Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Zhongguo Zhong Xi Yi Jie He Za Zhi ; 36(11): 1340-1344, 2016 Nov.
Artigo em Zh | MEDLINE | ID: mdl-30641628

RESUMO

Objective To observe the effect of Fuzheng Kang'ai Recipe (FKR) combined ge- fitinib on the proliferation and apoptosis of lung cancer A549 cells , and to study its potential synergistic mechanish with gefitinib. Methods The effects of FKR (0. 211, 0. 316, 0. 474, 0. 711, 1. 067, 1. 600, 2. 400, 3. 600 mg/mL) combined gefitinib (3. 95, 5. 92, 8. 18, 13. 33, 20. 00, 30. 00, 45. 00, 67. 50 µmol/ L) on the proliferation of A549 cells were detected by MTT assay. The apoptosis of A549 cells in the control group (complete culture medium) , FKR (1. 6 mg/mL) , gefitinib (45 µmol/L) , and FKR plus gefitinib (1. 6 mg/mL +45 µmol/L) were detected by flow cytometry (FCM). Their expressions of epidermal growth factor receptor (EGFR) , phosphorylating epidermal growth factor receptor ( p-EGFR) , enhancer of zeste homolog 2 (EZH2), peroxisome proliferator-activated receptor-γ ( PPAR-γ) , and P53 protein in A549 cells were detected by Western blot. Results Both FKR and gefitinib could inhibit the proliferation of A549 cells. The apoptotic rate was 12. 6% ±4. 5% in the FKR combined gefitinib group, obviously higher than that of the FKR group (4. 6% ± 0. 7%) and the gefitinib group (7. 8% ± 2. 7%) , showing statistical difference (P <0. 05). Compared with the control group, the expressions of p-EGFR and EZH2 were sig- nificantly down-regulated (P <0. 05) , the expressions of PPAR-γ and P53 protein were up-regulated in the FKR combined gefitinib group (P <0. 05); the expression of EZH2 was down-regulated in the gefitinib group and the FKR group (P <0. 05) ; the expression of PPAR-y was up-regulated in the FKR group (P < 0. 05). Compared with the gefitinib group, the expression of p-EGFR was down-regulated, and the expression of PPAR-γ was up-regulated in the FKR combined gefitinib group (both P <0. 05). Compared with the FKR group, the expression of p-EGFR was down-regulated in the FKR combined gefitinib group (P < 0. 05). Conclusions Combination of FKR and gefitinib could significantly inhibit the proliferation and growth of A549 cells,and induce cell apoptosis. Its potential synergistic mechanism of anti-tumor activities might be associated with down-regulating mRNA expressions of p-EGFR and EZH2, and up-regulating protein expressions of PPAR-y and P53.


Assuntos
Antineoplásicos , Apoptose , Carcinoma Pulmonar de Células não Pequenas , Medicamentos de Ervas Chinesas , Gefitinibe , Neoplasias Pulmonares , Células A549 , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Linhagem Celular Tumoral , Proliferação de Células , Medicamentos de Ervas Chinesas/uso terapêutico , Receptores ErbB , Gefitinibe/uso terapêutico , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Quinazolinas
2.
Carcinogenesis ; 35(11): 2544-54, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25194164

RESUMO

Accumulating evidence suggests that ß-catenin signaling in breast cancer stem cells (CSCs) is closely correlated to chemoresistance and adenosine triphosphate (ATP)-binding cassette subfamily G2 (ABCG2) expression. Targeting the aberrant ß-catenin signaling in CSCs has become a promising strategy to improve chemosensitivity in cancer treatment. In a pilot screening study, we found that the natural compound isoliquiritigenin (ISL) blocked ß-catenin transcription activity with the highest inhibition ratio. Here, we investigated the chemosensitizing effects of ISL on breast CSCs and the underlying mechanisms regulating the ß-catenin pathway. ISL could have synergistic effects with chemotherapeutic drugs to inhibit breast cancer cell proliferation and colony formation. In addition, ISL could significantly limit the side population and CSC ratios in breast cancer cells, accompanied by inhibited self-renewal and multidifferentiation abilities. A mechanistic study revealed that ISL could inhibit ß-catenin/ABCG2 signaling by activating the proteasome degradation pathway. The drug affinity responsive target stability strategy further identified GRP78 as the direct target of ISL. Subsequent molecular docking analysis and functional studies demonstrated that ISL could dock into the ATP domain of GRP78 and thereby inhibit its ATPase activity, resulting in its dissociation from ß-catenin. An in vivo study also suggested that ISL could chemosensitize breast CSCs via the GRP78/ß-catenin/ABCG2 pathway, with little toxicity in normal tissues and mammary stem cells. Taken together, the data from this study not only suggest ISL as a natural candidate to enhance breast CSC chemosensitivity but also highlight the significance of GRP78 in mediating cancer drug resistance and ß-catenin signaling in CSCs.


Assuntos
Transportadores de Cassetes de Ligação de ATP/biossíntese , Neoplasias da Mama/genética , Proteínas de Choque Térmico/metabolismo , Proteínas de Neoplasias/biossíntese , beta Catenina/biossíntese , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Animais , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Proliferação de Células , Chalconas/administração & dosagem , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Chaperona BiP do Retículo Endoplasmático , Feminino , Proteínas de Choque Térmico/química , Proteínas de Choque Térmico/genética , Humanos , Células MCF-7 , Camundongos , Simulação de Acoplamento Molecular , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Transdução de Sinais/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto , beta Catenina/genética
3.
Sci Total Environ ; 798: 149220, 2021 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-34315054

RESUMO

Continuous single tillage has the potential to increase greenhouse gas (GHG) emissions and decrease the accumulation of soil organic carbon (SOC), thus increasing carbon footprints (CFs). However, in a wheat-maize cropping system, limited information was available about the effects of strategic tillage on CFs. Thus, a four-year field experiment was conducted, including continuous rotary tillage (RT), continuous no-till (NT), RT + subsoiling (RS), and NT + subsoiling (NS), to investigate the effects of NS (strategic tillage) on the unit area and unit yield. The results showed that CO2 emission was the highest contributor to CFs (73.92%) in a winter wheat-summer maize cropping system, following the order of NS < NT < RS < RT. The direct N2O emissions from fertilizers and residues were 4.43-4.51 t CO2-eq ha-1 yr-1 during the wheat and maize seasons, and indirect N2O emissions from irrigation and fertilizer inputs had a proportion of >80% from total agricultural inputs. The differences in SOC storage significantly affected the CFs. Although the NS treatment increased the amount of GHG emissions from the residues returned and consumption of diesel, the enhancement of SOC storage by deeper SOC increased. Thus, lower area-scaled CFs were observed in the NS treatment. Furthermore, a higher grain yield and an annual change of SOC storage compared with other treatments were observed under the NS system, which helped to reduce the CFs. The yield-scaled CFs followed the order of RT > RS > NT > NS when considering the changes in SOC storage. Therefore, the NS treatment resulted in a higher grain yield and SOC sequestration with lower CFs, and thus, it could be recommended as the best tillage method to achieve sustainable production and environmental balance in a wheat-maize cropping system.


Assuntos
Triticum , Zea mays , Agricultura , Carbono , Pegada de Carbono , China , Óxido Nitroso/análise , Solo
4.
Am J Respir Cell Mol Biol ; 43(6): 684-91, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20081050

RESUMO

The matrix glycoprotein, fibronectin, stimulates the proliferation of non-small cell lung carcinoma in vitro through α5ß1 integrin receptor-mediated signals. However, the true role of fibronectin and its receptor in lung carcinogenesis in vivo remains unclear. To test this, we generated mouse Lewis lung carcinoma cells stably transfected with short hairpin RNA shRNA targeting the α5 integrin subunit. These cells were characterized and tested in proliferation, cell adhesion, migration, and soft agar colony formation assays in vitro. In addition, their growth and metastatic potential was tested in vivo in a murine model of lung cancer. We found that transfected Lewis lung carcinoma cells showed decreased expression of the α5 gene, which was associated with decreased adhesion to fibronectin and reduced cell migration, proliferation, and colony formation when compared with control cells and cells stably transfected with α2 integrin subunit in vitro. C57BL/6 mice injected with α5-silenced cells showed lower burden of implanted tumors, and a dramatic decrease in lung metastases resulting in higher survival as compared with mice injected with wild-type or α2 integrin-silenced cells. These observations reveal that recognition of host- and/or tumor-derived fibronectin via α5ß1 is important for tumor growth both in vitro and in vivo, and unveil α5ß1 as a potential target for the development of anti-lung cancer therapies.


Assuntos
Carcinoma Pulmonar de Lewis/metabolismo , Carcinoma Pulmonar de Lewis/patologia , Progressão da Doença , Integrina alfa5beta1/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Ágar , Animais , Adesão Celular , Movimento Celular , Proliferação de Células , Fibronectinas/metabolismo , Inativação Gênica , Injeções Intravenosas , Injeções Subcutâneas , Camundongos , Camundongos Endogâmicos C57BL , Metástase Neoplásica , Transplante de Neoplasias , RNA Interferente Pequeno/metabolismo , Análise de Sobrevida , Ensaio Tumoral de Célula-Tronco
5.
Mol Cancer Res ; 7(1): 108-17, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19147542

RESUMO

We previously showed that synthetic peroxisome proliferator-activated receptor gamma (PPARgamma) ligands inhibit non-small cell lung carcinoma (NSCLC) cell growth through multiple signaling pathways. Here, we show that dietary compounds, such as fish oil (which contains certain kinds of fatty acids like omega3 and omega6 polyunsaturated fatty acids), also inhibit NSCLC cell growth by affecting PPARgamma and by inhibiting the expression of integrin-linked kinase (ILK). Exogenous expression of ILK overcame, whereas silencing ILK enhanced the inhibitory effect of fish oil on cell growth. The inhibitor of p38 mitogen-activated protein kinase, SB239023, abrogated the inhibitory effect of fish oil on ILK expression, whereas the inhibitor of extracellular signal-regulated kinase, PD98059, had no effect. Transient transfection experiments showed that fish oil reduced ILK promoter activity, and this effect was abolished by AP-2alpha small interfering RNA and SB239023 and by deletion of a specific portion of the ILK gene promoter. Western blot analysis and gel mobility shift assay showed that fish oil significantly induced AP-2alpha protein expression and AP-2 DNA-binding activity in the ILK gene promoter and that this was dependent on PPARgamma activation. Blockade of AP-2alpha abrogated the effect of fish oil on ILK expression and on cell growth, whereas exogenous expression of AP-2alpha enhanced cell growth in the setting of fish oil exposure. Taken together, these findings show that fish oil inhibits ILK expression through activation of PPARgamma-mediated and p38 mitogen-activated protein kinase-mediated induction of AP-2alpha. In turn, this leads to inhibition of NSCLC cell proliferation. This study unveils a novel mechanism by which fish oil inhibits human lung cancer cell growth.


Assuntos
Carcinoma/patologia , Divisão Celular/efeitos dos fármacos , Óleos de Peixe/farmacologia , Neoplasias Pulmonares/patologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Carcinoma/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Óleos de Peixe/uso terapêutico , Deleção de Genes , Humanos , Neoplasias Pulmonares/genética , Regiões Promotoras Genéticas/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Serina-Treonina Quinases/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/genética , RNA Neoplásico/genética , RNA Interferente Pequeno/genética , Transfecção
6.
Mol Cancer Ther ; 8(1): 110-8, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19139119

RESUMO

We and others have shown previously that nicotine, a major component of tobacco, stimulates non-small cell lung carcinoma (NSCLC) proliferation through nicotinic acetylcholine receptor (nAChR)-mediated signals. Activation of peroxisome proliferator-activated receptor gamma (PPARgamma) has been shown to inhibit NSCLC cell growth, but the exact mechanisms responsible for this effect remain incompletely defined. Herein, we show that nicotine induces NSCLC cell proliferation in part through alpha4 nAChR, prompting us to explore the effects of rosiglitazone, a synthetic PPARgamma ligand, on the expression of this receptor. Rosiglitazone inhibited the expression of alpha4 nAChR, but this effect was through a PPARgamma-independent pathway, because GW9662, an antagonist of PPARgamma, and the transfection of cells with PPARgamma small interfering RNA failed to abolish the response. The inhibitory effect of rosiglitazone on alpha4 nAChR expression was accompanied by phosphorylation of p38 mitogen-activated protein kinase and extracellular signal-regulated kinase 1/2 and down-regulation of Akt phosphorylation. These signals mediated the inhibitory effects of rosiglitazone on alpha4 nAChR expression because chemical inhibitors prevented the effect. Rosiglitazone was also found to stimulate p53, a tumor suppressor known to mediate some of the effects of nicotine. Interestingly, p53 up-regulation was needed for rosiglitazone-induced inhibition of alpha4 nAChR. Thus, rosiglitazone inhibits alpha4 nAChR expression in NSCLC cells through activation of extracellular signal-regulated kinase and p38 mitogen-activated protein kinase, which triggers induction of p53. Finally, like others, we found that nicotine stimulated the expression of alpha4 nAChR. This process was also inhibited by rosiglitazone through similar pathways.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/metabolismo , Neoplasias Pulmonares/metabolismo , Receptores Nicotínicos/metabolismo , Tiazolidinedionas/farmacologia , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Neoplasias Pulmonares/patologia , PPAR gama/metabolismo , Rosiglitazona , Transdução de Sinais/efeitos dos fármacos , Proteína Supressora de Tumor p53/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
7.
Am J Respir Cell Mol Biol ; 40(3): 325-31, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18776129

RESUMO

We previously demonstrated that a selective agonist of peroxisome proliferator-activated receptor beta/delta (PPARbeta/delta), GW501516, stimulated human non-small cell lung carcinoma (NSCLC) growth, partly through inhibition of phosphatase and tensin homolog deleted on chromosome 10 expression. Here, we show that GW501516 also decreases the phosphorylation of AMP-activated protein kinase alpha (AMPKalpha), a major regulator of energy metabolism. This was mediated through specific activation of PPARbeta/delta, as a PPARbeta/delta small interfering RNA inhibited the effect. However, AMPKalpha did not mediate the growth-promoting effects of GW501516, as silencing of AMPKalpha did not inhibit GW501516-induced cell proliferation. Instead, we found that GW501516 stimulated peroxisome proliferator-activated receptor coactivator gamma (PGC)-1alpha, which activated the phosphatidylinositol 3 kinase (PI3-K)/Akt mitogenic pathway. An inhibitor of PI3-K, LY294002, had no effect on PGC-1alpha, consistent with PGC-1alpha being upstream of PI3-K/Akt. Of note, an activator of AMPKalpha, 5-amino-4-imidazole carboxamide riboside, inhibited the growth-promoting effects of GW501516, suggesting that although AMPKalpha is not responsible for the mitogenic effects of GW501516, its activation can oppose these events. This study unveils a novel mechanism by which GW501516 and activation of PPARbeta/delta stimulate human lung carcinoma cell proliferation, and suggests that activation of AMPKalpha may oppose this effect.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/metabolismo , Proteínas de Choque Térmico/metabolismo , Neoplasias Pulmonares/metabolismo , PPAR delta/agonistas , PPAR delta/metabolismo , PPAR beta/agonistas , PPAR beta/metabolismo , Tiazóis/metabolismo , Fatores de Transcrição/metabolismo , Proteínas Quinases Ativadas por AMP/metabolismo , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Ativação Enzimática , Proteínas de Choque Térmico/genética , Humanos , PPAR delta/genética , PPAR beta/genética , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Ribonucleotídeos/metabolismo , Transdução de Sinais/fisiologia , Fatores de Transcrição/genética
8.
Methods Mol Biol ; 512: 309-23, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19347285

RESUMO

Lung carcinoma remains one of the most common malignant tumors in the world despite recent advancements in the development of new chemotherapeutic agents for its treatment. Therefore, novel approaches for drug target discovery play an important role in the effort to help extend its dismal 5-year survival rate (<15%). Many mechanisms contribute to oncogenic transformation in carcinoma cells in the lung and recent evidence indicates that the overproduction of prostaglandin E(2) (PGE(2)), and the prostag-landin E(2) receptor subtype, EP4, promote the growth and progression of human nonsmall cell lung carcinoma (NSCLC), the most common lung carcinoma. Peroxisome proliferator-activated receptor beta/ delta (PPARbeta/delta), one of the nuclear hormone ligand-dependent transcription factors, has recently been reported to be involved in tumorigeniCity. We have shown that NSCLC cells express PPARbeta/delta protein and that treatment with a selective PPARbeta/delta agonist, GW501516, stimulated the expression of EP4 and induced NSCLC cell proliferation. In addition, this PPARbeta/delta agonist also induced EP4 promoter activity through the binding of C/EBP to the NF-IL6 site in the EP4 promoter. Therefore, PPARbeta/delta activation represents a novel molecular mechanism for regulating human cancer cell growth.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/metabolismo , Dinoprostona/metabolismo , Neoplasias Pulmonares/metabolismo , PPAR delta/agonistas , PPAR beta/agonistas , Receptores de Prostaglandina E/metabolismo , Western Blotting , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ensaio de Desvio de Mobilidade Eletroforética , Humanos , Neoplasias Pulmonares/patologia , PPAR delta/metabolismo , PPAR beta/metabolismo , Receptores de Prostaglandina E Subtipo EP4 , Tiazóis/farmacologia
9.
Cancer Res ; 66(1): 315-23, 2006 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-16397245

RESUMO

The Akt/mammalian target of rapamycin (mTOR)/ribosomal protein S6 kinase (p70S6K) pathway is considered a central regulator of protein synthesis and of cell proliferation, differentiation, and survival. However, the role of the Akt/mTOR/p70S6K pathway in lung carcinoma remains unknown. We previously showed that fibronectin, a matrix glycoprotein highly expressed in tobacco-related lung disease, stimulates non-small cell lung carcinoma (NSCLC) cell growth and survival. Herein, we explore the role of the Akt/mTOR/p70S6K pathway in fibronectin-induced NSCLC cell growth. We found that fibronectin stimulated the phosphorylation of Akt, an upstream inducer of mTOR, and induced the phosphorylation of p70S6K1 and eukaryotic initiation factor 4E-binding protein 1 (4E-BP1), two downstream targets of mTOR in NSCLC cells (H1792 and H1838), whereas it inhibited the phosphatase and tensin homologue deleted on chromosome 10, a tumor suppressor protein that antagonizes the phosphatidylinositol 3-kinase/Akt signal. In addition, treatment with fibronectin inhibited the mRNA and protein expression of LKB1 as well as the phosphorylation of AMP-activated protein kinase (AMPKalpha), both known to down-regulate mTOR. Rapamycin, an inhibitor of mTOR, blocked the fibronectin-induced phosphorylation of p70S6K and 4E-BP1. Akt small interfering RNA (siRNA) and an antibody against the fibronectin-binding integrin alpha5beta1 also blocked the p70S6K phosphorylation in response to fibronectin. In contrast, an inhibitor of extracellular signal-regulated kinase 1/2 (PD98095) had no effect on fibronectin-induced phosphorylation of p70S6K. Moreover, the combination of rapamycin and siRNA for Akt blocked fibronectin-induced cell proliferation. Taken together, these observations suggest that fibronectin-induced stimulation of NSCLC cell proliferation requires activation of the Akt/mTOR/p70S6K pathway and is associated with inhibition of LKB1/AMPK signaling.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/enzimologia , Fibronectinas/farmacologia , Neoplasias Pulmonares/enzimologia , Complexos Multienzimáticos/antagonistas & inibidores , Proteínas Quinases/metabolismo , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Quinases S6 Ribossômicas/metabolismo , Quinases Proteína-Quinases Ativadas por AMP , Proteínas Quinases Ativadas por AMP , Carcinoma Pulmonar de Células não Pequenas/patologia , Processos de Crescimento Celular/efeitos dos fármacos , Processos de Crescimento Celular/fisiologia , Relação Dose-Resposta a Droga , Ativação Enzimática/efeitos dos fármacos , Fibronectinas/antagonistas & inibidores , Humanos , Integrina alfa5beta1/fisiologia , Neoplasias Pulmonares/patologia , Complexos Multienzimáticos/biossíntese , Complexos Multienzimáticos/genética , Complexos Multienzimáticos/metabolismo , PTEN Fosfo-Hidrolase/genética , Fosforilação , Proteínas Serina-Treonina Quinases/biossíntese , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , RNA Mensageiro/antagonistas & inibidores , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Transdução de Sinais , Sirolimo/farmacologia , Serina-Treonina Quinases TOR
10.
Am J Respir Cell Mol Biol ; 37(6): 681-90, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17600315

RESUMO

The mechanisms by which tobacco promotes lung cancer remain incompletely understood. Herein, we report that nicotine, a major component of tobacco, promotes the proliferation of cultured non-small cell lung carcinoma (NSCLC) cells; this effect was most noticeable at 5 days. However, nicotine had no effect on apoptosis of NSCLC cells. In experiments designed to unveil the mechanisms for this effect, we found that nicotine also stimulated mRNA and protein expression of fibronectin. Fibronectin is a matrix glycoprotein that regulates important cellular processes (e.g., adhesion, proliferation, and differentiation) and is highly expressed in tobacco-related lung disorders. Of note, reagents against the integrin alpha5beta1 (antibodies, RGD peptides, alpha5 shRNA) blocked the mitogenic effects of nicotine. Thus, nicotine stimulated NSCLC cell proliferation indirectly via fibronectin induction. We then focused on the mechanisms responsible for nicotine-induced fibronectin expression in NSCLC cells and found that nicotine stimulated the surface expression of alpha7 nicotinic acetylcholine receptor (alpha7 nAChR), and that alpha-bungarotoxin, an inhibitor of alpha7 nAChR, abolished the nicotine-induced fibronectin response. The fibronectin-inducing effects of nicotine were associated with activation of extracellular signal-regulated kinase (ERK) and phosphoinositide 3-kinase (PI3-K)/mammalian target of rapamycin (mTOR) signaling pathways, and were abrogated by inhibitors of ERK (PD98059), PI3-K (LY294002), and mTOR (rapamycin), but not by inhibitors of protein kinase (PK)C (calphostin C) and PKA (H89). These observations suggest that nicotine stimulates NSCLC proliferation through induction of fibronectin, and that these events are mediated through nAChR-mediated signals that include ERK and PI3-K/mTOR pathways. This work highlights the role of fibronectin and alpha5beta1 integrins as potential targets for anti-lung cancer therapies.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/patologia , Fibronectinas/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Neoplasias Pulmonares/patologia , Nicotina/farmacologia , Anticorpos/farmacologia , Apoptose/efeitos dos fármacos , Carcinoma Pulmonar de Células não Pequenas/enzimologia , Carcinoma Pulmonar de Células não Pequenas/genética , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fibronectinas/metabolismo , Humanos , Integrina alfa5beta1/metabolismo , Neoplasias Pulmonares/enzimologia , Neoplasias Pulmonares/genética , Modelos Biológicos , Peptídeos/farmacologia , Fosforilação/efeitos dos fármacos , Proteínas Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Interferente Pequeno/metabolismo , Receptores Nicotínicos/metabolismo , Serina-Treonina Quinases TOR , Receptor Nicotínico de Acetilcolina alfa7
11.
Mol Cancer Ther ; 5(2): 430-7, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16505118

RESUMO

Peroxisome proliferator-activated receptors gamma (PPARgamma) exert diverse effects on cancer cells. Recent studies showed that rosiglitazone, a synthetic ligand for PPARgamma, inhibits cell growth. However, the exact mechanisms underlying this effect are still being explored, and the relevance of these findings to lung cancer remains unclear. Here, we report that rosiglitazone reduced the phosphorylation of Akt and increased phosphatase and tensin homologue (PTEN) protein expression in non-small cell lung carcinoma (NSCLC) cells (H1792 and H1838), and this was associated with inhibition of NSCLC cell proliferation. These effects were blocked or diminished by GW9662, a specific PPARgamma antagonist. However, transfection with a CMX-PPARgamma2 overexpression vector restored the effects of rosiglitazone on Akt, PTEN, and cell growth in the presence of GW9662. In addition, rosiglitazone increased the phosphorylation of AMP-activated protein kinase alpha (AMPKalpha), a downstream kinase target for LKB1, whereas it decreased phosphorylation of p70 ribosomal protein S6 kinase (p70S6K), a downstream target of mammalian target of rapamycin (mTOR). Of note, GW9662 did not affect the phosphorylation of AMPKalpha and p70S6K protein. The inhibitory effect of rosiglitazone on NSCLC cell growth was enhanced by the mTOR inhibitor rapamycin; however, it was blocked, in part, by the AMPKalpha small interfering RNA. Taken together, these findings show that rosiglitazone, via up-regulation of the PTEN/AMPK and down-regulation of the Akt/mTOR/p70S6K signal cascades, inhibits NSCLC cell proliferation through PPARgamma-dependent and PPARgamma-independent signals.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Neoplasias Pulmonares/tratamento farmacológico , PPAR gama/metabolismo , PTEN Fosfo-Hidrolase/metabolismo , Tiazolidinedionas/uso terapêutico , Proteínas Quinases Ativadas por AMP , Proliferação de Células/efeitos dos fármacos , Humanos , Neoplasias Pulmonares/metabolismo , Complexos Multienzimáticos/genética , Complexos Multienzimáticos/metabolismo , Fosforilação/efeitos dos fármacos , Proteínas Quinases/metabolismo , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Interferência de RNA , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Rosiglitazona , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR , Células Tumorais Cultivadas
12.
Lung Cancer ; 51(3): 283-96, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16376453

RESUMO

COX-2 has been implicated in the control of human non-small cell lung carcinoma (NSCLC) cell growth. The mechanisms by which COX-2 exerts its mitogenic effects have not been entirely elucidated, but stimulation of prostaglandin E2 production and alterations in the expression of the cyclin-dependent kinase inhibitor p21(WAF-1/CIP1/MDA-6)(p2i) have been suggested. Here, we demonstrate that two COX-2 inhibitors (NS398 and Nimesulide) inhibit proliferation and induce apoptosis in NSCLC cells, and these effects were associated with induction of p21 mRNA and protein expression. However, the anti-growth effect of the COX-2 inhibitors and their ability to induce p21 were not affected by COX-2 siRNA suggesting that their actions were COX-2 independent. Instead, activation of the MEK-1/Erk pathway was necessary since COX-2 inhibitors stimulated the phosphorylation of ERKs, and their effects were blocked by PD98095, an inhibitor of this pathway. Furthermore, we show that both NS398 and Nimesulide induced p21 gene promoter activity and this was prevented by PD98095. COX-2 inhibitors increased nuclear protein binding to the Spl site in the promoter region of the p21 gene. Consistent with a role for p21, we found that p21 antisense oligonucleotides prevented the effects of COX-2 inhibitors on cell growth. In summary, our results suggest that COX-2 inhibitors suppress NSCLC cell growth by inducing the expression of the p21 gene through MEK-1/ERK signaling and DNA-protein interactions involving Spl. These observations unveil a mechanism for p21 gene regulation by COX-2 inhibitors in lung carcinoma cell growth and this pathway represents a potential target for therapy.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/patologia , Inibidores de Ciclo-Oxigenase/farmacologia , Neoplasias Pulmonares/patologia , Nitrobenzenos/farmacologia , Sulfonamidas/farmacologia , Apoptose/efeitos dos fármacos , Western Blotting , Flavonoides/farmacologia , Regulação Neoplásica da Expressão Gênica , Humanos , MAP Quinase Quinase 1/efeitos dos fármacos , MAP Quinase Quinase Quinase 3/efeitos dos fármacos , Oligonucleotídeos Antissenso/farmacologia , Proteína Oncogênica p21(ras)/genética , Fosforilação , Regiões Promotoras Genéticas , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção , Células Tumorais Cultivadas
13.
Cancer Lett ; 219(1): 71-81, 2005 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-15694666

RESUMO

The cyclin-dependent kinase inhibitor p21(WAF-1/CIP1/MDA-6) (p21) plays a key role in cell cycle inhibition and apoptosis, and is negatively regulated during cell proliferation. Extracellular matrices can affect cellular proliferation, but their effects on p21 have not been entirely elucidated. Herein, we explore the effects of the matrix glycoprotein fibronectin on p21 expression in human lung carcinoma cells. Our studies show that fibronectin stimulates cell proliferation, and that this effect is associated with suppression of p21 and stimulation of cyclin D1 mRNA and protein levels in human lung non-small lung cell carcinoma cells (H1838). In contrast, the matrix protein collagen type 1 had no effect. The suppression of p21 by fibronectin was blocked by inhibitors of the extracellular signal-regulated kinase pathway (PD98095), and the Rho-kinase pathway (Y-27632). Fibronectin stimulated the phosphorylation of Erk and increased Rho protein expression. To determine the molecular mechanism(s) responsible for the inhibitory effects of fibronectin on p21 expression, transient transfection assays were performed with cells expressing a wild-type human p21 promoter construct. In these cells, fibronectin reduced p21 gene promoter activity. Finally, electrophoresis mobility shift experiments revealed that fibronectin decreased nuclear Sp1 binding activity in the promoter region of the p21 gene promoter, and a Sp1 competing oligonucleotide inhibited the fibronectin response. Taken together, our results suggest that fibronectin stimulates lung cancer carcinoma cell growth by reducing the cyclin-dependent kinase inhibitor p21 and by inducing cyclin D1 gene expression. The reduction of p21 by fibronectin appears to be mediated through Erk and Rho-kinase signaling and DNA-protein interactions at the Sp1 site in the p21 gene promoter. These observations unveil a novel mechanism for p21 gene regulation by fibronectin in lung carcinoma cell growth that represents a potential target for therapy.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/metabolismo , Proteínas de Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Fibronectinas/farmacologia , Neoplasias Pulmonares/metabolismo , Western Blotting , Proteínas de Ciclo Celular/metabolismo , Inibidor de Quinase Dependente de Ciclina p21 , Ensaio de Desvio de Mobilidade Eletroforética , MAP Quinases Reguladas por Sinal Extracelular/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Expressão Gênica/efeitos dos fármacos , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Regiões Promotoras Genéticas , Proteínas Serina-Treonina Quinases/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção , Células Tumorais Cultivadas , Quinases Associadas a rho
14.
Clin Cancer Res ; 9(12): 4627-35, 2003 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-14555539

RESUMO

PURPOSE: The peroxisome proliferator-activated receptor-gamma (PPARgamma), a ligand-dependent transcription factor belonging to the family of nuclear receptors, has been implicated in the control of cyclooxygenase (COX) 2 expression in some tissue, although the exact mechanism(s) of this activity has not been elucidated. In this study we explored the possible mechanism(s) of control of COX-2 gene expression through PPARgamma signaling in human cervical cancer. EXPERIMENTAL DESIGN: Using primary human cervical tissues and the CaSki human cervical cancer cell line, we assayed for PPARgamma and COX-2 mRNA expression by reverse transcription-PCR. Nuclear protein binding activities to three response elements located in the COX-2 promoter [nuclear factor kappaB (NFkappaB), cyclic AMP response element, and activator protein (AP)-2] were measured by gel mobility shift assays. We used transient transfection assays with COX-2 promoter reporter gene constructs to determine the regulatory sites in this promoter, which mediates PPARgamma regulation of COX-2 activity. RESULTS: We showed, for the first time, that primary human cervical cancer tissues express PPARgamma. Using CaSki cells, we demonstrated that COX-2 and PPARgamma mRNA levels were inversely regulated by PPARgamma ligands in that these compounds up-regulated PPARgamma but down-regulated COX-2. In contrast, epidermal growth factor (EGF), a potent activator of COX-2, decreased PPARgamma mRNA levels. This down-regulation of PPARgamma mRNA by EGF was blocked in the presence of NS-398, a selective COX-2 inhibitor. PPARgamma ligands suppressed the binding activities of AP-1 (binding to CRE) and NFkappaB but not AP-2. Transient transfection results indicated that EGF stimulated whereas PPARgamma ligands inhibited COX-2 promoter (-327/+59) activity. This effect by PPARgamma ligands on the COX-2 promoter was blocked when the CRE, but not the NFkappaB, binding site was mutagenized. CONCLUSION: Cervical cancer cells express readily detectable levels of PPARgamma. There is reciprocal negative regulation between COX-2 and PPARgamma signaling in human cervical cancer cells. The ability of PPARgamma ligands to inhibit COX-2 appears to be mediated predominantly through inhibition of AP-1 protein binding to the CRE site in the COX-2 promoter.


Assuntos
Regulação Enzimológica da Expressão Gênica , Isoenzimas/genética , Prostaglandina-Endoperóxido Sintases/genética , Receptores Citoplasmáticos e Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Neoplasias do Colo do Útero/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Ciclo-Oxigenase 2 , Inibidores de Ciclo-Oxigenase 2 , Inibidores de Ciclo-Oxigenase/farmacologia , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Ensaio de Desvio de Mobilidade Eletroforética , Fator de Crescimento Epidérmico/farmacologia , Feminino , Humanos , Isoenzimas/antagonistas & inibidores , Isoenzimas/metabolismo , Proteínas de Membrana , NF-kappa B/genética , NF-kappa B/metabolismo , Nitrobenzenos/farmacologia , Regiões Promotoras Genéticas/genética , Prostaglandina-Endoperóxido Sintases/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores Citoplasmáticos e Nucleares/antagonistas & inibidores , Receptores Citoplasmáticos e Nucleares/genética , Elementos de Resposta , Sulfonamidas/farmacologia , Fator de Transcrição AP-1/metabolismo , Fator de Transcrição AP-2 , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/genética , Ativação Transcricional , Transfecção , Células Tumorais Cultivadas , Neoplasias do Colo do Útero/genética
15.
Clin Cancer Res ; 10(6): 1911-9, 2004 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-15041706

RESUMO

PURPOSE: The peroxisome proliferator-activated receptor gamma (PPARgamma), a ligand-dependent transcription factor belonging to the family of nuclear receptors, has been implicated in the regulation of cell growth and differentiation although the exact mechanism(s) of this activity has not been elucidated. In this study, we explored the role of PPARgamma signaling on the control of gene expression of the cycle-dependent kinase inhibitor p21 in human lung carcinoma cells. EXPERIMENTAL DESIGN: Using several human lung carcinoma cell lines (small and non-small carcinoma cells), we assayed for cell growth inhibition and apoptosis induction. We also assayed for p21 mRNA and protein expression by reverse transcription-PCR, real-time reverse transcription-PCR, and Western blot analysis. Nuclear protein binding activities to three response elements located in the p21 promoter [nuclear factor (NF)-kappaB, Sp1, and NF-interleukin 6 (IL6) CAAT/enhancer binding protein (C/EBP)] were measured by gel mobility shift assays. We used transient transfection assays with p21 promoter reporter gene constructs to determine the transcriptional regulation by PPARgamma ligands. Finally, by using p21 antisense oligonucleotides, we tested the link between PPARgamma activation and p21 signaling in cell growth inhibition assays and by Western blot analysis. RESULTS: We showed that the PPARgamma ligands PGJ2 and ciglitazone inhibit the growth and induce the apoptosis of several human lung carcinoma cell lines, whereas the PPARalpha agonist WY14643 has little effect. Treatment of lung carcinoma cells with the PPARgamma ligands PGJ2, ciglitazone, troglizaone, and GW1929 elevated p21 mRNA and protein levels and reduced cyclin D1 mRNA levels. These results were supported by transient transfection assays, which indicated that PPARgamma ligands increased p21 gene promoter activity in human lung carcinoma cells. In addition, p21 antisense oligonucleotides inhibited PPARgamma ligand-induced p21 protein expression and significantly blocked lung carcinoma cell growth inhibition induced by PPARgamma ligands. Finally, electrophoresis mobility shift experiments demonstrated that PPARgamma ligands increased the nuclear binding activities of Sp1 and NF-IL6 (C/EBP), two transcription factors with regulatory elements in the promoter region of the p21 gene. CONCLUSION: PPARgamma ligands inhibit human lung carcinoma cell growth and induce apoptosis by stimulating the cyclin-dependent kinase inhibitor p21 and by reducing cyclin D1 gene expression. The induction of p21 gene expression by PPARgamma ligands may be mediated through increased Sp1- and NF-IL6 (C/EBP)-dependent transcriptional activation. These observations unveil a mechanism for p21 gene regulation in lung carcinoma that represents a potential target for therapy.


Assuntos
Regulação Neoplásica da Expressão Gênica/genética , Neoplasias Pulmonares/genética , Proteína Oncogênica p21(ras)/genética , PPAR gama/fisiologia , Apoptose/efeitos dos fármacos , Sequência de Bases , Divisão Celular , Linhagem Celular Tumoral , Primers do DNA , Humanos , Hipoglicemiantes/farmacologia , Ligantes , Neoplasias Pulmonares/patologia , NF-kappa B/metabolismo , Oligodesoxirribonucleotídeos Antissenso/farmacologia , Reação em Cadeia da Polimerase , Transdução de Sinais/efeitos dos fármacos , Tiazolidinedionas/farmacologia , Fatores de Transcrição/metabolismo , Transfecção
16.
J Clin Endocrinol Metab ; 88(2): 713-9, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12574204

RESUMO

Previous studies indicated that the antiprogestin RU486 could directly inhibit the growth of normal and malignant human endometrial cells. However, the mechanism by which this occurs is poorly understood. In this study we explore further details of endometrial cell growth regulation by RU486. Gel shift assays using the endometrial cell line EM42 demonstrated that RU486, at concentrations ranging from 20-100 micro M, significantly stimulated the cellular binding activity of the nuclear transcription factor nuclear factor-kappa B (NF-kappa B) while having little effect on activating protein-1 (AP-1) binding. This effect on NF-kappa B binding was blocked in the presence of the NF-kappa B inhibitor, pyrrolidine dithiocarbamate (PDTC). The data also showed that the activity of RU486 on NF-kappa B binding correlated with the ability of this compound to induce apoptosis of EM42 cells. To investigate a cause and effect relationship between these two phenomena, we evaluated the effects of RU486 treatment on the expression of two apoptosis-related genes, bax and bcl-2, known to be regulated through NF-kappa B binding on their promoter. RT-PCR demonstrated that RU486 significantly induced bax mRNA levels, while suppressing mRNA of bcl-2. Alteration of these genes by RU486 was inhibited in the presence of 100 microM PDTC. Correspondingly, PDTC antagonized the ability of RU486 to inhibit the growth and induce apoptosis of EM42 cells. This study demonstrates that the inhibition of growth and apoptosis of human endometrial cells by RU486 involves stimulation of NF-kappa B binding with subsequent modulation of apoptosis regulatory genes bax and bcl-2.


Assuntos
Endométrio/citologia , Antagonistas de Hormônios/farmacologia , Mifepristona/farmacologia , NF-kappa B/metabolismo , Prolina/análogos & derivados , Transdução de Sinais/efeitos dos fármacos , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Linhagem Celular , Feminino , Expressão Gênica/efeitos dos fármacos , Humanos , NF-kappa B/antagonistas & inibidores , Prolina/farmacologia , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-bcl-2/genética , RNA Mensageiro/metabolismo , Tiocarbamatos/farmacologia , Proteína X Associada a bcl-2
17.
Fertil Steril ; 79 Suppl 1: 763-9, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12620489

RESUMO

OBJECTIVE: To determine whether peroxisome proliferator-activated receptor (PPAR)-gamma ligands can affect the expression of interleukin-6 (IL-6) and cytokines related to the pathogenesis of endometriosis. DESIGN: In vitro study to determine whether PPARs are expressed in human endometrial cells and determine the effects of various PPAR-gamma ligands on IL-6 and other cytokine expression in these cells. SETTING: Academic medical center. PATIENT(S): Women presenting for infertility workup. INTERVENTION(S): Endometrial cell cultures were treated with PPAR-gamma ligands. MAIN OUTCOME MEASURE(S): Interleukin-6, IL-8, colony stimulating factor-1 (CSF-1) and macrophage chemotactic factor (MCP-1) protein secretion, messenger RNA expression of IL-6, PPAR-alpha, -beta, and -gamma. RESULT(S): Using a human endometrial cell line (EM42), as well as primary stromal and epithelial endometrial cells, we show the presence of PPAR-alpha, -beta, and -gamma by reverse transcription-polymerase chain reaction (RT-PCR) in these cells. PPAR-gamma ligands stimulated IL-6 secretion and induced enhancement of IL-6 mRNA levels. These ligands also stimulated the secretion of IL-8 and CSF-1. CONCLUSION(S): PPAR-gamma may play a role in the pathogenesis of endometriosis related to the production of IL-6 and some other cytokines.


Assuntos
Citocinas/biossíntese , Endométrio/metabolismo , Receptores Citoplasmáticos e Nucleares/fisiologia , Tiazolidinedionas , Fatores de Transcrição/fisiologia , Tirosina/análogos & derivados , Benzofenonas/farmacologia , Fatores Estimuladores de Colônias/biossíntese , Fatores Estimuladores de Colônias/metabolismo , Citocinas/metabolismo , Endométrio/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Feminino , Humanos , Interleucina-6/biossíntese , Interleucina-6/metabolismo , Ligantes , Fenilacetatos/farmacologia , Prostaglandina D2/análogos & derivados , Prostaglandina D2/farmacologia , Análise Serial de Proteínas , Isoformas de Proteínas , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Receptores Citoplasmáticos e Nucleares/biossíntese , Receptores Citoplasmáticos e Nucleares/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Tiazóis/farmacologia , Fatores de Transcrição/biossíntese , Fatores de Transcrição/metabolismo , Tirosina/farmacologia
18.
PLoS One ; 8(2): e56631, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23457597

RESUMO

Aerobic glycolysis is an important feature of cancer cells. In recent years, lactate dehydrogenase A (LDH-A) is emerging as a novel therapeutic target for cancer treatment. Seeking LDH-A inhibitors from natural resources has been paid much attention for drug discovery. Spatholobus suberectus (SS) is a common herbal medicine used in China for treating blood-stasis related diseases such as cancer. This study aims to explore the potential medicinal application of SS for LDH-A inhibition on breast cancer and to determine its bioactive compounds. We found that SS manifested apoptosis-inducing, cell cycle arresting and anti-LDH-A activities in both estrogen-dependent human MCF-7 cells and estrogen-independent MDA-MB-231 cell. Oral herbal extracts (1 g/kg/d) administration attenuated tumor growth and LDH-A expression in both breast cancer xenografts. Bioactivity-guided fractionation finally identified epigallocatechin as a key compound in SS inhibiting LDH-A activity. Further studies revealed that LDH-A plays a critical role in mediating the apoptosis-induction effects of epigallocatechin. The inhibited LDH-A activities by epigallocatechin is attributed to disassociation of Hsp90 from HIF-1α and subsequent accelerated HIF-1α proteasome degradation. In vivo study also demonstrated that epigallocatechin could significantly inhibit breast cancer growth, HIF-1α/LDH-A expression and trigger apoptosis without bringing toxic effects. The preclinical study thus suggests that the potential medicinal application of SS for inhibiting cancer LDH-A activity and the possibility to consider epigallocatechin as a lead compound to develop LDH-A inhibitors. Future studies of SS for chemoprevention or chemosensitization against breast cancer are thus warranted.


Assuntos
Antineoplásicos/farmacologia , Neoplasias da Mama/patologia , Inibidores Enzimáticos/farmacologia , Fabaceae/química , L-Lactato Desidrogenase/antagonistas & inibidores , Extratos Vegetais/farmacologia , Transdução de Sinais/efeitos dos fármacos , Animais , Antineoplásicos/química , Apoptose/efeitos dos fármacos , Catequina/análogos & derivados , Catequina/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Proteínas de Choque Térmico HSP90/metabolismo , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Isoenzimas/antagonistas & inibidores , Isoenzimas/genética , L-Lactato Desidrogenase/genética , Lactato Desidrogenase 5 , Pontos de Checagem da Fase M do Ciclo Celular/efeitos dos fármacos , Camundongos , Extratos Vegetais/química , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteólise/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
19.
PLoS One ; 8(7): e68566, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23861918

RESUMO

Angiogenesis is crucial for cancer initiation, development and metastasis. Identifying natural botanicals targeting angiogenesis has been paid much attention for drug discovery in recent years, with the advantage of increased safety. Isoliquiritigenin (ISL) is a dietary chalcone-type flavonoid with various anti-cancer activities. However, little is known about the anti-angiogenic activity of isoliquiritigenin and its underlying mechanisms. Herein, we found that ISL significantly inhibited the VEGF-induced proliferation of human umbilical vein endothelial cells (HUVECs) at non-toxic concentration. A series of angiogenesis processes including tube formation, invasion and migration abilities of HUVECs were also interrupted by ISL in vitro. Furthermore, ISL suppressed sprout formation from VEGF-treated aortic rings in an ex-vivo model. Molecular mechanisms study demonstrated that ISL could significantly inhibit VEGF expression in breast cancer cells via promoting HIF-1α (Hypoxia inducible factor-1α) proteasome degradation and directly interacted with VEGFR-2 to block its kinase activity. In vivo studies further showed that ISL administration could inhibit breast cancer growth and neoangiogenesis accompanying with suppressed VEGF/VEGFR-2 signaling, elevated apoptosis ratio and little toxicity effects. Molecular docking simulation indicated that ISL could stably form hydrogen bonds and aromatic interactions within the ATP-binding region of VEGFR-2. Taken together, our study shed light on the potential application of ISL as a novel natural inhibitor for cancer angiogenesis via the VEGF/VEGFR-2 pathway. Future studies of ISL for chemoprevention or chemosensitization against breast cancer are thus warranted.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Chalconas/farmacologia , Neovascularização Patológica/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Trifosfato de Adenosina , Animais , Sítios de Ligação , Neoplasias da Mama/tratamento farmacológico , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Chalconas/química , Suplementos Nutricionais , Modelos Animais de Doenças , Feminino , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Camundongos , Conformação Molecular , Simulação de Acoplamento Molecular , Neovascularização Patológica/tratamento farmacológico , Neovascularização Fisiológica/efeitos dos fármacos , Complexo de Endopeptidases do Proteassoma/metabolismo , Ligação Proteica , Proteólise/efeitos dos fármacos , Carga Tumoral/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/farmacologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/química , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA