Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Mais filtros

Medicinas Complementares
Métodos Terapêuticos e Terapias MTCI
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
J Appl Microbiol ; 122(3): 770-784, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28004480

RESUMO

AIMS: To investigate the in vivo effects of Lactobacillus rhamnosus GG (LGG) on intestinal polyp development and the interaction between this single-organism probiotic and the gut microbiota therein. METHODS AND RESULTS: The ApcMin/+ mouse model was used to study the potential preventive effect of LGG on intestinal polyposis, while shotgun metagenomic sequencing was employed to characterize both taxonomic and functional changes within the gut microbial community. We found that the progression of intestinal polyps in the control group altered the community functional profile remarkably despite small variation in the taxonomic diversity. In comparison, the consumption of LGG helped maintain the overall functional potential and taxonomic profile in the resident microbes, thereby leading to a 25% decrease of total polyp counts. Furthermore, we found that LGG enriched those microbes or microbial activities related to short-chain fatty acid production (e.g. Roseburia and Coprococcus), as well as suppressed the ones that can lead to inflammation (e.g. Bilophila wadsworthia). CONCLUSIONS: Our study using shotgun metagenomics highlights how single probiotic LGG may exert its beneficial effects and decrease polyp formation in mice by maintaining gut microbial functionality. SIGNIFICANCE AND IMPACT OF THE STUDY: This probiotic intervention targeting microbiota may be used in conjugation with other dietary supplements or drugs as part of prevention strategies for early-stage colon cancer, after further clinical validations in human.


Assuntos
Pólipos Intestinais/prevenção & controle , Lacticaseibacillus rhamnosus/crescimento & desenvolvimento , Microbiota/efeitos dos fármacos , Probióticos/uso terapêutico , Sulindaco/uso terapêutico , Proteína da Polipose Adenomatosa do Colo/genética , Animais , Humanos , Metagenômica/métodos , Camundongos , Filogenia , Probióticos/farmacologia , Organismos Livres de Patógenos Específicos , Sulindaco/farmacologia
2.
Toxicol Lett ; 258: 126-133, 2016 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-27339904

RESUMO

Sulindac has anti-neoplastic properties against colorectal cancers; however, its use as a chemopreventive agent has been limited due to toxicity and efficacy concerns. Combinatorial treatment of colorectal cancers has been attempted to maximize anti-cancer efficacy with minimal side effects by administrating NSAIDs in combination with other inhibitory compounds or drugs such as l-ascorbic acid (vitamin C), which is known to exhibit cytotoxicity towards various cancer cells at high concentrations. In this study, we evaluated a combinatorial strategy utilizing sulindac and vitamin C. The death of HCT116 cells upon combination therapy occurred via a p53-mediated mechanism. The combination therapeutic resistance developed in isogenic p53 null HCT116 cells and siRNA-mediated p53 knockdown HCT116 cells, but the exogenous expression of p53 in p53 null isogenic cells resulted in the induction of cell death. In addition, we investigated an increased level of intracellular ROS (reactive oxygen species), which was preceded by p53 activation. The expression level of PUMA (p53-upregulated modulator of apoptosis), but not Bim, was significantly increased in HCT116 cells in response to the combination treatment. Taken together, our results demonstrate that combination therapy with sulindac and vitamin C could be a novel anti-cancer therapeutic strategy for p53 wild type colon cancers.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Ácido Ascórbico/metabolismo , Neoplasias do Colo/tratamento farmacológico , Espécies Reativas de Oxigênio/agonistas , Sulindaco/farmacologia , Proteína Supressora de Tumor p53/agonistas , Anti-Inflamatórios não Esteroides/farmacologia , Antioxidantes/metabolismo , Proteínas Reguladoras de Apoptose/agonistas , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Carcinoma/dietoterapia , Carcinoma/tratamento farmacológico , Carcinoma/metabolismo , Neoplasias do Colo/dietoterapia , Neoplasias do Colo/metabolismo , Terapia Combinada , Suplementos Nutricionais , Resistencia a Medicamentos Antineoplásicos , Interações Alimento-Droga , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células HCT116 , Humanos , Concentração Osmolar , Oxidantes/metabolismo , Proteínas Proto-Oncogênicas/agonistas , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Interferência de RNA , Espécies Reativas de Oxigênio/metabolismo , Proteína Supressora de Tumor p53/antagonistas & inibidores , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
3.
Br J Pharmacol ; 173(11): 1728-41, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26894321

RESUMO

BACKGROUND AND PURPOSE: Pulmonary disease is the main cause of morbidity and mortality in cystic fibrosis (CF) patients due to exacerbated inflammation. To date, the only anti-inflammatory drug available to CF patients is high-dose ibuprofen, which can slow pulmonary disease progression, but whose cyclooxygenase-dependent digestive adverse effects limit its clinical use. Here we have tested sulindac, another non-steroidal anti-inflammatory drug with an undefined anti-inflammatory effect in CF airway epithelial cells. EXPERIMENTAL APPROACH: Using in vitro and in vivo models, we NF-κB activity and IL-8 secretion. In HeLa-F508del cells, we performed luciferase reporter gene assays in order to measure i) IL-8 promoter activity, and ii) the activity of synthetic promoter containing NF-κB responsive elements. We quantified IL-8 secretion in airway epithelial CFBE cells cultured at an air-liquid interface and in a mouse model of CF. KEY RESULTS: Sulindac inhibited the transcriptional activity of NF-κB and decreased IL-8 transcription and secretion in TNF-α stimulated CF cells via a cyclooxygenase-independent mechanism. This effect was confirmed in vivo in a mouse model of CF induced by intra-tracheal instillation of LPS, with a significant decrease of the induction of mRNA for MIP-2, following treatment with sulindac. CONCLUSION AND IMPLICATIONS: Overall, sulindac decrease lung inflammation by a mechanism independent of cycolooxygenase. This drug could be beneficially employed in CF.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Fibrose Cística/tratamento farmacológico , Prostaglandina-Endoperóxido Sintases/metabolismo , Sulindaco/farmacologia , Animais , Anti-Inflamatórios não Esteroides/administração & dosagem , Linhagem Celular , Fibrose Cística/metabolismo , Células HeLa , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Sulindaco/administração & dosagem
4.
Rev. Esc. Enferm. USP ; 48(spe): 45-52, 08/2014. tab
Artigo em Inglês | LILACS, BDENF | ID: lil-731291

RESUMO

Objective To explore beliefs, values and practices related to the use of medicinal plants among low-income black families. Method The research method was ethnography and the participant observation process was done in a low-income community in the peripheral area of the City of São Paulo. Twenty black women were interviewed. Results Two cultural sub-themes, I do use medicines that I learned to make with my mother and with religious practitioners to care for diseases and Home medicines are to treat problems that are not serious, and the cultural theme I do use home medicines to treat simple diseases because I always have them at my disposal, they are free and I don’t need a medical prescription represent beliefs, values, and practices related to the use of medicinal plants among low-income black families. Conclusion The development of such practices, which can hide ethnic and social vulnerability, reveals the resilience of low-income black women in the process of confronting problems during the health-illness process. .


Objetivo Explorar las creencias, valores y prácticas sobre el uso de las plantas medicinales entre las familias negras de bajos ingresos. Método El método de investigación fue la etnografía y el proceso de observación participante fue desarrollado en una comunidad de bajos ingresos en las afueras de la Ciudad de São Paulo. Se entrevistó a veinte mujeres negras. Resultados Dos subtemas culturales Uso remedios que aprendí a hacer con mi madre y con los religiosos para cuidar de enfermedades y Remedios caseros se utilizan para resolver problemas que no son graves y el tema cultural Uso remedio casero para resolver enfermedades simples porque tengo todo lo que necesito, es gratuito y no necesita una receta médica simbolizam las prácticas de las mujeres. Conclusión Estas prácticas, que pueden estar enmascarando vulnerabilidades étnicas y sociales, ponen de manifiesto la resiliencia de las mujeres negras de bajos ingresos en el confrontamiento de los problemas del proceso salud-enfermedad. .


Objetivo Explorar crenças, valores e práticas relativas ao uso das plantas medicinais entre famílias negras de baixa renda. Método Pesquisa etnográfica cujo processo de observação participante foi desenvolvido em uma comunidade de baixa renda da periferia da Cidade de São Paulo. Vinte mulheres negras foram entrevistadas. Resultados Dois subtemas culturais, Uso remédios que aprendi a fazer com minha mãe e com os religiosos para cuidar das doenças e Remédios caseiros servem para resolver problemas que não são graves, e o tema cultural Uso remédio caseiro para resolver doenças simples, pois tenho sempre que preciso, é de graça e não precisa de receita médica representam as crenças, valores e práticas relativos ao uso das plantas medicinais entre famílias negras de baixa renda. Conclusão O desenvolvimento dessas práticas, que pode estar mascarando vulnerabilidades étnicas e sociais, revela a resiliência das mulheres negras de baixa renda no enfrentamento dos problemas que encontram no processo saúde-enfermidade. .


Assuntos
Humanos , Anti-Inflamatórios não Esteroides/farmacologia , Apoptose/efeitos dos fármacos , Endotélio Vascular/efeitos dos fármacos , Sulindaco/análogos & derivados , Sulindaco/farmacologia , Inibidores da Angiogênese/farmacologia , Ciclo Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Relação Dose-Resposta a Droga , Endotélio Vascular/citologia , Endotélio Vascular/ultraestrutura , Microscopia Eletrônica , Veias Umbilicais
5.
Mediators Inflamm ; 2013: 289789, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23935247

RESUMO

Anticancer activity of many herbs was observed for hundreds of years. They act as modifiers of biologic response, and their effectiveness may be increased by combining multiple herbal extracts . PERVIVO, traditional digestive herbal remedy, contains some of them, and we previously described its antiangiogenic activity. Numerous studies documented anticancer effects of nonsteroidal anti-inflammatory drugs. We were the first to show that sulindac and its metabolites inhibit angiogenesis. In the present paper the combined in vivo effect of multicomponent herbal remedy PERVIVO and nonsteroidal anti-inflammatory drug sulindac on tumor growth, tumor angiogenesis, and tumor volume in Balb/c mice was studied. These effects were checked after grafting cells collected from syngeneic sarcoma L-1 tumors into mice skin. The strongest inhibitory effect was observed in experimental groups treated with PERVIVO and sulindac together. The results of our investigation showed that combined effect of examined drugs may be the best way to get the strongest antiangiogenic and antitumor effect.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Preparações de Plantas/farmacologia , Sarcoma/tratamento farmacológico , Sulindaco/farmacologia , Inibidores da Angiogênese/farmacologia , Animais , Antineoplásicos/farmacologia , Artemisia absinthium/metabolismo , Linhagem Celular Tumoral , Quimioterapia Combinada , Feminino , Zingiber officinale/química , Camundongos , Camundongos Endogâmicos BALB C , Neovascularização Patológica/tratamento farmacológico , Extratos Vegetais/farmacologia , Pele/irrigação sanguínea
6.
Anticancer Agents Med Chem ; 13(10): 1540-50, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23848206

RESUMO

Previous studies indicate that extracts and purified components from Garcinia species inhibit the growth of human colon cancer cells. Garcinia benzophenones activate the expression of genes in the endoplasmic reticulum and cellular energy stress (mTOR) pathways. This study examines the growth inhibitory and synergistic effects of Garcinia benzophenones, alone or combined with chemopreventive agents, on human colon cancer cells. To find optimal combination treatments, HT29 colon cancer cells were treated with benzophenones alone, or combined with chemopreventive agents, and cell growth measured using the MTT assay. To reveal effects on signaling pathways, we assessed effects of the MEK inhibitor U0126 and the ER IP3 receptor antagonist heparin, as well as effects on the phosphorylation of 4E-BP-1 (mTOR pathway), using Western blot analysis. New and known benzophenones from Garcinia intermedia inhibited the growth of human colon cancer cells; an alcohol extract of Garcinia xanthochymus, as well as purified guttiferones (guttiferone E and xanthochymol), preferentially inhibited the growth of colon cancer versus nonmalignant intestinal epithelial cells. Guttiferone E exhibited synergy with the NSAID sulindac sulfide and xanthochymol, with the spice turmeric. Guttiferone A did not alter phosphorylation of 4E-BP-1, indicating that the mTORC1 pathway is not involved in its action. The effects of xanthochymol were enhanced by U0126, at low doses, and were blocked by heparin, indicating that the MEK pathway is involved, while the ER IP3 receptor is critical for its action. These studies indicate the potential of benzophenones, alone or combined with sulindac sulfide or turmeric, to prevent and treat colon cancer.


Assuntos
Antineoplásicos Fitogênicos/química , Benzofenonas/química , Garcinia/química , Regulação Neoplásica da Expressão Gênica , Extratos Vegetais/química , Sulindaco/análogos & derivados , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Antineoplásicos Fitogênicos/isolamento & purificação , Antineoplásicos Fitogênicos/farmacologia , Benzofenonas/isolamento & purificação , Benzofenonas/farmacologia , Butadienos/farmacologia , Celecoxib , Proteínas de Ciclo Celular , Linhagem Celular Tumoral , Curcuma , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/metabolismo , Inibidores Enzimáticos/farmacologia , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Heparina/farmacologia , Humanos , Receptores de Inositol 1,4,5-Trifosfato/genética , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina , Proteínas Quinases Ativadas por Mitógeno/genética , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , Nitrilas/farmacologia , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Extratos Vegetais/farmacologia , Pirazóis/farmacologia , Transdução de Sinais , Relação Estrutura-Atividade , Sulfonamidas/farmacologia , Sulindaco/farmacologia , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo
7.
J Hematol Oncol ; 6: 8, 2013 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-23327547

RESUMO

BACKGROUND: Both selenium and non-steroidal anti-inflammatory drug (NSAID) sulindac are effective in cancer prevention, but their effects are affected by several factors including epigenetic alterations and gene expression. The current study was designed to determine the effects of the combination of selenium and sulindac on tumor inhibition and the underlying mechanisms. RESULTS: We fed the intestinal tumor model Apc/p21 mice with selenium- and sulindac-supplemented diet for 24 weeks, and found that the combination of selenium and sulindac significantly inhibited intestinal tumorigenesis, in terms of reducing tumor incidence by 52% and tumor multiplicities by 80% (p<0.01). Mechanistic studies revealed that the combination of selenium and sulindac led to the significant induction of the expression of p27 and p53 and JNK1 phosphorylation, and led to the suppression of ß-catenin and its downstream targets. Impressively, the data also showed that demythelation on p21 promoter was associated with tumor inhibition by the combination of selenium and sulindac. CONCLUSIONS: The selenium is synergistic with sulindac to exert maximal effects on tumor inhibition. This finding provides an important chemopreventive strategy using combination of anti-cancer agents, which has a great impact on cancer prevention and has a promising translational potential.


Assuntos
Proteína da Polipose Adenomatosa do Colo/fisiologia , Anti-Inflamatórios não Esteroides/farmacologia , Transformação Celular Neoplásica/efeitos dos fármacos , Inibidor de Quinase Dependente de Ciclina p21/fisiologia , Neoplasias Intestinais/prevenção & controle , Selênio/farmacologia , Sulindaco/farmacologia , Animais , Western Blotting , Inibidor de Quinase Dependente de Ciclina p27/genética , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Metilação de DNA , Modelos Animais de Doenças , Sinergismo Farmacológico , Quimioterapia Combinada , Feminino , Neoplasias Intestinais/genética , Neoplasias Intestinais/patologia , Masculino , Camundongos , Camundongos Knockout , Proteína Quinase 8 Ativada por Mitógeno/genética , Proteína Quinase 8 Ativada por Mitógeno/metabolismo , Fosforilação , Regiões Promotoras Genéticas , Reação em Cadeia da Polimerase em Tempo Real , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
8.
Br J Pharmacol ; 162(7): 1521-33, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21175575

RESUMO

BACKGROUND AND PURPOSE: The use of non-steroidal anti-inflammatory drugs (NSAIDs) in the treatment of rheumatoid arthritis (RA) is limited by their toxicity. We evaluated the anti-inflammatory efficacy and safety of three novel modified NSAIDs, phospho-aspirin, phospho-ibuprofen and phospho-sulindac. EXPERIMENTAL APPROACH: We determined the anti-inflammatory effects and gastrointestinal safety of the phospho-NSAIDs in the rat adjuvant arthritis model and studied their mechanism of action in cultured cells, Cytokines were measured with elisa and activation of nuclear factor-κB (NF-κB) by immunohistochemistry. KEY RESULTS: All three phospho-NSAIDs showed less gastrointestinal toxicity than their parent compounds and demonstrated strong anti-inflammatory effects, essentially reversing joint inflammation and oedema. They have a broad but not uniform effect on the expression of relevant cytokines, in general decreasing IL-6 and IL-1ß and increasing IL-10 levels in rat plasma and cultured cells. Phospho-sulindac and phospho-ibuprofen but not phospho-aspirin suppressed PGE(2) production in vitro, whereas phospho-aspirin (in contrast to aspirin) showed the same effect in vivo. In joint tissues, phospho-aspirin inhibited NF-κB activation, and suppressed inflammation and bone resorption. Phospho-aspirin also inhibited Jurkat T cell proliferation. In general, phospho-aspirin had greater efficacy but different effects upon inflammatory mediators compared with aspirin. The chemical modification of the parent NSAIDs seems crucial for their safety and efficacy. CONCLUSIONS AND IMPLICATIONS: Phospho-aspirin, phospho-ibuprofen and phospho-sulindac were safer than their parent NSAIDs, were highly effective in rat adjuvant arthritis and inhibited many key mediators in the pathophysiology of RA. These novel compounds are promising candidate drugs for the treatment of RA and merit further evaluation.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Artrite Experimental/tratamento farmacológico , Animais , Anti-Inflamatórios não Esteroides/toxicidade , Artrite Experimental/imunologia , Aspirina/análogos & derivados , Aspirina/farmacologia , Aspirina/toxicidade , Linhagem Celular , Citocinas/biossíntese , Citocinas/genética , Dinoprostona/antagonistas & inibidores , Dinoprostona/biossíntese , Dinoprostona/metabolismo , Gastroenteropatias/induzido quimicamente , Humanos , Ibuprofeno/análogos & derivados , Ibuprofeno/farmacologia , Ibuprofeno/toxicidade , Células Jurkat , Camundongos , NF-kappa B/metabolismo , Células NIH 3T3 , Organofosfatos/farmacologia , Organofosfatos/toxicidade , Compostos Organofosforados/farmacologia , Compostos Organofosforados/toxicidade , Ratos , Sulindaco/análogos & derivados , Sulindaco/farmacologia , Sulindaco/toxicidade
9.
J Pathol ; 223(3): 378-89, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21171083

RESUMO

TNF-related apoptosis-inducing ligand (TRAIL) receptor agonistic agents and non-steroidal anti-inflammatory drugs (NSAIDs) are interesting agents for the chemoprevention and treatment of colorectal cancer. We investigated whether NSAIDs sensitize colon cancer and adenoma cell lines and ex vivo cultured human adenomas to recombinant human (rh)TRAIL. Involvement of the crucial Wnt signalling pathway in the sensitization of colon cancer cells was examined. Five colon cancer and two adenoma cell lines, human ex vivo adenomas and normal colonic epithelium were treated with aspirin or sulindac combined with rhTRAIL. Apoptosis levels, expression of intracellular proteins and TRAIL receptor membrane expression were assessed. Ls174T cells stably transfected with an inducible dominant negative TCF-4 (dnTCF-4) construct served to analyse the role of Wnt pathway activation. Both rhTRAIL-sensitive and -resistant colon cancer cell lines were strongly sensitized to rhTRAIL by aspirin (maximum enhancement ratio, 7.1). Remarkably, in adenoma cell lines sulindac enhanced rhTRAIL-induced apoptosis most effectively (maximum enhancement ratio, 2.5). Although membrane TRAIL receptor expression was not affected by NSAIDs, caspase-8 activation was enhanced by combinational treatment. Several proteins from different biological pathways were affected by NSAIDs, indicating complex mechanisms of sensitization. Elimination of TCF-4 completely blocked the sensitizing effect in colon cancer cells. In ex vivo adenomas the combination of sulindac and rhTRAIL increased apoptosis from 18.4% (sulindac) and 17.8% (rhTRAIL) to 28.0% (p = 0.003 and p = 0.005, respectively). It was concluded that NSAID-induced sensitization to rhTRAIL requires TCF-4 activity. Thus, the combination of TRAIL-receptor agonistic agents and NSAIDs is a potentially attractive treatment option for (pre)malignant tumours with constitutively active Wnt signalling, such as colorectal tumours.


Assuntos
Adenoma/patologia , Anti-Inflamatórios não Esteroides/farmacologia , Antineoplásicos/farmacologia , Neoplasias do Colo/patologia , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Adenoma/metabolismo , Adulto , Idoso , Apoptose/efeitos dos fármacos , Aspirina/farmacologia , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/fisiologia , Células CACO-2 , Colo/efeitos dos fármacos , Neoplasias do Colo/metabolismo , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos/métodos , Sinergismo Farmacológico , Feminino , Humanos , Mucosa Intestinal/efeitos dos fármacos , Masculino , Pessoa de Meia-Idade , Proteínas de Neoplasias/metabolismo , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Proteínas Recombinantes/farmacologia , Transdução de Sinais/efeitos dos fármacos , Sulindaco/farmacologia , Fator de Transcrição 4 , Fatores de Transcrição/fisiologia , Células Tumorais Cultivadas , Proteínas Wnt/fisiologia
10.
Cell Mol Life Sci ; 67(5): 797-806, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20091083

RESUMO

Sulindac is a non-selective inhibitor of cyclooxygenases (COX) used to treat inflammation and pain. Additionally, non-COX targets may account for the drug's chemo-preventive efficacy against colorectal cancer and reduced gastrointestinal toxicity. Here, we demonstrate that the pharmacologically active metabolite of sulindac, sulindac sulfide (SSi), targets 5-lipoxygenase (5-LO), the key enzyme in the biosynthesis of proinflammatory leukotrienes (LTs). SSi inhibited 5-LO in ionophore A23187- and LPS/fMLP-stimulated human polymorphonuclear leukocytes (IC(50) approximately 8-10 microM). Importantly, SSi efficiently suppressed 5-LO in human whole blood at clinically relevant plasma levels (IC(50) = 18.7 microM). SSi was 5-LO-selective as no inhibition of related lipoxygenases (12-LO, 15-LO) was observed. The sulindac prodrug and the other metabolite, sulindac sulfone (SSo), failed to inhibit 5-LO. Mechanistic analysis demonstrated that SSi directly suppresses 5-LO with an IC(50) of 20 muM. Together, these findings may provide a novel molecular basis to explain the COX-independent pharmacological effects of sulindac under therapy.


Assuntos
Anti-Inflamatórios/farmacologia , Inibidores de Lipoxigenase , Sulindaco/análogos & derivados , Proteínas Ativadoras de 5-Lipoxigenase , Anti-Inflamatórios/uso terapêutico , Araquidonato 5-Lipoxigenase/metabolismo , Sangue/efeitos dos fármacos , Sangue/metabolismo , Proteínas de Transporte/antagonistas & inibidores , Proteínas de Transporte/metabolismo , Sistema Livre de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Humanos , Inibidores de Lipoxigenase/farmacologia , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/metabolismo , Modelos Biológicos , Neutrófilos/efeitos dos fármacos , Neutrófilos/enzimologia , Neutrófilos/metabolismo , Concentração Osmolar , Transporte Proteico/efeitos dos fármacos , Sulindaco/farmacologia , Sulindaco/uso terapêutico
11.
Mol Cancer Ther ; 6(6): 1736-44, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17541034

RESUMO

The design of novel targeted or combination therapies may improve treatment options for pancreatic cancer. Two targets of recent interest are nuclear factor-kappaB (NF-kappaB) and cyclooxygenase (COX), known to be activated or overexpressed, respectively, in pancreatic cancer. We have previously shown that parthenolide, a proapoptotic drug associated with NF-kappaB inhibition, enhanced the growth suppression of pancreatic cancer cells by the COX inhibitor sulindac in vitro. In the present study, a bioavailable analogue of parthenolide, LC-1, and sulindac were evaluated in vivo using a xenograft model of human pancreatic cancer. Treatment groups included placebo, low-dose/high-dose LC-1 (20 and 40 mg/kg), low-dose/high-dose sulindac (20 and 60 mg/kg), and low-dose combination LC-1/sulindac (20 mg/kg each). In MiaPaCa-2 xenografts, tumor growth was inhibited by either high-dose sulindac or LC-1. In BxPC-3 xenografts, tumor size was significantly reduced by treatment with the low-dose LC-1/sulindac combination or high-dose sulindac alone (P < 0.05). Immunohistochemistry of BxPC-3 tumors revealed a significant decrease in Ki-67 and CD31 staining by high-dose sulindac, with no significant changes in COX-1/COX-2 levels or activity in any of the treatment groups. NF-kappaB DNA-binding activity was significantly decreased by high-dose LC-1. Cyclin D1 protein levels were reduced by the low-dose LC-1/sulindac combination or high-dose sulindac alone, correlating with BxPC-3 tumor suppression. These results suggest that LC-1 and sulindac may mediate their antitumor effects, in part, by altering cyclin D1 levels. Furthermore, this study provides preclinical evidence for the therapeutic efficacy of these agents.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Ciclina D1/antagonistas & inibidores , Neoplasias Pancreáticas/patologia , Sesquiterpenos/farmacologia , Sulindaco/farmacologia , Animais , Linhagem Celular Tumoral , Humanos , Camundongos , Camundongos Nus , NF-kappa B/metabolismo , Neoplasias Pancreáticas/metabolismo , Prostaglandina-Endoperóxido Sintases/metabolismo
12.
J Biol Chem ; 281(42): 31279-89, 2006 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-16899457

RESUMO

Gamma-secretase is one of the critical enzymes required for the generation of amyloid-beta peptides from the beta-amyloid precursor protein. Because amyloid-beta peptides are generally accepted to play a key role in Alzheimer disease, gamma-secretase inhibition holds the promise for a disease-modifying therapy for this neurodegenerative condition. Although recent progress has enhanced the understanding of the biology and composition of the gamma-secretase enzyme complex, less information is available on the actual interaction of various inhibitor classes with the enzyme. Here we show that the two principal classes of inhibitor described in the scientific and patent literature, aspartyl protease transition state analogue and small molecule non-transition state inhibitors, display fundamental differences in the way they interact with the enzyme. Taking advantage of a gamma-secretase enzyme overexpressing cellular system and different radiolabeled gamma-secretase inhibitors, we observed that the maximal binding of non-transition state gamma-secretase inhibitors accounts only for half the number of catalytic sites of the recombinant enzyme complex. This characteristic stoichiometry can be best accommodated with a model whereby the non-transition state inhibitors bind to a unique site at the interface of a dimeric enzyme. Subsequent competition studies confirm that this site appears to be targeted by the main classes of small molecule gamma-secretase inhibitor. In contrast, the non-steroidal anti-inflammatory drug gamma-secretase modulator sulindac sulfide displayed noncompetitive antagonism for all types of inhibitor. This finding suggests that non-steroidal anti-inflammatory drug-type gamma-secretase modulators target an alternative site on the enzyme, thereby changing the conformation of the binding sites for gamma-secretase inhibitors.


Assuntos
Secretases da Proteína Precursora do Amiloide/química , Secretases da Proteína Precursora do Amiloide/metabolismo , Anti-Inflamatórios não Esteroides/farmacologia , Ligação Competitiva , Bioquímica/métodos , Domínio Catalítico , Linhagem Celular , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos/métodos , Inibidores Enzimáticos/farmacologia , Humanos , Cinética , Ligantes , Modelos Químicos , Ligação Proteica , Sulindaco/análogos & derivados , Sulindaco/farmacologia
13.
Int J Mol Med ; 14(4): 707-11, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15375605

RESUMO

Angiogenesis, the process of new blood vessel formation, is the key event in the mechanism of several pathological processes including diabetic retinopathy. The physiological control of angiogenesis depends on the balance between stimulatory and inhibitory factors. Therefore, a number of anti-angiogenic approaches has been developed, many of them based on the inhibition of the functional activity of pro-angiogenic factors. The aim of the present study was to compare the anti-angiogenic effectiveness of sulindac sulfone and some herbal compounds in the serum-induced angiogenesis test performed in Balb/c mice. Pooled sera from 35 patients with diabetes type 2 and retinopathy were used as pro-angiogenic stimuli. The strongest inhibitory effect was observed for the sulindac sulfone and ursolic acid in the highest concentration of 200 micro g/ml, as well as for the low-dosage concomitant treatment with 2 micro g/ml of epigallocatechin gallate (EGCG, green tea flavanol), ursolic acid (plant-derived triterpenoid), sulindac sulfone and convalamaroside (steroidal saponin). Combination treatment was significantly more effective than monotherapy with medium (20 micro g/ml) or lowest doses of tested compounds. The present study is the first to demonstrate the potent anti-angiogenic effect of the combination therapy comprising of plant-derived extracts and sulindac sulfone, as tested in the in vivo angiogenesis experimental model with sera of non-proliferative diabetic retinopathy patients used as the pro-angiogenic stimuli. We think that it might be the first step toward application of some of these compounds, in the future, in preventive anti-angiogenic therapy of these patients, as well, as in the treatment of later, proliferative stage of this disease.


Assuntos
Indutores da Angiogênese/antagonistas & inibidores , Diabetes Mellitus Tipo 2/sangue , Diabetes Mellitus Tipo 2/complicações , Retinopatia Diabética/complicações , Neovascularização Fisiológica/efeitos dos fármacos , Plantas Medicinais/química , Sulindaco/análogos & derivados , Sulindaco/farmacologia , Animais , Proliferação de Células/efeitos dos fármacos , Retinopatia Diabética/sangue , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Soro/química
14.
J Biol Chem ; 278(48): 47762-75, 2003 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-14506281

RESUMO

Sulindac, a non-steroidal anti-inflammatory prodrug, is metabolized into pharmacologically active sulfide and sulfone derivatives. Sulindac sulfide, but not sulindac sulfone, inhibits cyclooxygenase (COX) enzyme activities, yet both derivatives have growth inhibitory effects on colon cancer cells. Microarray analysis was used to detect COX-independent effects of sulindac on gene expression in human colorectal cells. Spermidine/sperm-ine N1-acetyltransferase (SSAT) gene, which encodes a polyamine catabolic enzyme, was induced by clinically relevant sulindac sulfone concentrations. Northern blots confirmed increased SSAT RNA levels in these colon cancer cells. Deletion analysis and mutational studies were done to map the sulindac sulfone-dependent response sequences in the SSAT 5'-flanking sequences. This led us to the identification of two peroxisome proliferator-activated receptor (PPAR) response elements (PPREs) in the SSAT gene. PPRE-2, at +48 bases relative to the transcription start site, is required for the induction of SSAT by sulindac sulfone and is specifically bound by PPAR gamma in the Caco-2 cells as shown by transfection and gel shift experiments. PPRE-1, at-323 bases relative to the start site, is not required for the induction of SSAT by sulindac sulfone but can be bound by both PPAR delta and PPAR gamma. Sulindac sulfone reduced cellular polyamine contents in the absence but not in the presence of verapamil, an inhibitor of the export of monoacetyl diamines, inhibited cell proliferation and induced apoptosis. The induced apoptosis could be partially rescued by exogenous putrescine. These data suggest that apoptosis induced by sulindac sulfone is mediated, in part, by the COX-independent, PPAR-dependent transcriptional activation of SSAT, leading to reduced tissue polyamine contents in human colon cancer cells.


Assuntos
Apoptose , Neoplasias do Colo/metabolismo , Poliaminas/química , Prostaglandina-Endoperóxido Sintases/metabolismo , Sulindaco/análogos & derivados , Sulindaco/farmacologia , Acetiltransferases/genética , Northern Blotting , Western Blotting , Células CACO-2 , Membrana Celular/metabolismo , Sobrevivência Celular , Ciclo-Oxigenase 2 , DNA Complementar/metabolismo , Relação Dose-Resposta a Droga , Humanos , Immunoblotting , Isoenzimas/metabolismo , Proteínas de Membrana , Modelos Biológicos , Modelos Genéticos , Análise de Sequência com Séries de Oligonucleotídeos , Oligonucleotídeos/química , Plasmídeos/metabolismo , Regiões Promotoras Genéticas , Ligação Proteica , Biossíntese de Proteínas , Putrescina/química , RNA/metabolismo , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/metabolismo , Elementos de Resposta , Espermidina/metabolismo , Fatores de Tempo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transcrição Gênica , Ativação Transcricional , Transfecção
15.
J Pharm Pharmacol ; 55(4): 519-26, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12803774

RESUMO

Abnormal vascular smooth muscle cell (VSMC) proliferation plays an important role in the pathogenesis of both atherosclerosis and restenosis. Recent studies suggest that high-dose salicylates, in addition to inhibiting cyclooxygenase activity, exert an antiproliferative effect on VSMC growth both in-vitro and in-vivo. However, whether all non-steroidal anti-inflammatory drugs (NSAIDs) exert similar antiproliferative effects on VSMCs, and do so via a common mechanism of action, remains to be shown. In this study, we demonstrate that the NSAIDs aspirin, sodium salicylate, diclofenac, ibuprofen, indometacin and sulindac induce a dose-dependent inhibition of proliferation in rat A10 VSMCs in the absence of significant cytotoxicity. Flow cytometric analyses showed that exposure of A10 cells to diclofenac, indometacin, ibuprofen and sulindac, in the presence of the mitotic inhibitor, nocodazole, led to a significant G0/G1 arrest. In contrast, the salicylates failed to induce a significant G1 arrest since flow cytometry profiles were not significantly different from control cells. Cyclin A levels were elevated, and hyperphosphorylated p107 was present at significant levels, in salicylate-treated A10 cells, consistent with a post-G1/S block, whereas cyclin A levels were low, and hypophosphorylated p107 was the dominant form, in cells treated with other NSAIDs consistent with a G1 arrest. The ubiquitously expressed cyclin-dependent kinase (CDK) inhibitors, p21 and p27, were increased in all NSAID-treated cells. Our results suggest that diclofenac, indometacin, ibuprofen and sulindac inhibit VSMC proliferation by arresting the cell cycle in the G1 phase, whereas the growth inhibitory effect of salicylates probably affects the late S and/or G2/M phases. Irrespective of mechanism, our results suggest that NSAIDs might be of benefit in the treatment of certain vasculoproliferative disorders.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Divisão Celular/efeitos dos fármacos , Animais , Anti-Inflamatórios não Esteroides/administração & dosagem , Aorta/citologia , Aorta/efeitos dos fármacos , Aspirina/administração & dosagem , Aspirina/farmacologia , Linhagem Celular/efeitos dos fármacos , Linhagem Celular/metabolismo , Diclofenaco/administração & dosagem , Diclofenaco/farmacologia , Relação Dose-Resposta a Droga , Citometria de Fluxo , Ibuprofeno/administração & dosagem , Ibuprofeno/farmacologia , Indometacina/administração & dosagem , Indometacina/farmacologia , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Ratos , Salicilato de Sódio/administração & dosagem , Salicilato de Sódio/farmacologia , Sulindaco/administração & dosagem , Sulindaco/farmacologia
16.
Mutat Res ; 523-524: 119-25, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12628509

RESUMO

Herbal medicines are now attracting attention as potential sources of cancer preventive agents. Using inhibition of tumor necrosis factor-alpha (TNF-alpha) release assay, we studied Acer nikoense, Megusurino-ki in Japanese. Inhibitory potential was found in the leaf extract, and the main active principles were identified as geraniin and corilagin. The IC(50) values for TNF-alpha release inhibition were 43 microM for geraniin and 76 microM for corilagin, whereas that for (-)-epigallocatechin gallate (EGCG), the green tea polyphenol, as control was 26 microM. Furthermore, treatment with geraniin inhibited okadaic acid tumor promotion in a two-stage carcinogenesis experiment on mouse skin. Geraniin and corilagin are present in another well-known Japanese traditional herb, Geranium thunbergii, Genno-shoko in Japanese. Considering seasonal variations of the agents and sites of cultivation of herbs, this paper reviews the significance of geraniin as a new cancer preventive agent. In addition, based on accumulated results of green tea as a cancer preventive, we review two important results with EGCG: the synergistic effects of EGCG with sulindac or tamoxifen on cancer preventive activity in PC-9 cells, and cancer prevention of intestinal tumor development in multiple intestinal neoplasia (Min) mice by cotreatment using EGCG with sulindac. We report here new findings on additional gene expression resulting from cotreatment with EGCG and sulindac in PC-9 cells compared with gene expression by EGCG alone or sulindac alone. Overall, our results indicate that, with the continuing spread of cancer chemoprevention as a fundamental medical strategy, both clinicians and researchers should take a closer look at herbal medicine.


Assuntos
Anticarcinógenos/farmacologia , Antineoplásicos/farmacologia , Catequina/análogos & derivados , Catequina/farmacologia , Taninos Hidrolisáveis , Estruturas Vegetais/química , Sulindaco/farmacologia , Tamoxifeno/farmacologia , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/metabolismo , Células 3T3 , Animais , Glucosídeos/farmacologia , Medicina Herbária , Japão , Medicina Tradicional , Camundongos , Taninos/farmacologia
17.
Carcinogenesis ; 24(2): 263-7, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12584176

RESUMO

Epidemiological and animal studies suggest that tea may be protective towards cancers of the GI tract. White tea, the least processed form of tea, contains high levels of polyphenols and, like green tea, is chemopreventive towards heterocyclic amine-initiated colonic aberrant crypt formation in male F344 rats. We examined for the first time the relative effectiveness of white and green tea in suppressing intestinal tumorigenesis in C57BL/6J-Apc(Min/+) (Apc(min)) mice. Each tea was also compared with sulindac, a non-steroidal anti-inflammatory drug known to be highly effective in Apc(min) mice. Male C57BL/6J (+/+) (wild-type) and Apc(min) mice were treated in the drinking water with white tea or green tea (1.5% w/v, 2 min brew-time), 80 p.p.m. sulindac, a combination of 80 p.p.m. sulindac in 1.5% white tea, or pH buffered water. After 12 weeks of treatment, Apc(min) mice given white tea, green tea, or sulindac had significantly fewer tumors than controls (P < 0.05). The protection provided by 1.5% green or white tea was comparable to that provided by 80 p.p.m. sulindac. Mice treated with a combination of white tea plus sulindac had significantly fewer tumors than either treatment alone (P < 0.05). beta-catenin and beta-catenin/Tcf-4 regulated proteins Cyclin D(1) and c-Jun were readily detected in polyps, but markedly reduced in normal-looking intestines of mice treated with both tea and sulindac. This research provides evidence that teas, particularly when administered in combination with sulindac, are highly effective at inhibiting intestinal neoplasia in male Apc(min) mice via direct or indirect effects on the beta-catenin/APC pathway.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Proteínas do Citoesqueleto/metabolismo , Regulação para Baixo/efeitos dos fármacos , Genes APC , Transdução de Sinais/efeitos dos fármacos , Sulindaco/farmacologia , Chá , Transativadores/metabolismo , Animais , Pólipos Intestinais/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , beta Catenina
18.
Physiol Genomics ; 11(2): 45-52, 2002 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-12388792

RESUMO

Oxidative injury and the resulting death of neurons is a major pathological factor involved in numerous neurodegenerative diseases. However, the development of drugs that target this mechanism remains limited. The goal of this study was to test a compound library of approved Food and Drug Administration drugs against a hydrogen peroxide-induced oxidant injury model in neuroblastoma cells. We identified 26 neuroprotective compounds, of which megestrol, meclizine, verapamil, methazolamide, sulindac, and retinol were examined in greater detail. Using large-scale oligonucleotide microarray analysis, we identified genes modulated by these drugs that might underlie the cytoprotection. Five key genes were either uniformly upregulated or downregulated by all six drug treatments, namely, tissue inhibitor of matrix metalloproteinase (TIMP1), ret-proto-oncogene, clusterin, galanin, and growth associated protein (GAP43). Exogenous addition of the neuropeptide galanin alone conferred survival to oxidant-stressed cells, comparable to that seen with the drugs. Our approach, which we term "interventional profiling," represents a general and powerful strategy for identifying new bioactive agents for any biological process, as well as identifying key downstream genes and pathways that are involved.


Assuntos
Bioensaio/métodos , Perfilação da Expressão Gênica/métodos , Doenças Neurodegenerativas/prevenção & controle , Fármacos Neuroprotetores/farmacologia , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Avaliação Pré-Clínica de Medicamentos/métodos , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Peróxido de Hidrogênio/antagonistas & inibidores , Peróxido de Hidrogênio/metabolismo , Meclizina/farmacologia , Megestrol/farmacologia , Metazolamida/farmacologia , Neuroblastoma/patologia , Doenças Neurodegenerativas/induzido quimicamente , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Oxidantes/efeitos adversos , Oxidantes/antagonistas & inibidores , Proto-Oncogene Mas , Sulindaco/farmacologia , Células Tumorais Cultivadas , Verapamil/farmacologia , Vitamina A/farmacologia
19.
Mutat Res ; 506-507: 121-7, 2002 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-12351151

RESUMO

There is growing interest in the potential health benefits of tea, and a recent report described the potent antimutagenic activity of white tea in comparison with green tea against several heterocyclic amines, including 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP) [Mutat. Res. 495 (2001) 61]. We compared the inhibitory effects of white and green teas with sulindac, a nonsteroidal anti-inflammatory agent, in two different mouse models of intestinal tumorigenesis. In the Apc(min) mouse, white and green teas given at human-relevant concentrations (1.5% w/v, 2-min brew), and sulindac (80 ppm in the drinking water), each suppressed polyp formation by approximately 50%, and the combination of white tea plus sulindac was more effective than either treatment alone (P=0.05). Mice expressing an N-terminally truncated, oncogenic version of beta-catenin (A 33(delta N beta-cat) mutant mice) developed colonic aberrant crypt foci (ACF) spontaneously, but PhIP treatment increased the incidence and number of ACF per colon. In the normal-looking intestinal mucosa of Apc(min) and A 33(delta N beta-cat) mice, white tea plus sulindac treatment markedly attenuated the expression of beta-catenin protein, and this was recapitulated in vitro in cells transiently transfected with beta-catenin plus Tcf-4 and treated with tea or the major tea polyphenol epigallocatechin-3-gallate (EGCG). Expression of a beta-catenin/Tcf reporter was inhibited by EGCG in the transfected cells, and the beta-catenin/Tcf target genes cyclin D1 and c-jun were downregulated in vivo by tea plus sulindac treatment. Collectively, the data support a chemopreventive role for tea and sulindac against intermediate and late stages of colon cancer, via effects on the beta-catenin/Tcf signaling pathway.


Assuntos
Proteína da Polipose Adenomatosa do Colo/metabolismo , Anti-Inflamatórios não Esteroides/farmacologia , Catequina/análogos & derivados , Neoplasias Intestinais/prevenção & controle , Extratos Vegetais/farmacologia , Pólipos/prevenção & controle , Sulindaco/farmacologia , Chá , Proteína da Polipose Adenomatosa do Colo/deficiência , Proteína da Polipose Adenomatosa do Colo/genética , Animais , Catequina/farmacologia , Linhagem Celular , Proteínas do Citoesqueleto/deficiência , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/metabolismo , Combinação de Medicamentos , Imidazóis/toxicidade , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Neoplasias Intestinais/induzido quimicamente , Neoplasias Intestinais/metabolismo , Luciferases/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Pólipos/induzido quimicamente , Pólipos/metabolismo , Transativadores/deficiência , Transativadores/genética , Transativadores/metabolismo , beta Catenina
20.
J Cancer Res Clin Oncol ; 127(1): 69-72, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11206275

RESUMO

Green tea is the most effective beverage for cancer prevention in humans. Looking at the concept of combination cancer chemoprevention, we previously reported the synergistic effects of (-)-epigallocatechin gallate (EGCG) with sulindac, and the additive effects of EGCG with tamoxifen, on cancer-preventive activity in human lung cancer cell line PC-9. This paper reports confirmation of the synergistic effects of EGCG with sulindac on the inhibition of intestinal tumors in multiple intestinal neoplasia (Min) mice. Treatment with both green tea extract and sulindac significantly reduced the number of tumors from 72.3 +/- 28.3 to 32.0 +/- 18.7 tumors per mouse, a decrease of 44.3%, whereas treatment with green tea extract alone or with sulindac alone reduced it to 56.7 +/- 3.5 and 49.0 +/- 12.7, respectively. The results also indicated that green tea extract inhibited tumor growth in Min mice almost as potently as sulindac itself did. The three treated groups did not show any adenocarcinomas, whereas 10.8% of the control group did. Since cancer-preventive agents like sulindac and tamoxifen are associated with adverse effects, we discuss the possibility of non-toxic, combination cancer chemoprevention with green tea, looking at the goal of truly effective cancer prevention.


Assuntos
Catequina/análogos & derivados , Neoplasias Intestinais/prevenção & controle , Extratos Vegetais/farmacologia , Sulindaco/farmacologia , Chá , Adenocarcinoma/prevenção & controle , Animais , Catequina/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA