfected with the DNase γ gene. We observed selective cytotoxicity of cladribine on the DNase γ-expressing sub-cell line of A549. Cladribine induces selective apoptosis in DNase γ-expressing A549 cells, which depends on activation of caspases. These results suggest that a combination therapy that includes cladribine along with the introduction of DNase γ has potential as a new therapeutic strategy for lung cancer.


Assuntos
Adenocarcinoma/patologia , Apoptose/efeitos dos fármacos , Cladribina/farmacologia , Endodesoxirribonucleases/metabolismo , Neoplasias Pulmonares/patologia , Adenocarcinoma/enzimologia , Linhagem Celular Tumoral , Citometria de Fluxo , Humanos , Neoplasias Pulmonares/enzimologia
17.
Int Immunopharmacol ; 90: 107184, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33316741

RESUMO

Inflammation is implicated in various diseases, such as inflammatory bowel disease and cancer. Ascochlorin (ASC) and its derivatives have been shown to modulate inflammatory responses in many previous studies. However, the effects of 4-O-methylascochlorin (MAC), one of the ASC derivatives, on inflammatory responses have yet to be reported. In addition, the consequences of chemical modification of ASC on protein signaling and immunity have yet to be fully understood. The fourth carbon in MAC is methylated, which may result in modulation of immune response differently compared with ASC. Hence, we have investigated the role of MAC in inflammatory response induced by lipopolysaccharide in murine macrophage cells. Here, we found that MAC treatment decreased the inflammatory response by murine macrophages. When murine macrophages were treated with MAC, the transcription and translation of various pro-inflammatory indicators such as iNOS and COX-2 decreased. In addition, the ELISA results showed that the expression of TNF-α, IL-6, and IL-1ß, which are pro-inflammatory cytokines, was successfully decreased by MAC. Such effects of MAC appear to be mediated via downregulation of MAPK signaling and the transactivational activity of NF-κB. Lipopolysaccharide upregulates MAPK protein phosphorylation and NF-κB translocation, which in turn enhances the transactivation of genes related to NF-κB. Such results of lipopolysaccharide were attenuated by MAC. Collectively, our results indicate that MAC alleviated the inflammatory responses induced by lipopolysaccharide in murine macrophages successfully by modulating MAPK signaling pathway and NF-κB-related genes. This study shows that MAC, similar to other ASC derivatives, can potentially be used therapeutically to reduce the harmful damage induced by prolonged inflammation. In addition, the structural differences between ASC and its derivatives as well as their effect on intracellular signaling will also be discussed.


Assuntos
Anti-Inflamatórios/farmacologia , Mediadores da Inflamação/metabolismo , Inflamação/prevenção & controle , Lipopolissacarídeos/farmacologia , Macrófagos/efeitos dos fármacos , Terpenos/farmacologia , Animais , Ciclo-Oxigenase 2/metabolismo , Citocinas/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Inflamação/imunologia , Inflamação/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , NF-kappa B/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Fosforilação , Células RAW 264.7
18.
Biochem Biophys Res Commun ; 396(4): 967-72, 2010 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-20460103

RESUMO

The current study presents that ascofuranone isolated from a phytopathogenic fungus, Ascochyta viciae, has antitumor activity against various transplantable tumors and a considerable hypolipidemic activity. AMP-activated protein kinase (AMPK) plays a critical role in cellular glucose and lipid homeostasis. We found that ascofuranone improves ER stress-induced insulin resistance by activating AMPK through the LKB1 pathway. In L6 myotube cells, ascofuranone treatment increased the phosphorylation of the Thr-172 residue of the AMPK alpha subunit and the Ser-79 subunit of acetyl-CoA carboxylase (ACC) and cellular glucose uptake. Ascofuranone-induced phosphorylation of AMPK and ACC was not increased in A549 cells lacking LKB1. Interestingly, ascofuranone treatment also improved insulin signaling impaired by ER stress in L6 myotube cells. These effects were all reversed by pretreatment with Compound C, an AMPK inhibitor or with adenoviral-mediated dominant-negative AMPK alpha 2. Taken together, these results indicated that ascofuranone-mediated enhancement of glucose uptake and reduction of impaired insulin sensitivity in L6 cells is predominantly accomplished by activating AMPK, thereby mediating beneficial effects in type 2 diabetes and insulin resistance.


Assuntos
Proteínas Quinases Ativadas por AMP/biossíntese , Antifúngicos/farmacologia , Retículo Endoplasmático/efeitos dos fármacos , Antagonistas da Insulina/farmacologia , Resistência à Insulina , Sesquiterpenos/farmacologia , Estresse Fisiológico , Animais , Linhagem Celular , Retículo Endoplasmático/enzimologia , Ativação Enzimática , Glucose/metabolismo , Humanos , Insulina/farmacologia , Fibras Musculares Esqueléticas/efeitos dos fármacos , Fibras Musculares Esqueléticas/enzimologia , Ratos
19.
World J Surg ; 34(9): 2197-203, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20458581

RESUMO

BACKGROUND: Skin-sparing partial mastectomy (SSPM) has yet to be investigated as a breast-conserving therapy for early-stage breast cancer. We report the clinical outcomes for video-assisted SSPM (VA-SSPM) with immediate breast reconstruction using autogenous tissue. METHODS: VA-SSPM is indicated for early-stage breast cancer arising in the upper-outer or lower-outer quadrant without skin involvement. An incision is placed along the midaxillary line, and SSPM is performed under endoscopic guidance using subcutaneous tunneling and lifting methods. Through the same incision, a latissimus dorsi muscle flap is harvested for breast reconstruction. From January 2000 to October 2007, 168 patients (Tis, n = 24; T1, n = 37; T2, n = 107) underwent VA-SSPM, and morbidity, curability, and postoperative patient satisfaction were investigated. RESULTS: Postoperative complications included skin necrosis (2.4%, n = 4) and muscle flap necrosis (0.6%, n = 1), but no severe complications were observed. After a mean follow-up of 58.6 months, eight patients (4.8%) experienced local recurrence. Sixty-month distant metastasis-free survival rates for Tis, T1, and T2 were 100%, 97%, and 83.3%, respectively, with an overall rate of 88.4%. Furthermore, overall survival rates for Tis, T1, and T2 were 100%, 94.1%, and 94.4%, respectively, with an overall survival rate of 95% for all patients. A patient satisfaction survey showed that 81.6% of patients evaluated the surgery as "good." CONCLUSIONS: VA-SSPM for early-stage breast cancer improves cosmetic results and achieves high patient satisfaction without increasing local or distant organ recurrence. This method offers a useful local therapy for early-stage breast cancer.


Assuntos
Neoplasias da Mama/cirurgia , Mamoplastia , Mastectomia Segmentar/métodos , Cirurgia Vídeoassistida , Adulto , Feminino , Humanos , Mamoplastia/métodos , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/epidemiologia , Satisfação do Paciente , Retalhos Cirúrgicos , Resultado do Tratamento
20.
Biol Pharm Bull ; 33(4): 568-71, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20410587

RESUMO

A peptide antibiotic, edeine B(1), exerts a lethal action in Bacillus subtilis causing filamentous morphology. This antibiotic assumes to inhibit cell division by interacting with FtsZ and inhibiting FtsZ polymerization. The temperature-sensitive mutant ftsZ ts1 was shown to be hypersensitive to the antibiotic as compared to the parent 168 with respect to its lethal action and the sensitivity to the antibiotic of the revertant of ftsZ ts1 was shown to be intermediate between those of the parent 168 and the ftsZ ts1. Alteration of FtsZ sequence may be responsible for sensitivity to edeine B(1). The residues at 240, 278, 345 and 346 in the FtsZ sequence of the parent 168 were A240, A278, D345 and A346. Those of ftsZ ts1 were V240, V278, E345 and P346. Those of the revertant of ftsZ ts1 were A240, A278, E345 and P346. The difference in sensitivity to edeine B(1) among these strains is presumably due to the difference in the residues at 240, 278, 345 and 346 in the FtsZ sequence. The sequential events of the inhibition of FtsZ assembly and the inhibition of protein biosynthesis by edeine B(1) may progress synergistically, resulting in cell death.


Assuntos
Antibacterianos/farmacologia , Bacillus subtilis/efeitos dos fármacos , Proteínas de Bactérias/biossíntese , Divisão Celular/efeitos dos fármacos , Proteínas do Citoesqueleto/biossíntese , Edeína/análogos & derivados , Espermidina/análogos & derivados , Antibacterianos/isolamento & purificação , Bacillus subtilis/genética , Bacillus subtilis/fisiologia , Proteínas de Bactérias/genética , Sequência de Bases , Divisão Celular/genética , Proteínas do Citoesqueleto/genética , Edeína/isolamento & purificação , Edeína/farmacologia , Mutação , Espermidina/isolamento & purificação , Espermidina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA