Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Semin Cancer Biol ; 57: 111-116, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30213689

RESUMO

Prostate cancer is the most prevalent type of cancer in men. The etiology of prostate cancer development and the mechanisms underlying androgen-independent progression remains to be further investigated. There are many known targets for prostate cancer therapy including the androgen receptor (AR) axis, but resistance eventually develops in advanced disease suggesting the need to better understand mechanisms of resistance and consideration of multi-targeted therapy. Mechanisms contributing to resistance may include gene amplifications, gene mutations, AR splice variants, and changes in expression of androgen receptor co-regulatory proteins. Given the limitations of approved therapies, further study of additional potential targets is warranted. This review focuses on the roles of autophagy pathway, p62, Yes-associated protein (YAP), cancer stem cells, and epigenetics. Therapies targeting these potential mechanisms of resistance may interact with currently approved therapies either additively or synergistically. Thus, the study of combination therapy against multiple targets may be critically important to achieve more impact against lethal forms of prostate cancer resistant to all approved current therapies.


Assuntos
Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Suscetibilidade a Doenças , Neoplasias da Próstata/etiologia , Neoplasias da Próstata/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Autofagia/genética , Terapia Combinada , Progressão da Doença , Epigênese Genética , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Neoplasias da Próstata/patologia , Neoplasias da Próstata/terapia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteínas de Sinalização YAP
2.
Prostate ; 78(5): 390-400, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29368435

RESUMO

BACKGROUND: To test the hypothesis that p62 is an optimal target for autophagy inhibition and Verteporfin, a clinically available drug approved by FDA to treat macular degeneration that inhibits autophagy by targeting p62 protein, can be developed clinically to improve therapy for advanced prostate cancer. METHODS: Forced expression of p62 in PC-3 cells and normal prostate epithelial cells, RWPE-1 and PZ-HPV7, were carried out by transfection of these cells with pcDNA3.1/p62 or p62 shRNA plasmid. Autophagosomes and autophagic flux were measured by transfection of tandem fluorescence protein mCherry-GFP-LC3 construct. Apoptosis was measured by Annexin V/PI staining. Tumorigenesis was measured by a xenograft tumor growth model. RESULTS: Verteporfin inhibited cell growth and colony formation in PC-3 cells. Verteporfin generated crosslinked p62 oligomers, resulting in inhibition of autophagy and constitutive activation of Nrf2 as well as its target genes, Bcl-2 and TNF-α. In normal prostate epithelial cells, forced expression of p62 caused constitutive Nrf2 activation, development of apoptosis resistance, and Verteporfin treatment exhibited inhibitory effects. Verteporfin treatment also inhibited starvation-induced autophagic flux of these cells. Verteporfin inhibited tumorigenesis of both normal prostate epithelial cells with p62 expression and prostate cancer cells and decreased p62, constitutive Nrf2, and Bcl-xL in xenograft tumor tissues, indicating that p62 can be developed as a drug target against prostate cancer. CONCLUSIONS: p62 has a high potential to be developed as a therapeutic target. Verteporfin represents a prototypical agent with therapeutic potential against prostate cancer through inhibition of autophagy by a novel mechanism of p62 inhibition.


Assuntos
Neoplasias da Próstata/tratamento farmacológico , Proteínas de Ligação a RNA/antagonistas & inibidores , Verteporfina/farmacologia , Animais , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Processos de Crescimento Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Humanos , Masculino , Redes e Vias Metabólicas/efeitos dos fármacos , Camundongos , Camundongos Nus , Terapia de Alvo Molecular , Fator 2 Relacionado a NF-E2/metabolismo , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Proteínas de Ligação a RNA/metabolismo , Distribuição Aleatória , Espécies Reativas de Oxigênio/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
3.
J Biol Chem ; 291(31): 16271-81, 2016 07 29.
Artigo em Inglês | MEDLINE | ID: mdl-27226640

RESUMO

Hexavalent chromium (Cr(VI))-containing compounds are well established environmental carcinogens. Most mechanistic investigations of Cr(VI)-induced carcinogenesis focus on oxidative stress and various cellular responses, leading to malignant cell transformation or the first stage of metal-induced carcinogenesis. The development of malignantly transformed cells into tumors that require angiogenesis is the second stage. This study focuses on the second stage, in particular, the role of EGF receptor (EGFR) signaling in angiogenesis and tumorigenesis of Cr(VI)-transformed cells. Our preliminary studies have shown that EGFR is constitutively activated in Cr(VI)-transformed cells, in lung tissue from Cr(VI)-exposed animals, and in lung tumor tissue from a non-smoking worker occupationally exposed to Cr(VI) for 19 years. Using in vitro and in vivo models, the present study has investigated the role of EGFR in angiogenesis of Cr(VI)-transformed cells. The results show that Cr(VI)-transformed cells are angiogenic. Hypoxia-inducible factor-1α, pro-angiogenic protein matrix metalloproteinase 1, and VEGF are all highly expressed in Cr(VI)-transformed cells, in lung tissue from animals exposed to Cr(VI), and in lung tumor tissue from a non-smoking worker occupationally exposed to Cr(VI) for 19 years. p38 MAPK is also activated in Cr(VI)-transformed cells and in human lung tumor tissue. Inhibition of EGFR reduces p38 MAPK, resulting in decreased expression of hypoxia-inducible factor-1α, metalloproteinase 1, and VEGF, leading to suppressions of angiogenesis and tumorigenesis. Overall, the present study has demonstrated that EGFR plays an important role in angiogenesis and tumorigenesis of Cr(VI)-transformed cells.


Assuntos
Transformação Celular Neoplásica , Cromo/toxicidade , Receptores ErbB/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Neoplasias Pulmonares , Neovascularização Patológica , Exposição Ocupacional/efeitos adversos , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Transformação Celular Neoplásica/induzido quimicamente , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Receptores ErbB/genética , Feminino , Células Endoteliais da Veia Umbilical Humana , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Neoplasias Pulmonares/induzido quimicamente , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Endogâmicos BALB C , Neovascularização Patológica/induzido quimicamente , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia
4.
Toxicol Appl Pharmacol ; 331: 24-32, 2017 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-28416455

RESUMO

Cr(VI) is a well known environmental carcinogen, but its mechanism of action and the measures required to mitigate its effects remain to be investigated. Our previous studies showed that exposure of human bronchial epithelial (BEAS-2B) cells to Cr(VI) caused malignant transformation, that these transformed cells progressed through tumorigenesis, and that luteolin, a natural compound, inhibited both of these processes. The present study investigates the underlying mechanisms by which luteolin protects cells against Cr(VI)-induced transformation and tumorigenesis. The present study shows that luteolin activates inducible Nrf2 to inhibit Cr(VI)-generated reactive oxygen species (ROS) in normal BEAS-2B cells. The decreased ROS level is likely responsible for the protective effect of luteolin against Cr(VI)-induced malignant cell transformation in normal cells. By contrast, in cells that have been transformed by Cr(VI), Nrf2 is constitutively activated, and its target proteins, heme oxygenase 1 (HO-1), NAD(P)H:quinone oxidoreductase 1 (NQO1), and superoxide dismutase 1/2 (SOD1/SOD2) are all constitutively activated, and ROS levels are low. Bcl-2, an anti-apoptotic protein and target protein of Nrf2 is elevated. Cr(VI)-transformed BEAS-2B cells develop apoptosis resistance, increasing the survival of these transformed cells. Luteolin decreases interactions between Nrf2 and the antioxidant response element sites of its target anti-apoptotic and antioxidant proteins, Bcl-2, Bcl-XL, and HO-1, which results in decreased constitutive Nrf2 activation. The decreased constitutive Nrf2 activation, decrease in Nrf2 target proteins and consequent apoptosis resistance by luteolin are possible mechanisms that mediate the protective effect of luteolin in Cr(VI)-transformed cells.


Assuntos
Carcinogênese/metabolismo , Transformação Celular Neoplásica/metabolismo , Cromo/toxicidade , Citoproteção/fisiologia , Luteolina/farmacologia , Fator 2 Relacionado a NF-E2/metabolismo , Carcinogênese/induzido quimicamente , Carcinogênese/efeitos dos fármacos , Carcinógenos Ambientais/toxicidade , Linhagem Celular , Transformação Celular Neoplásica/efeitos dos fármacos , Citoproteção/efeitos dos fármacos , Relação Dose-Resposta a Droga , Humanos , Fator 2 Relacionado a NF-E2/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
5.
Toxicol Appl Pharmacol ; 331: 164-173, 2017 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-28624442

RESUMO

Hexavalent chromium (Cr(VI)) compounds are confirmed human carcinogens for lung cancer. Our previous studies has demonstrated that chronic exposure of human bronchial epithelial BEAS-2B cells to low dose of Cr(VI) causes malignant cell transformation. The acquisition of cancer stem cell-like properties is involved in the initiation of cancers. The present study has observed that a small population of cancer stem-like cells (BEAS-2B-Cr-CSC) exists in the Cr(VI)-transformed cells (BEAS-2B-Cr). Those BEAS-2B-Cr-CSC exhibit extremely reduced capability of generating reactive oxygen species (ROS) and apoptosis resistance. BEAS-2B-Cr-CSC are metabolic inactive as evidenced by reductions in oxygen consumption, glucose uptake, ATP production, and lactate production. Most importantly, BEAS-2B-Cr-CSC are more tumorigenic with high levels of cell self-renewal genes, Notch1 and p21. Further study has found that fructose-1,6-bisphosphatase (FBP1), an rate-limiting enzyme driving glyconeogenesis, was lost in BEAS-2B-Cr-CSC. Forced expression of FBP1 in BEAS-2B-Cr-CSC restored ROS generation, resulting in increased apoptosis, leading to inhibition of tumorigenesis. In summary, the present study suggests that loss of FBP1 is a critical event in tumorigenesis of Cr(VI)-transformed cells.


Assuntos
Apoptose/efeitos dos fármacos , Carcinogênese/efeitos dos fármacos , Cromo/toxicidade , Frutose-Bifosfatase/metabolismo , Glicólise/efeitos dos fármacos , Células-Tronco Neoplásicas/efeitos dos fármacos , Animais , Apoptose/fisiologia , Carcinogênese/metabolismo , Linhagem Celular , Linhagem Celular Transformada , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Deficiência de Frutose-1,6-Difosfatase/metabolismo , Glicólise/fisiologia , Humanos , Camundongos , Camundongos Nus , Células-Tronco Neoplásicas/metabolismo , Mucosa Respiratória/efeitos dos fármacos , Mucosa Respiratória/metabolismo
6.
J Biol Chem ; 290(4): 2213-24, 2015 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-25477514

RESUMO

Hexavalent chromium (Cr(VI)) compounds are well-established lung carcinogens. Epidermal growth factor receptor (EGFR) is a tyrosine kinase transmembrane receptor that regulates cell survival, tumor invasion, and angiogenesis. Our results show that chronic exposure of human bronchial epithelial (BEAS-2B) cells to Cr(VI) is able to cause malignant cell transformation. These transformed cells exhibit apoptosis resistance with reduced poly ADP-ribose polymerase cleavage (C-PARP) and Bax expression and enhanced expressions of Bcl-2 and Bcl-xL. These transformed cells also exhibit reduced capacity of reactive oxygen species (ROS) generation along with elevated expression of antioxidant manganese superoxide dismutase 2 (SOD2). The expression of this antioxidant was also elevated in lung tumor tissue from a worker exposed to Cr(VI) for 19 years. EGFR was activated in Cr(VI)-transformed BEAS-2B cells, lung tissue from animals exposed to Cr(VI) particles, and human lung tumor tissue. Further study indicates that constitutive activation of EGFR in Cr(VI)-transformed cells was due to increased binding to its ligand amphiregulin (AREG). Inhibition of EGFR or AREG increased Bax expression and reduced Bcl-2 expression, resulting in reduced apoptosis resistance. Furthermore, inhibition of AREG or EGFR restored capacity of ROS generation and decreased SOD2 expression. PI3K/AKT was activated, which depended on EGFR in Cr(VI)-transformed BEAS-2B cells. Inhibition of PI3K/AKT increased ROS generation and reduced SOD2 expression, resulting in reduced apoptosis resistance with commitment increase in Bax expression and reduction of Bcl-2 expression. Xenograft mouse tumor study further demonstrates the essential role of EGFR in tumorigenesis of Cr(VI)-transformed cells. In summary, the present study suggests that ligand-dependent constitutive activation of EGFR causes reduced ROS generation and increased antioxidant expression, leading to development of apoptosis resistance, contributing to Cr(VI)-induced tumorigenesis.


Assuntos
Apoptose , Cromo/química , Receptores ErbB/metabolismo , Regulação Neoplásica da Expressão Gênica , Espécies Reativas de Oxigênio/metabolismo , Anfirregulina/química , Animais , Antioxidantes/química , Arsênio/química , Sequência de Bases , Linhagem Celular Tumoral , Feminino , Inativação Gênica , Humanos , Ligantes , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Microscopia de Fluorescência , Dados de Sequência Molecular , Transplante de Neoplasias , Níquel/química , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Homologia de Sequência do Ácido Nucleico , Proteína X Associada a bcl-2/metabolismo
7.
Toxicol Appl Pharmacol ; 311: 106-112, 2016 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-27664007

RESUMO

Apigenin is a natural flavonoid which possesses multiple anti-cancer properties such as anti-proliferation, anti-inflammation, and anti-metastasis in many types of cancers including colorectal cancer. Neural precursor cell expressed developmentally downregulated 9 (NEDD9) is a multi-domain scaffolding protein of the Cas family which has been shown to correlate with cancer metastasis and progression. The present study investigates the role of NEDD9 in apigenin-inhibited cell migration, invasion, and metastasis of colorectal adenocarcinoma DLD1 and SW480 cells. The results show that knockdown of NEDD9 inhibited cell migration, invasion, and metastasis and that overexpression of NEDD9 promoted cell migration and invasion of DLD1 cells and SW4890 cells. Apigenin treatment attenuated NEDD9 expression at protein level, resulting in reduced phosphorylations of FAK, Src, and Akt, leading to inhibition on cell migration, invasion, and metastasis of both DLD1 and SW480 cells. The present study has demonstrated that apigenin inhibits cell migration, invasion, and metastasis through NEDD9/Src/Akt cascade in colorectal cancer cells. NEDD9 may function as a biomarker for evaluation of cancer aggressiveness and for selection of therapeutic drugs against cancer progression.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Apigenina/farmacologia , Neoplasias Colorretais/patologia , Regulação para Baixo/efeitos dos fármacos , Invasividade Neoplásica , Metástase Neoplásica , Fosfoproteínas/metabolismo , Animais , Linhagem Celular Tumoral , Neoplasias Colorretais/metabolismo , Feminino , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Humanos , Camundongos , Camundongos Nus , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Quinases da Família src/metabolismo
9.
Biochem Biophys Res Commun ; 456(2): 643-8, 2015 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-25499816

RESUMO

Arsenic is an environmental carcinogen, its mechanisms of carcinogenesis remain to be investigated. Reactive oxygen species (ROS) are considered to be important. A previous study (Carpenter et al., 2011) has measured ROS level in human lung bronchial epithelial (BEAS-2B) cells and arsenic-transformed BEAS-2B cells and found that ROS levels were higher in transformed cells than that in parent normal cells. Based on these observations, the authors concluded that cell transformation induced by arsenic is mediated by increased cellular levels of ROS. This conclusion is problematic because this study only measured the basal ROS levels in transformed and parent cells and did not investigate the role of ROS in the process of arsenic-induced cell transformation. The levels of ROS in arsenic-transformed cells represent the result and not the cause of cell transformation. Thus question concerning whether ROS are important in arsenic-induced cell transformation remains to be answered. In the present study, we used expressions of catalase (antioxidant against H2O2) and superoxide dismutase 2 (SOD2, antioxidant against O2(-)) to decrease ROS level and investigated their role in the process of arsenic-induced cell transformation. Our results show that inhibition of ROS by antioxidant enzymes decreased arsenic-induced cell transformation, demonstrating that ROS are important in this process. We have also shown that in arsenic-transformed cells, ROS generation was lower and levels of antioxidants are higher than those in parent cells, in a disagreement with the previous report. The present study has also shown that the arsenic-transformed cells acquired apoptosis resistance. The inhibition of catalase to increase ROS level restored apoptosis capability of arsenic-transformed BEAS-2B cells, further showing that ROS levels are low in these cells. The apoptosis resistance due to the low ROS levels may increase cells proliferation, providing a favorable environment for tumorigenesis of arsenic-transformed cells.


Assuntos
Arsênio/toxicidade , Brônquios/efeitos dos fármacos , Carcinógenos Ambientais/toxicidade , Transformação Celular Neoplásica/induzido quimicamente , Espécies Reativas de Oxigênio/metabolismo , Mucosa Respiratória/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Brônquios/metabolismo , Brônquios/patologia , Catalase/antagonistas & inibidores , Linhagem Celular , Humanos , Mucosa Respiratória/metabolismo , Mucosa Respiratória/patologia
10.
Toxicol Appl Pharmacol ; 287(3): 240-5, 2015 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-26091798

RESUMO

Occupational and environmental exposure to arsenic (III) and chromium VI (Cr(VI)) have been confirmed to cause lung cancer. Mechanisms of these metals carcinogenesis are still under investigation. Selection of cell lines to be used is essential for the studies. Human bronchial epithelial BEAS-2B cells are the cells to be utilized by most of scientists. However, due to p53 missense mutation (CCG→TCG) at codon 47 and the codon 72 polymorphism (CGC→CCC) in BEAS-2B cells, its usage has frequently been questioned. The present study has examined activity and expression of 53 and its downstream target protein p21 upon acute or chronic exposure of BEAS-2B cells to arsenic and Cr(VI). The results show that short-term exposure of BEAS-2B cells to arsenic or Cr(VI) was able to activate both p53 and p21. Chronic exposure of BEAS-2B cells to these two metals caused malignant cell transformation and tumorigenesis. In arsenic-transformed BEAS-2B cells reductions in p53 promoter activity, mRNA expression, and phosphorylation of p53 at Ser392 were observed, while the total p53 protein level remained the same compared to those in passage-matched parent ones. p21 promoter activity and expression were decreased in arsenic-transformed cells. Cr(VI)-transformed cells exhibit elevated p53 promoter activity, mRNA expression, and phosphorylation at Ser15, but reduced phosphorylation at Ser392 and total p53 protein level compared to passage-matched parent ones. p21 promoter activity and expression were elevated in Cr(VI)-transformed cells. These results demonstrate that p53 is able to respond to exposure of arsenic or Cr(VI), suggesting that BEAS-2B cells are an appropriate in vitro model to investigate arsenic or Cr(VI) induced lung cancer.


Assuntos
Arsenitos/toxicidade , Brônquios/efeitos dos fármacos , Transformação Celular Neoplásica/induzido quimicamente , Cromatos/toxicidade , Células Epiteliais/efeitos dos fármacos , Intoxicação por Metais Pesados , Neoplasias Pulmonares/induzido quimicamente , Intoxicação/etiologia , Compostos de Sódio/toxicidade , Animais , Brônquios/metabolismo , Brônquios/patologia , Linhagem Celular , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Relação Dose-Resposta a Droga , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Metais Pesados/metabolismo , Camundongos Nus , Mutação de Sentido Incorreto , Fosforilação , Intoxicação/genética , Intoxicação/metabolismo , Intoxicação/patologia , Regiões Promotoras Genéticas , Medição de Risco , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Ativação Transcricional , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
11.
Toxicol Appl Pharmacol ; 288(2): 232-9, 2015 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-26220687

RESUMO

Arsenic is a known carcinogen to humans, and chronic exposure to environmental arsenic is a worldwide health concern. As a dietary factor, ethanol carries a well-established risk for malignancies, but the effects of co-exposure to arsenic and ethanol on tumor development are not well understood. In the present study, we hypothesized that ethanol would enhance the function of an environmental carcinogen such as arsenic through increase in COX-2 expression. Our in vitro results show that ethanol enhanced arsenic-induced COX-2 expression. We also show that the increased COX-2 expression associates with intracellular ROS generation, up-regulated AKT signaling, with activation of both NFAT and NF-κB pathways. We demonstrate that antioxidant enzymes have an inhibitory effect on arsenic/ethanol-induced COX-2 expression, indicating that the responsive signaling pathways from co-exposure to arsenic and ethanol relate to ROS generation. In vivo results also show that co-exposure to arsenic and ethanol increased COX-2 expression in mice. We conclude that ethanol enhances arsenic-induced COX-2 expression in colorectal cancer cells via both the NFAT and NF-κB pathways. These results imply that, as a common dietary factor, ethanol ingestion may be a compounding risk factor for arsenic-induced carcinogenesis/cancer development.


Assuntos
Consumo de Bebidas Alcoólicas/metabolismo , Arsenitos/toxicidade , Carcinógenos Ambientais/toxicidade , Neoplasias Colorretais/enzimologia , Ciclo-Oxigenase 2/biossíntese , Etanol/toxicidade , NF-kappa B/metabolismo , Fatores de Transcrição NFATC/metabolismo , Transdução de Sinais/efeitos dos fármacos , Compostos de Sódio/toxicidade , Consumo de Bebidas Alcoólicas/genética , Neoplasias Colorretais/genética , Ciclo-Oxigenase 2/genética , Relação Dose-Resposta a Droga , Indução Enzimática , Células HCT116 , Células HT29 , Humanos , Masculino , NF-kappa B/genética , Fatores de Transcrição NFATC/genética , Estresse Oxidativo/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Medição de Risco
12.
Toxicol Appl Pharmacol ; 284(1): 92-99, 2015 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-25680589

RESUMO

Extensive exposure of solar ultraviolet-B (UVB) radiation to skin induces oxidative stress and inflammation that play a crucial role in the induction of skin cancer. Photochemoprevention with natural products represents a simple but very effective strategy for the management of cutaneous neoplasia. In this study, we investigated whether blackberry extract (BBE) reduces chronic inflammatory responses induced by UVB irradiation in SKH-1 hairless mice skin. Mice were exposed to UVB radiation (100 mJ/cm(2)) on alternate days for 10 weeks, and BBE (10% and 20%) was applied topically a day before UVB exposure. Our results show that BBE suppressed UVB-induced hyperplasia and reduced infiltration of inflammatory cells in the SKH-1 hairless mice skin. BBE treatment reduced glutathione (GSH) depletion, lipid peroxidation (LPO), and myeloperoxidase (MPO) in mouse skin by chronic UVB exposure. BBE significantly decreased the level of pro-inflammatory cytokines IL-6 and TNF-α in UVB-exposed skin. Likewise, UVB-induced inflammatory responses were diminished by BBE as observed by a remarkable reduction in the levels of phosphorylated MAP Kinases, Erk1/2, p38, JNK1/2 and MKK4. Furthermore, BBE also reduced inflammatory mediators such as cyclooxygenase-2 (COX-2), prostaglandin E2 (PGE2), and inducible nitric oxide synthase (iNOS) levels in UVB-exposed skin. Treatment with BBE inhibited UVB-induced nuclear translocation of NF-κB and degradation of IκBα in mouse skin. Immunohistochemistry analysis revealed that topical application of BBE inhibited the expression of 8-oxo-7, 8-dihydro-2'-deoxyguanosine (8-oxodG), cyclobutane pyrimidine dimers (CPD), proliferating cell nuclear antigen (PCNA), and cyclin D1 in UVB-exposed skin. Collectively, these data indicate that BBE protects from UVB-induced oxidative damage and inflammation by modulating MAP kinase and NF-κB signaling pathways.


Assuntos
Anti-Inflamatórios/farmacologia , Antioxidantes/farmacologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , NF-kappa B/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Extratos Vegetais/farmacologia , Rubus , Transdução de Sinais/efeitos dos fármacos , Pele/efeitos dos fármacos , Queimadura Solar/prevenção & controle , Protetores Solares/farmacologia , Raios Ultravioleta , Transporte Ativo do Núcleo Celular , Animais , Anti-Inflamatórios/isolamento & purificação , Antioxidantes/isolamento & purificação , Biomarcadores/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proliferação de Células/efeitos dos fármacos , Dano ao DNA , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Feminino , Frutas , Mediadores da Inflamação/metabolismo , Peroxidação de Lipídeos/efeitos dos fármacos , Camundongos Pelados , Neoplasias Induzidas por Radiação/enzimologia , Neoplasias Induzidas por Radiação/imunologia , Neoplasias Induzidas por Radiação/prevenção & controle , Fosforilação , Fitoterapia , Extratos Vegetais/isolamento & purificação , Plantas Medicinais , Rubus/química , Pele/enzimologia , Pele/imunologia , Pele/patologia , Neoplasias Cutâneas/enzimologia , Neoplasias Cutâneas/imunologia , Neoplasias Cutâneas/prevenção & controle , Queimadura Solar/enzimologia , Queimadura Solar/imunologia , Queimadura Solar/patologia , Protetores Solares/isolamento & purificação , Fatores de Tempo
13.
Toxicol Appl Pharmacol ; 280(1): 127-37, 2014 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-25062774

RESUMO

Skin cancer is one of the most commonly diagnosed cancers in the United States. Exposure to ultraviolet-B (UVB) radiation induces inflammation and photocarcinogenesis in mammalian skin. Cyanidin-3-glucoside (C3G), a member of the anthocyanin family, is present in various vegetables and fruits especially in edible berries, and displays potent antioxidant and anticarcinogenic properties. In this study, we have assessed the in vivo effects of C3G on UVB irradiation induced chronic inflammatory responses in SKH-1 hairless mice, a well-established model for UVB-induced skin carcinogenesis. Here, we show that C3G inhibited UVB-induced skin damage and inflammation in SKH-1 hairless mice. Our results indicate that C3G inhibited glutathione depletion, lipid peroxidation and myeloperoxidation in mouse skin by chronic UVB exposure. C3G significantly decreased the production of UVB-induced pro-inflammatory cytokines, such as IL-6 and TNF-α, associated with cutaneous inflammation. Likewise, UVB-induced inflammatory responses were diminished by C3G as observed by a remarkable reduction in the levels of phosphorylated MAP kinases, Erk1/2, p38, JNK1/2 and MKK4. Furthermore, C3G also decreased UVB-induced cyclooxygenase-2 (COX-2), PGE2 and iNOS levels, which are well-known key mediators of inflammation and cancer. Treatment with C3G inhibited UVB-induced nuclear translocation of NF-κB and degradation of IκBα in mice skin. Immunofluorescence assay revealed that topical application of C3G inhibited the expression of 8-hydroxy-2'-deoxyguanosine, proliferating cell nuclear antigen, and cyclin D1 in chronic UVB exposed mouse skin. Collectively, these data indicates that C3G can provide substantial protection against the adverse effects of UVB radiation by modulating UVB-induced MAP kinase and NF-κB signaling pathways.


Assuntos
Antocianinas/farmacologia , Glucosídeos/farmacologia , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , NF-kappa B/antagonistas & inibidores , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/efeitos da radiação , Raios Ultravioleta/efeitos adversos , Animais , Relação Dose-Resposta a Droga , Feminino , Inflamação/metabolismo , Inflamação/patologia , Inflamação/prevenção & controle , Camundongos , Camundongos Pelados , Proteínas Quinases Ativadas por Mitógeno/fisiologia , NF-kappa B/fisiologia , Estresse Oxidativo/fisiologia , Distribuição Aleatória , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Transdução de Sinais/efeitos da radiação , Absorção Cutânea/efeitos dos fármacos , Absorção Cutânea/fisiologia , Absorção Cutânea/efeitos da radiação
14.
Toxicol Appl Pharmacol ; 281(2): 230-41, 2014 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-25448439

RESUMO

Hexavalent chromium [Cr(VI)] is a well-known human carcinogen associated with the incidence of lung cancer. Inhibition of metal induced carcinogenesis by a dietary antioxidant is a novel approach. Luteolin, a natural dietary flavonoid found in fruits and vegetables, possesses potent antioxidant and anti-inflammatory activity. We found that short term exposure of human bronchial epithelial cells (BEAS-2B) to Cr(VI) (5µM) showed a drastic increase in ROS generation, NADPH oxidase (NOX) activation, lipid peroxidation, and glutathione depletion, which were significantly inhibited by the treatment with luteolin in a dose dependent manner. Treatment with luteolin decreased AP-1, HIF-1α, COX-2, and iNOS promoter activity induced by Cr(VI) in BEAS-2B cells. In addition, luteolin protected BEAS-2B cells from malignant transformation induced by chronic Cr(VI) exposure. Moreover, luteolin also inhibited the production of pro-inflammatory cytokines (IL-1ß, IL-6, IL-8, TNF-α) and VEGF in chronic Cr(VI) exposed BEAS-2B cells. Western blot analysis showed that luteolin inhibited multiple gene products linked to survival (Akt, Fak, Bcl-2, Bcl-xL), inflammation (MAPK, NF-κB, COX-2, STAT-3, iNOS, TNF-α) and angiogenesis (HIF-1α, VEGF, MMP-9) in chronic Cr(VI) exposed BEAS-2B cells. Nude mice injected with BEAS-2B cells chronically exposed to Cr(VI) in the presence of luteolin showed reduced tumor incidence compared to Cr(VI) alone treated group. Overexpression of catalase (CAT) or SOD2, eliminated Cr(VI)-induced malignant transformation. Overall, our results indicate that luteolin protects BEAS-2B cells from Cr(VI)-induced carcinogenesis by scavenging ROS and modulating multiple cell signaling mechanisms that are linked to ROS. Luteolin, therefore, serves as a potential chemopreventive agent against Cr(VI)-induced carcinogenesis.


Assuntos
Anticarcinógenos/farmacologia , Brônquios/efeitos dos fármacos , Transformação Celular Neoplásica/efeitos dos fármacos , Cromo/toxicidade , Células Epiteliais/efeitos dos fármacos , Sequestradores de Radicais Livres/farmacologia , Neoplasias Pulmonares/prevenção & controle , Luteolina/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Dicromato de Potássio/toxicidade , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteínas Angiogênicas/metabolismo , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Brônquios/metabolismo , Brônquios/patologia , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Transformação Celular Neoplásica/induzido quimicamente , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Citoproteção , Relação Dose-Resposta a Droga , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Humanos , Mediadores da Inflamação/metabolismo , Neoplasias Pulmonares/induzido quimicamente , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Camundongos Nus , Transfecção , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Toxicol Appl Pharmacol ; 271(2): 239-48, 2013 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-23707771

RESUMO

Cr(VI) compounds are known human carcinogens that primarily target the lungs. Cr(VI) produces reactive oxygen species (ROS), but the exact effects of ROS on the signaling molecules involved in Cr(VI)-induced carcinogenesis have not been extensively studied. Chronic exposure of human bronchial epithelial cells to Cr(VI) at nanomolar concentrations (10-100nM) for 3months not only induced cell transformation, but also increased the potential of these cells to invade and migrate. Injection of Cr(VI)-stimulated cells into nude mice resulted in the formation of tumors. Chronic exposure to Cr(VI) increased levels of intracellular ROS and antiapoptotic proteins. Transfection with catalase or superoxide dismutase (SOD) prevented Cr(VI)-mediated increases in colony formation, cell invasion, migration, and xenograft tumors. While chronic Cr(VI) exposure led to activation of signaling cascades involving PI3K/AKT/GSK-3ß/ß-catenin and PI3K/AKT/mTOR, transfection with catalase or SOD markedly inhibited Cr(VI)-mediated activation of these signaling proteins. Inhibitors specific for AKT or ß-catenin almost completely suppressed the Cr(VI)-mediated increase in total and active ß-catenin proteins and colony formation. In particular, Cr(VI) suppressed autophagy of epithelial cells under nutrition deprivation. Furthermore, there was a marked induction of AKT, GSK-3ß, ß-catenin, mTOR, and carcinogenic markers in tumor tissues formed in mice after injection with Cr(VI)-stimulated cells. Collectively, our findings suggest that ROS is a key mediator of Cr(VI)-induced carcinogenesis through the activation of PI3K/AKT-dependent GSK-3ß/ß-catenin signaling and the promotion of cell survival mechanisms via the inhibition of apoptosis and autophagy.


Assuntos
Carcinógenos , Cromo/toxicidade , Quinase 3 da Glicogênio Sintase/fisiologia , Proteína Oncogênica v-akt/fisiologia , Fosfatidilinositol 3-Quinases/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , beta Catenina/fisiologia , Animais , Western Blotting , Linhagem Celular Tumoral , Movimento Celular , Células Clonais/efeitos dos fármacos , Espectroscopia de Ressonância de Spin Eletrônica , Glicogênio Sintase Quinase 3 beta , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Nus , Plasmídeos , Transfecção
16.
Toxicol Appl Pharmacol ; 269(2): 89-99, 2013 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-23545178

RESUMO

Exposure of the skin to ultraviolet B (UVB) radiation causes oxidative damage to skin, resulting in sunburn, photoaging, and skin cancer. It is generally believed that the skin damage induced by UV irradiation is a consequence of generation of reactive oxygen species (ROS). Recently, there is an increased interest in the use of natural products as chemopreventive agents for non-melanoma skin cancer (NMSC) due to their antioxidants and anti-inflammatory properties. Quercitrin, glycosylated form of quercetin, is the most common flavonoid in nature with antioxidant properties. The present study investigated the possible beneficial effects of quercitrin to inhibit UVB irradiation-induced oxidative damage in vitro and in vivo. Our results showed that quercitrin decreased ROS generation induced by UVB irradiation in JB6 cells. Quercitrin restored catalase expression and GSH/GSSG ratio reduced by UVB exposure, two major antioxidant enzymes, leading to reductions of oxidative DNA damage and apoptosis and protection of the skin from inflammation caused by UVB exposure. The present study demonstrated that quercitrin functions as an antioxidant against UVB irradiation-induced oxidative damage to skin.


Assuntos
Antioxidantes/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/efeitos da radiação , Quercetina/análogos & derivados , Pele/efeitos da radiação , Raios Ultravioleta/efeitos adversos , Animais , Linhagem Celular , Dano ao DNA , Reparo do DNA , Células Epidérmicas , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos da radiação , Camundongos , NF-kappa B , Quercetina/farmacologia , Espécies Reativas de Oxigênio
17.
Mol Pharm ; 10(1): 406-16, 2013 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-23215050

RESUMO

OATP1B3 is a member of the OATP (organic anion transporting polypeptides) superfamily, responsible for mediating the transport of numerous endogenous and xenobiotic substances. Although initially reported to be exclusively expressed in the liver, several studies reported that OATP1B3 is frequently expressed in multiple types of cancers and may be associated with differing clinical outcomes. However, a detailed investigation on the expression and function of OATP1B3 protein in cancer has been lacking. In this study, we confirmed that colon and pancreatic cancer cells express variant forms of OATP1B3, different from OATP1B3 wild-type (WT) expressed in the normal liver. OATP1B3 variant 1 (V1), the most prevalent form among the variants, contains alternative exonic sequences (exon 2a) instead of exons 1 and 2 present in OATP1B3 WT. The translated product of OATP1B3 V1 is almost identical to OATP1B3 WT, with exception to the first 28 amino acids at the N-terminus. Exogenous expression of OATP1B3 V1 revealed that OATP1B3 V1 undergoes post-translational modifications and proteasomal degradation to a differing extent compared to OATP1B3 WT. OATP1B3 V1 showed only modest transport activity toward cholecystokin-8 (CCK-8, a prototype OATP1B3 substrate) in contrast to OATP1B3 WT showing a markedly efficient uptake of CCK-8. Consistent with these results, OATP1B3 V1 was localized mainly in the cytoplasm with a much lower extent of trafficking to the surface membrane compared to OATP1B3 WT. In summary, our results demonstrate that colon and pancreatic cancer cells express variant forms of OATP1B3 with only limited transport activity and different subcellular localization compared to OATP1B3 WT. These observed differences at the molecular and functional levels will be important considerations for further investigations of the biological and clinical significance of OATP1B3 expression in cancer.


Assuntos
Neoplasias do Colo/genética , Neoplasias do Colo/metabolismo , Transportadores de Ânions Orgânicos Sódio-Independentes/genética , Neoplasias Pancreáticas/genética , Transporte Biológico , Linhagem Celular Tumoral , Citoplasma/genética , Citoplasma/metabolismo , Éxons , Variação Genética , Células HCT116 , Humanos , Fígado/metabolismo , Transportadores de Ânions Orgânicos Sódio-Independentes/metabolismo , Neoplasias Pancreáticas/metabolismo , Processamento de Proteína Pós-Traducional , Sincalida/genética , Sincalida/metabolismo , Membro 1B3 da Família de Transportadores de Ânion Orgânico Carreador de Soluto
18.
Pharm Biol ; 51(8): 1066-76, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23746221

RESUMO

CONTEXT: Recently, there has been renewed interest in barley (Hordeum vulgare L. Poaceae) as a functional food and for its medicinal properties. OBJECTIVE: This study examines the anti-inflammatory potential of the active fractions of barley and the mechanisms involved. MATERIALS AND METHODS: The macrophages were exposed to 100 µg/mL of each of the barley extracts in the presence of 1 µg/mL lipopolysaccharide (LPS) and after 24 or 48 h of incubation, cells or culture supernatants were analyzed by various assays. The anti-inflammatory potential of barley fractions was also investigated using the LPS-injected septic mouse model. The active constituents in the fractions were identified using gas chromatography-mass spectrometry (GC-MS). RESULTS: The active fractions, named F4, F7, F9 and F12, inhibited almost completely the LPS-induced production of nitric oxide (NO) and inducible NO synthase. Pre-treatment with these fractions at 100 µg/mL diminished the tumor necrosis factor-α (TNF-α) levels to 19.8, 3.5, 1.2 and 1.7 ng/mL, respectively, compared to LPS treatment alone (41.5 ng/mL). These fractions at 100 µg/mL also suppressed apparently the secretion of interleukin (IL)-6 and IL-1ß and the DNA-binding activity of nuclear factor-κB in LPS-stimulated cells. Mice injected intraperitoneally with LPS (30 mg/kg BW) showed 20% survival at 48 h after injection, whereas oral administration of the fractions improved the survival rates to 80%. GC-MS analysis revealed the presence of the derivatives of benzoic and cinnamic acids and fatty acids in the fractions. DISCUSSION AND CONCLUSION: The aerial parts of barley are useful as functional food to prevent acute inflammatory responses.


Assuntos
Anti-Inflamatórios/farmacologia , Hordeum/química , Inflamação/tratamento farmacológico , Extratos Vegetais/farmacologia , Animais , Anti-Inflamatórios/isolamento & purificação , Modelos Animais de Doenças , Feminino , Cromatografia Gasosa-Espectrometria de Massas , Inflamação/fisiopatologia , Lipopolissacarídeos , Metanol/química , Camundongos , Camundongos Endogâmicos BALB C , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo II/efeitos dos fármacos , Óxido Nítrico Sintase Tipo II/metabolismo , Componentes Aéreos da Planta , Extratos Vegetais/química , Sepse/tratamento farmacológico , Sepse/fisiopatologia , Taxa de Sobrevida , Fatores de Tempo
19.
Mol Pharm ; 9(8): 2197-205, 2012 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-22734651

RESUMO

Carfilzomib is a novel class of peptidyl epoxyketone proteasome inhibitor and has demonstrated promising activity in multiple clinical trials to treat patients with multiple myeloma and other types of cancers. Here, we investigated molecular mechanisms underlying acquired resistance to carfilzomib and a potential strategy to restore cellular sensitivity to carfilzomib. H23 and DLD-1 cells (human lung and colon adenocarcinoma cell lines) with acquired resistance to carfilzomib displayed marked cross-resistance to YU-101, a closely related proteasome inhibitor, and paclitaxel, a known substrate of Pgp. However, carfilzomib-resistant cells remained sensitive to bortezomib, a clinically used dipeptide with boronic acid pharmacophore. In accordance with these observations, carfilzomib-resistant H23 and DLD-1 cells showed marked upregulation of P-glycoprotein (Pgp) as compared to their parental controls, and coincubation with verapamil, a Pgp inhibitor, led to an almost complete restoration of cellular sensitivity to carfilzomib. These results indicate that Pgp upregulation plays a major role in the development of carfilzomib resistance in these cell lines. In developing a potential strategy to overcome carfilzomib resistance, we as a proof of concept prepared a small library of peptide analogues derived from the peptide backbone of carfilzomib and screened these molecules for their activity to restore carfilzomib sensitivity when cotreated with carfilzomib. We found that compounds as small as dipeptides are sufficient in restoring carfilzomib sensitivity. Taken together, we found that Pgp upregulation plays a major role in the development of resistance to carfilzomib in lung and colon adenocarcinoma cell lines and that small peptide analogues lacking the pharmacophore can be used as agents to reverse acquired carfilzomib resistance. Our findings may provide important information in developing a potential strategy to overcome drug resistance.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Oligopeptídeos/farmacologia , Peptídeos/farmacologia , Ácidos Borônicos/farmacologia , Bortezomib , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos , Humanos , Peptídeos/química , Inibidores de Proteassoma/farmacologia , Pirazinas/farmacologia , Verapamil/farmacologia
20.
Biochem Biophys Res Commun ; 409(4): 792-8, 2011 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-21621523

RESUMO

Major histocompatibility complex (MHC) class I is a major host defense mechanism against viral infections such as type 16 and type 18 of the human papillomavirus (HPV). Here, we found that the E6 oncogene from HPV16, but not HPV18, suppressed MHC I expression. Ectopic expression of HPV16E6 in HeLa cells, which are infected with HPV18, suppressed MHC I expression, and that knockdown by antisense or siRNA of the HPV16E6 strongly enhanced MHC I expression in Caski cells, which are infected with HPV18, but not HPV16. The expression of HPV16E6 strongly enhanced cellular resistance to cytotoxic T lymphocytes (CTLs)-mediated lytic activity, and knockdown of HPV16E6 by antisense had the opposite effect. The regulation of HPV16E6-mediated MHC I suppression might be through the regulation of lymphotoxin (LT) and its receptor, LTßR. In addition, cells from the spleen and liver of LTα- or LTßR-deficient mice showed increased MHC I expression. Overall, these results demonstrated that the E6 oncogene of HPV16 might play an important role in cell transformation and cancer development through LT-mediated MHC I downregulation in humans.


Assuntos
Regulação Neoplásica da Expressão Gênica , Antígenos de Histocompatibilidade Classe I/genética , Papillomavirus Humano 16/genética , Linfotoxina-alfa/genética , Linfotoxina-beta/genética , Proteínas Oncogênicas Virais/genética , Proteínas Repressoras/genética , Neoplasias do Colo do Útero/genética , Neoplasias do Colo do Útero/virologia , Animais , Feminino , Técnicas de Silenciamento de Genes , Células HeLa , Antígenos de Histocompatibilidade Classe I/imunologia , Papillomavirus Humano 18/genética , Humanos , Receptor beta de Linfotoxina/genética , Camundongos , Camundongos Endogâmicos C57BL , Linfócitos T Citotóxicos/imunologia , Regulação para Cima , Neoplasias do Colo do Útero/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA