Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 209
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Childs Nerv Syst ; 36(11): 2675-2684, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32385563

RESUMO

INTRODUCTION: Pilocytic astrocytoma (PA) is the most common brain tumor that affects the pediatric population. Even though PA is benign and treatment only involves surgery, recurrent or unresectable tumors require chemo- and radiotherapy. Besides BRAF, CDKN2A, or IDH mutations, the hyperactivation of the nuclear factor NF-κB contributes to tumor growth and survival. METHODS: In the present study, we used publicly available data for the in silico analysis of NF-κB subunits (RELA, RELB, REL, NF-κB1, and NF-κB2) expression in PA samples. Besides, in vitro assays were performed to evaluate proliferation, migration, cell death, on the PA cell line Res286 comparing to human primary astrocytes. Sensitization to radiation therapy and temozolomide (TMZ) was also assayed. RESULTS: Our results showed that all the members of the NF-kB family are upregulated in PA datasets compared to normal brain tissues. Moreover, DHMEQ treatment significantly reduced cell growth and motility, while sensitized cells to ionizing radiation and TMZ, as previously seen in high-grade gliomas. CONCLUSIONS: This drug presents a potential application in clinical practice for the treatment of recurrent or inoperable PA. Moreover, its use might assist adjuvant chemotherapy and reduce irradiation doses to avoid toxicity to the surrounding tissues.


Assuntos
Astrocitoma , Neoplasias Encefálicas , Astrocitoma/tratamento farmacológico , Neoplasias Encefálicas/tratamento farmacológico , Proliferação de Células , Criança , Humanos , NF-kappa B , Temozolomida/farmacologia
2.
Br J Cancer ; 110(12): 2965-74, 2014 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-24867687

RESUMO

BACKGROUND: Although T-cell immunity is thought to be involved in the prognosis of epithelial ovarian cancer (EOC) patients, immunosuppressive conditions hamper antitumour immune responses. Thus, their mechanisms and overcoming strategies need to be investigated. METHODS: The role of NF-κB in human EOC cells and macrophages was evaluated by in vitro production of immunosuppressive IL-6 and IL-8 by EOC cells and in vivo analysis of immune responses in nude mice implanted with human EOC cells using an NF-κB inhibitor DHMEQ. RESULTS: In EOC patients, increased plasma IL-6, IL-8, and arginase were observed. The NF-κB inhibitor DHMEQ inhibited the production of IL-6 and IL-8 by EOC cell lines. Immunosuppression of human DCs and macrophages by culture supernatant of EOC cells was reversed with the pretreatment of DHMEQ. Administration of DHMEQ to nude mice implanted with human EOC resulted in the restoration of T-cell stimulatory activity of murine DCs along with the reduction of tumour accumulation and arginase expression of MDSCs. Nuclear factor-κB inhibition in tumour-bearing mice also enhanced antitumour effects of transferred murine naive T cells. CONCLUSIONS: NF-κB is involved in the immunosuppression induced by human EOC, and its inhibitor may restore antitumour immune responses, indicating that NF-κB is an attractive target for EOC treatment.


Assuntos
Tolerância Imunológica , Interleucina-6/imunologia , Interleucina-8/imunologia , Neoplasias Epiteliais e Glandulares/imunologia , Neoplasias Ovarianas/imunologia , Fator de Transcrição RelA/imunologia , Transferência Adotiva , Animais , Arginase/sangue , Benzamidas/farmacologia , Carcinoma Epitelial do Ovário , Linhagem Celular Tumoral , Meios de Cultivo Condicionados/farmacologia , Cicloexanonas/farmacologia , Células Dendríticas/imunologia , Feminino , Humanos , Interleucina-6/biossíntese , Interleucina-6/sangue , Interleucina-8/biossíntese , Interleucina-8/sangue , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Transplante de Neoplasias , Transdução de Sinais/imunologia , Fator de Transcrição RelA/antagonistas & inibidores , Fator de Transcrição RelA/genética , Transplante Heterólogo
3.
Br J Cancer ; 107(4): 652-7, 2012 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-22805327

RESUMO

BACKGROUND: Inducible activation of nuclear factor (NF)-κB is one of the principal mechanisms through which resistant prostate cancer cells are protected from radiotherapy. We hypothesised that inactivation of inducible NF-κB with a novel NF-κB inhibitor, DHMEQ, would increase the therapeutic effects of radiotherapy. METHODS: PC-3 and LNCaP cells were exposed to irradiation and/or DHMEQ. Cell viability, cell cycle analysis, western blotting assay, and NF-κB activity were measured. The antitumour effect of irradiation combined with DHMEQ in vivo was also assessed. RESULTS: The combination of DHMEQ with irradiation resulted in cell growth inhibition and G2/M arrest relative to treatment with irradiation alone. Inducible NF-κB activity by irradiation was inhibited by DHMEQ treatment. The expression of p53 and p21 in LNCaP, and of 14-3-3σ in PC-3 cells, was increased in the combination treatment. In the in vivo study, 64 days after the start of treatment, tumour size was 85.1%, 77.1%, and 64.7% smaller in the combination treatment group than that of the untreated control, DHMEQ-treated alone, and irradiation alone groups, respectively. CONCLUSION: Blockade of NF-κB activity induced by radiation with DHMEQ could overcome radio-resistant responses and may become a new therapeutic modality for treating prostate cancer.


Assuntos
Antineoplásicos/uso terapêutico , Benzamidas/farmacologia , Cicloexanonas/farmacologia , NF-kappa B/antagonistas & inibidores , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/radioterapia , Radiossensibilizantes/uso terapêutico , Animais , Antineoplásicos/farmacologia , Benzamidas/uso terapêutico , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/efeitos da radiação , Divisão Celular/efeitos dos fármacos , Divisão Celular/efeitos da radiação , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Cicloexanonas/uso terapêutico , Humanos , Masculino , Camundongos , Camundongos Nus , Tolerância a Radiação/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Clin Exp Allergy ; 42(8): 1273-81, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22805475

RESUMO

BACKGROUND: Dehydroxymethylepoxyquinomicin (DHMEQ) is a newly developed compound that inhibits nuclear factor κB activation and is reported to ameliorate animal models of various inflammatory diseases without significant adverse effects. Because nuclear factor κB is a transcription factor that plays a critical role in the pathophysiology of asthma, DHMEQ may be of therapeutic benefit in asthma. OBJECTIVE: The purpose of this study was to evaluate the effects of DHMEQ on airway inflammation and remodelling in murine models of asthma. METHODS: The BALB/c mice were sensitized and then challenged acutely or chronically with ovalbumin and administered DHMEQ intraperitoneally before each challenge. Inflammation of airways, lung histopathology and airway hyper responsiveness to methacholine challenge were evaluated. In addition, the effect of DHMEQ on production of cytokines and eotaxin-1 by murine splenocytes, human peripheral blood mononuclear cells and bronchial epithelial cells was investigated. RESULTS: Airway hyper responsiveness was ameliorated in both acutely and chronically challenged models by treatment with DHMEQ. DHMEQ significantly reduced eosinophilic airway inflammation and levels of Th2 cytokines in bronchoalveolar lavage fluid in the acute model. It also inhibited parameters of airway remodelling including mucus production, peribronchial fibrosis and the expression of α-smooth muscle actin. Moreover, the production of Th2 cytokines from murine splenocytes and human peripheral blood mononuclear cells and the production of eotaxin-1 by bronchial epithelial cells were inhibited by DHMEQ. CONCLUSIONS AND CLINICAL RELEVANCE: These results indicate that DHMEQ inhibits allergic airway inflammation and airway remodelling in murine models of asthma. DHMEQ may have therapeutic potential in the treatment of asthma.


Assuntos
Remodelação das Vias Aéreas/efeitos dos fármacos , Asma/tratamento farmacológico , Benzamidas/farmacologia , Cicloexanonas/farmacologia , Inflamação/tratamento farmacológico , NF-kappa B/antagonistas & inibidores , Animais , Asma/metabolismo , Asma/patologia , Benzamidas/administração & dosagem , Líquido da Lavagem Broncoalveolar/imunologia , Células Cultivadas , Cicloexanonas/administração & dosagem , Citocinas/imunologia , Citocinas/metabolismo , Modelos Animais de Doenças , Eosinófilos/imunologia , Eosinófilos/patologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Feminino , Humanos , Inflamação/metabolismo , Inflamação/patologia , Mediadores da Inflamação/metabolismo , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , NF-kappa B/metabolismo , Hipersensibilidade Respiratória/tratamento farmacológico , Hipersensibilidade Respiratória/metabolismo
5.
Phys Rev Lett ; 108(3): 037002, 2012 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-22400776

RESUMO

We have performed high-resolution angle-resolved photoemission spectroscopy on Fe-based superconductor LiFeAs (T(c)=18 K). We reveal multiple nodeless superconducting (SC) gaps with 2Δ/k(B)T(c) ratios varying from 2.8 to 6.4, depending on the Fermi surface (FS). We also succeeded in directly observing a gap anisotropy along the FS with magnitude up to ~30%. The anisotropy is fourfold symmetric with an antiphase between the hole and electron FSs, suggesting complex anisotropic interactions for the SC pairing. The observed momentum dependence of the SC gap offers an excellent opportunity to investigate the underlying pairing mechanism.

6.
Clin Exp Immunol ; 166(2): 299-306, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21985376

RESUMO

Dehydroxymethylepoxyquinomicin (DHMEQ), a new nuclear factor (NF)-κB inhibitor, has several beneficial effects, including the suppression of tumour growth and anti-inflammatory effects. DHMEQ can also suppress the production of tumour necrosis factor (TNF)-α induced by lipopolysaccharide (LPS) in vitro. In the present study, we examine the effects of DHMEQ on TNF-α production in vivo and on the survival of mice injected with LPS. When DHMEQ was injected into mice 2 h before LPS injection, the survival of the LPS-injected mice was prolonged. When DHMEQ was injected twice (2 h before LPS injection and the day after LPS injection), all the mice were rescued. The injection of DHMEQ 1 h after LPS injection and the day after LPS injection also resulted in the rescue of all mice. The serum levels of TNF-α in the mice that received both LPS and DHMEQ were suppressed compared to the mice that received only LPS. These results suggest that DHMEQ can be utilized for the prevention and treatment of endotoxin shock.


Assuntos
Benzamidas/farmacologia , Cicloexanonas/farmacologia , Choque Séptico/tratamento farmacológico , Choque Séptico/imunologia , Fator de Necrose Tumoral alfa/biossíntese , Animais , Apoptose/efeitos dos fármacos , Núcleo Celular/metabolismo , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Interleucina-1beta/biossíntese , Interleucina-6/biossíntese , Lipopolissacarídeos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , RNA Mensageiro/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Choque Séptico/prevenção & controle , Baço/citologia , Fator de Necrose Tumoral alfa/sangue
7.
Br J Cancer ; 102(1): 206-12, 2010 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-19997106

RESUMO

BACKGROUND: Tumour-initiating cells (TICs) or cancer stem cells can exist as a small population in malignant tissues. The signalling pathways activated in TICs that contribute to tumourigenesis are not fully understood. METHODS: Several breast cancer cell lines were sorted with CD24 and CD44, known markers for enrichment of breast cancer TICs. Tumourigenesis was analysed using sorted cells and total RNA was subjected to gene expression profiling and gene set enrichment analysis (GSEA). RESULTS: We showed that several breast cancer cell lines have a small population of CD24(-/low)/CD44(+) cells in which TICs may be enriched, and confirmed the properties of TICs in a xenograft model. GSEA revealed that CD24(-/low)/CD44(+) cell populations are enriched for genes involved in transforming growth factor-beta, tumour necrosis factor, and interferon response pathways. Moreover, we found the presence of nuclear factor-kappaB (NF-kappaB) activity in CD24(-/low)/CD44(+) cells, which was previously unrecognised. In addition, NF-kappaB inhibitor dehydroxymethylepoxyquinomicin (DHMEQ) prevented tumourigenesis of CD24(-/low)/CD44(+) cells in vivo. CONCLUSION: Our findings suggest that signalling pathways identified using GSEA help to identify molecular targets and biomarkers for TIC-like cells.


Assuntos
Neoplasias da Mama/patologia , Antígeno CD24/análise , Separação Celular/métodos , Perfilação da Expressão Gênica , Receptores de Hialuronatos/análise , Proteínas de Neoplasias/análise , Células-Tronco Neoplásicas/fisiologia , Animais , Benzamidas/farmacologia , Benzamidas/uso terapêutico , Biomarcadores , Neoplasias da Mama/genética , Cicloexanonas/farmacologia , Cicloexanonas/uso terapêutico , Feminino , Vetores Genéticos/farmacologia , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , NF-kappa B/antagonistas & inibidores , NF-kappa B/fisiologia , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/fisiologia , Células-Tronco Neoplásicas/química , Células-Tronco Neoplásicas/transplante , Análise de Sequência com Séries de Oligonucleotídeos , Transdução de Sinais/fisiologia , Transdução Genética , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Oncogene ; 26(42): 6184-93, 2007 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-17420722

RESUMO

Rituximab (chimeric anti-CD20 monoclonal antibody) is currently being used, alone or in combination with chemotherapy, in the treatment of B-non-Hodgkin's lymphoma (B-NHL). We have reported that rituximab treatment of B-NHL cell lines sensitizes the drug-resistant tumor cells to apoptosis by various chemotherapeutic drugs and chemosensitization was, in large part, owing to the selective inhibition of the anti-apoptotic Bcl-(XL) gene product. The constitutive activation of the Akt pathway in B-NHL results in overexpression and functional activation of Bcl-(xL). Hence, we hypothesized that rituximab-induced inhibition of Bcl-(xL) expression and chemosensitization may result, in part, from its inhibitory activity of the Akt pathway. This hypothesis was tested using the drug-resistant Ramos and Daudi B-NHL cell lines. Time kinetic analysis revealed that treatment with rituximab inhibited phosphorylation of Akt, but not unphosphorylated Akt, and the inhibition was first detected at 6 h post-rituximab treatment. Similar time kinetics revealed rituximab-induced inhibition of p-PDK1, p-Bad, p-IKKalpha/beta and p-Ikappabetaalpha and no inhibition of unphosphorylated proteins. In addition, rituximab treatment resulted in significant increase of Bcl-(xL)-Bad heterodimeric complexes as compared to untreated cells. The role of the Akt pathway in the regulation of resistance was corroborated by the use of the Akt inhibitor, LY294002, and by transfection with siRNA Akt. Treatment of tumor cells with LY294002 or with Akt siRNA, but not control siRNA, resulted in inhibition of Bcl-(xL) expression and sensitization to drug-induced apoptosis. Although rituximab did not inhibit the Akt pathway nor sensitized the rituximab-resistant Ramos RR1 clone, treatment with LY294002 or Akt siRNA sensitized the clone to drug-induced apoptosis. The present findings demonstrate for the first time that rituximab inhibits the constitutively activated Akt pathway in B-NHL cell lines, and this inhibition contributes to sensitization of drug-resistant cells to apoptosis by chemotherapeutic drugs. The findings also identify the Akt pathway as target for therapeutic intervention in the reversal of rituximab and drug-resistant B-NHL.


Assuntos
Anticorpos Monoclonais/fisiologia , Apoptose/imunologia , Resistencia a Medicamentos Antineoplásicos/imunologia , Linfoma de Células B/enzimologia , Linfoma de Células B/imunologia , Inibidores de Fosfoinositídeo-3 Quinase , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Transdução de Sinais/imunologia , Anticorpos Monoclonais Murinos , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Humanos , Linfoma de Células B/tratamento farmacológico , Fosfatidilinositol 3-Quinases/fisiologia , Proteínas Proto-Oncogênicas c-akt/fisiologia , Rituximab , Transdução de Sinais/efeitos dos fármacos
9.
J Clin Invest ; 100(7): 1822-30, 1997 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-9312183

RESUMO

4 wk after intraperitoneal inoculation of 0.2 LD50 (50% lethal dose) of murine cytomegalovirus (MCMV) in adult BALB/c mice, MCMV remained detectable in the salivary glands, but not in the lungs or other organs. When the T cells of these mice were activated in vivo by a single injection of anti-CD3 monoclonal antibody, interstitial pneumonitis was induced in the lungs that were free of the virus with an excessive production of the cytokines. In the lungs of such mice persistently infected with MCMV, the mRNA of the cytokines such as IL-2, IL-6, TNF-alpha, and IFN-gamma were abundantly expressed 3 h after the anti-CD3 injection, and the elevated levels continued thereafter. A marked expression of inducible nitric oxide synthetase (iNOS) was then noted in the lungs, suggesting that such cytokines as TNF-alpha and IFN-gamma may have induced iNOS. Although the increase in NO formation was demonstrated by the significant elevation of the serum levels of nitrite and nitrate, the interstitial pneumonitis was not associated with either increased superoxide formation or peroxynitrite-induced tyrosine nitration. Nevertheless, the administration of an NO antagonist also alleviated the interstitial pneumonitis provoked by anti-CD3 mAb. Based on these findings, it was concluded that MCMV-associated pneumonitis is mediated by a molecule of cytokine-induced NO other than peroxynitrite.


Assuntos
Infecções por Citomegalovirus/complicações , Doenças Pulmonares Intersticiais/etiologia , Pulmão/metabolismo , Muromegalovirus , Óxido Nítrico/metabolismo , Animais , Anticorpos Monoclonais/farmacologia , Líquido da Lavagem Broncoalveolar , Complexo CD3/imunologia , Óxidos N-Cíclicos , Citocinas/biossíntese , Infecções por Citomegalovirus/imunologia , Infecções por Citomegalovirus/patologia , Pulmão/patologia , Doenças Pulmonares Intersticiais/induzido quimicamente , Doenças Pulmonares Intersticiais/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , NG-Nitroarginina Metil Éster/farmacologia , Nitratos/sangue , Óxido Nítrico Sintase/biossíntese , Nitritos/sangue , Óxidos de Nitrogênio/farmacologia , Reação em Cadeia da Polimerase , RNA Mensageiro/análise , Espécies Reativas de Oxigênio , Tirosina/análise , Xantina Desidrogenase/análise , Xantina Oxidase/análise
10.
Leukemia ; 20(5): 800-6, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16525497

RESUMO

Chronic lymphocytic leukemia (CLL) is a low-grade lymphoid malignancy incurable with conventional modalities of chemotherapy. Strong and constitutive nuclear factor kappa B (NF-kappaB) activation is a characteristic of CLL cells. We examined the effects of a new NF-kappaB inhibitor, dehydroxymethylepoxyquinomicin (DHMEQ), on CLL cells. Dehydroxymethylepoxyquinomicin completely abrogated constitutive NF-kappaB activity and induced apoptosis of CLL cells. Apoptosis induced by DHMEQ was accompanied by downregulation of NF-kappaB-dependent antiapoptotic genes: c-IAP, Bfl-1, Bcl-X(L) and c-FLIP. Dehydroxymethylepoxyquinomicin also inhibited NF-kappaB induced by CD40 and enhanced fludarabine-mediated apoptosis of CLL cells. Results of this study suggest that inhibition of constitutive and inducible NF-kappaB by DHMEQ in combination with fludarabine is a promising strategy for the treatment of CLL.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Benzamidas/farmacologia , Cicloexanonas/farmacologia , Leucemia Linfocítica Crônica de Células B/tratamento farmacológico , NF-kappa B/antagonistas & inibidores , Vidarabina/análogos & derivados , Idoso , Idoso de 80 Anos ou mais , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD , Antígenos CD40/efeitos dos fármacos , Caspases/efeitos dos fármacos , Caspases/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Regulação para Baixo , Sinergismo Farmacológico , Feminino , Humanos , Proteínas Inibidoras de Apoptose/efeitos dos fármacos , Proteínas Inibidoras de Apoptose/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Leucemia Linfocítica Crônica de Células B/diagnóstico , Leucemia Linfocítica Crônica de Células B/metabolismo , Masculino , Pessoa de Meia-Idade , Antígenos de Histocompatibilidade Menor , NF-kappa B/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Células Tumorais Cultivadas , Vidarabina/farmacologia , Proteína bcl-X/efeitos dos fármacos , Proteína bcl-X/metabolismo
11.
Bone Marrow Transplant ; 52(2): 245-251, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27643869

RESUMO

Recombinant human soluble thrombomodulin (rTM) counteracted capillary leakage and alleviated edema in individuals with sinusoidal obstruction syndrome and engraftment syndrome after hematopoietic stem cell transplantation. We previously showed that rTM increased levels of antiapoptotic protein Mcl-1 and protected endothelial cells from calcineurin inhibitor cyclosporine A (CsA)-induced apoptosis. However, the molecular mechanisms by which rTM enhances barrier function in vascular endothelial cells remain unknown. Here we show that exposure of vascular endothelial EA.hy926 cells to CsA induced phosphorylation of Src/vascular endothelial cadherin (VE-cadherin) and translocation of VE-cadherin from cell surface to cytoplasm, resulting in an increase in vascular permeability. In addition, CsA increased production of inflammatory cytokines, including interleukin (IL)-1ß and IL-6, associated with an increase in nuclear levels of nuclear factor-κB (NF-κB) which also enhanced vascular permeability. Importantly, the fourth and fifth regions of epidermal growth factor-like domain of TM (TME45) attenuated CsA-induced p-Src/VE-cadherin and vascular permeability in parallel with a decrease in nuclear levels of NF-κB and cytokine production in EA.hy926 cells. In conclusion, TM, especially TME45, maintains vascular integrity, at least in part, via Src signaling.


Assuntos
Antígenos CD/metabolismo , Caderinas/metabolismo , Inibidores de Calcineurina/farmacologia , Permeabilidade Capilar/efeitos dos fármacos , Ciclosporina/farmacologia , Trombomodulina/administração & dosagem , Quinases da Família src/metabolismo , Animais , Linhagem Celular , Feminino , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Camundongos , Camundongos Endogâmicos ICR , Proteína de Sequência 1 de Leucemia de Células Mieloides/metabolismo
12.
Biomed Pharmacother ; 60(9): 578-86, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16978829

RESUMO

Previously, we designed and synthesized a new NF-kappaB inhibitor, dehydroxymethylepoxyquinomicin (DHMEQ). In the present research we looked into the effect of DHMEQ on the activation of macrophages, especially on the phagocytotic activity of cells of the mouse macrophage-like cell line RAW264.7. DHMEQ inhibited lipopolysaccharide (LPS)-induced NF-kappaB activation by inhibiting its nuclear translocation from the cytoplasm. It also inhibited the expression of inducible NO synthase (iNOS) and nitric oxide (NO) production induced by LPS and interferon-gamma. Using enzyme-linked immunosorbent assays (ELISAs) we showed DHMEQ to inhibit LPS-induced secretion of IL-6, IL-12, interleukin-1beta (IL-1beta), and TNF-alpha. Furthermore, DHMEQ also inhibited the phagocytosis of fluorescently labeled Escherichia coli by RAW264.7 cells treated with LPS or IL-1beta, thus being the first evidence for the involvement of NF-kappaB in the regulation of phagocytosis by use of this inhibitor. Deletion of p65 by siRNA also inhibited the phagocytosis. DHMEQ inhibited the LPS-induced but not IL-1beta-induced phagocytosis of glass beads, indicating that activation of not only NF-kappaB but also Toll-like receptor 4 (TLR-4) is essential for the phagocytosis of E. coli. Previously we found that DHMEQ inhibited type 2 collagen-induced rheumatoid arthritis and the growth of various human carcinomas in mice. It is thus likely that inhibition of macrophage activation is involved in the mechanism of these anti-inflammatory and antitumor activities of DHMEQ in mice.


Assuntos
Benzamidas/farmacologia , Cicloexanonas/farmacologia , Ativação de Macrófagos/efeitos dos fármacos , NF-kappa B/antagonistas & inibidores , Fagocitose/efeitos dos fármacos , Animais , Células Cultivadas , Citocinas/biossíntese , Lipopolissacarídeos/farmacologia , Camundongos , Óxido Nítrico/biossíntese , Óxido Nítrico Sintase Tipo II/antagonistas & inibidores , Receptor 4 Toll-Like/fisiologia
13.
Cancer Res ; 38(6): 1782-4, 1978 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-348304

RESUMO

An antitumor antibiotic aclacinomycin A, was nonmutagenic in a Salmonella test, but its derivative, N-demethylaclacinomycin A, was mutagenic. Similarly, 1-deoxypyrromycin, a hydrolysis product of aclacinomycin A, was nonmutagenic, but N-demethyl-1-deoxypyrromycin was mutagenic. Daunomycin was highly mutagenic, but N-methyldaunomycin showed only weak mutagenicity, and N-dimethyldaunomycin was nonmutagenic. The aglycones of aclacinomycin A and daunomycin were not mutagenic. Thus, the amino moiety of anthracycline glycosides is concluded to be essential for mutagenesis.


Assuntos
Antibióticos Antineoplásicos/farmacologia , Daunorrubicina/análogos & derivados , Mutagênicos , Naftacenos/farmacologia , Salmonella typhimurium/efeitos dos fármacos , Aclarubicina/análogos & derivados , Daunorrubicina/farmacologia , Glicosídeos/farmacologia , Mutação/efeitos dos fármacos , Relação Estrutura-Atividade
14.
Cancer Res ; 53(20): 4896-9, 1993 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-8402678

RESUMO

Isolated from a culture filtrate of Phellinus sp., cyclophellitol is a specific inhibitor of beta-glucosidase, but unlike castanospermine, it does not inhibit experimental metastasis. However, its structural analogue, 1,6-epi-cyclophellitol, inhibited alpha-glucosidase as well as beta-glucosidase, and inhibited experimental metastasis. 1,6-Epi-cyclophellitol depressed alpha-glucosidase activity in cultured B16/F10 cells after 48 h of incubation. Preincubation of B16/F10 cells for 48 h with 1,6-epi-cyclophellitol inhibited invasion of the cells in a Boyden chamber assay at the doses effective in inhibiting alpha-glucosidase in situ. Pulmonary metastasis of B16/F10 cells in mice was inhibited by pretreatment of the cells with 1,6-epi-cyclophellitol in culture. The inhibitor reduced the collagen type I- and IV-mediated attachment of the cells, whereas it had no effect on laminin-mediated attachment. These results suggest that alpha-glucosidase in tumor cells is essential for the metastatic process through the cellular interaction with collagen type I and IV.


Assuntos
Antineoplásicos/toxicidade , Cicloexanóis/toxicidade , Inibidores de Glicosídeo Hidrolases , Neoplasias Pulmonares/prevenção & controle , Neoplasias Pulmonares/secundário , Melanoma Experimental/patologia , Metástase Neoplásica/prevenção & controle , Animais , Antineoplásicos/uso terapêutico , Adesão Celular/efeitos dos fármacos , Linhagem Celular , Cicloexanóis/uso terapêutico , Feminino , Cinética , Neoplasias Pulmonares/patologia , Melanoma Experimental/tratamento farmacológico , Melanoma Experimental/enzimologia , Camundongos , Camundongos Endogâmicos C57BL , Invasividade Neoplásica , alfa-Glucosidases/metabolismo , beta-Glucosidase/antagonistas & inibidores , beta-Glucosidase/metabolismo
15.
Cancer Res ; 49(4): 996-1000, 1989 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-2643467

RESUMO

Oxanosine, a guanosine analogue antibiotic, altered the "transformed" morphology of rat kidney cells integrating a temperature-sensitive K-ras gene into "normal" morphology at 34 degrees C, a permissive temperature. Oxanosine also increased the cellular content of fibronectin to the normal level. The cells under these conditions had lower levels of guanine nucleotides and unstable and less palmitylated Mr 21,000 protein (K-ras gene product, referred to as p21). These changes in p21 were the same as those which occurred to p21 in the cells cultured in the absence of oxanosine at 39.5 degrees C, a nonpermissive temperature.


Assuntos
Antibióticos Antineoplásicos/farmacologia , Transformação Celular Neoplásica/efeitos dos fármacos , Genes ras , Guanosina/análogos & derivados , Animais , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Rim , Nucleotídeos/metabolismo , Fenótipo , Ratos , Ribonucleosídeos/farmacologia
16.
Cancer Res ; 56(21): 4978-82, 1996 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-8895753

RESUMO

Tyrosine kinase inhibitor, erbstatin, induced morphological apoptosis and DNA fragmentation in human small cell lung carcinoma (SCLC) cells. Erbstatin-induced apoptosis was inhibited by antioxidants, whereas erbstatin-inhibited tyrosine phosphorylation was not affected by them. Erbstatin was shown by means of flow cytometry to induce hydrogen peroxide generation. Furthermore, hydrogen peroxide induced morphological apoptosis and DNA fragmentation in the SCLC cells. We also demonstrated that erbstatin-induced hydrogen peroxide production and DNA fragmentation were partially suppressed by inhibition of protein synthesis. Thus, erbstatin-induced apoptosis would be due to hydrogen peroxide generation via newly synthesized protein.


Assuntos
Apoptose/efeitos dos fármacos , Carcinoma de Células Pequenas/patologia , Inibidores Enzimáticos/farmacologia , Peróxido de Hidrogênio/toxicidade , Hidroquinonas/farmacologia , Neoplasias Pulmonares/patologia , Proteínas Tirosina Quinases/antagonistas & inibidores , Antioxidantes/farmacologia , Carcinoma de Células Pequenas/metabolismo , Cicloeximida/farmacologia , Humanos , Peróxido de Hidrogênio/metabolismo , Neoplasias Pulmonares/metabolismo , Células Tumorais Cultivadas
17.
Cancer Res ; 40(7): 2539-42, 1980 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-7388809

RESUMO

The inhibitory effects of protease inhibitors on blood-borne metastasis in male Donryu rat lung were studied. Injection i.v. of 10(6) Yoshida ascites hepatoma AH7974 cells induced about 118 +/- 92 (S.D.) metastatic foci in rat lung after 3 weeks. Leupeptin (50 mg/kg body weight twice a day), injected i.p. from 2 days before to 4 days after the inoculation of tumor cells, reduced the number of metastatic foci to about 49 +/- 45 (p less than 0.005). Leupeptin also suppressed the formation of metastatic foci of Yoshida ascites hepatoma AH100B cells (p less than 0.001). Elastatinal (100 mg/kg body weight twice a day) and chymostatin (100 mg/kg body weight once a day) did not inhibit formation of metastatic foci of AH7974 cells. Injection i.v. of 10(6) AH7974 cells induced pulmonary thrombi within 1 hr. Leupeptin (50 mg/kg body weight twice a day) reduced the number of thrombi from 1298 +/- 395 to 646 +/- 218, when injected i.p. for 2 days before the inoculation of the cells (p less than 0.005). Chymostatin and elastatinal did not significantly change the number of pulmonary thrombi. These results indicate that leupeptin inhibited metastasis formation and suggest that this effect may be due to the inhibition of thrombus formation after the arrest of circulating tumor cells.


Assuntos
Neoplasias Pulmonares/secundário , Células Neoplásicas Circulantes , Inibidores de Proteases/farmacologia , Animais , Leupeptinas/farmacologia , Neoplasias Hepáticas Experimentais/patologia , Masculino , Metástase Neoplásica , Transplante de Neoplasias , Embolia Pulmonar/prevenção & controle , Ratos
18.
J Neurosci ; 20(22): 8401-9, 2000 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-11069947

RESUMO

The epsilon4 genotype of apolipoprotein E (apoE4) is the most established predisposing factor in Alzheimer's disease (AD); however, it remains unclear how apoE4 contributes to the pathophysiology. Here, we report that the apoE4 protein (ApoE4) evokes apoptosis in neuronal cells through the low-density lipoprotein receptor-related protein (LRP) and heterotrimeric GTPases. We examined neuron/neuroblastoma hybrid F11 cells and found that these cells were killed by 30 microg/ml ApoE4, but not by 30 microg/ml ApoE3. ApoE4-induced death occurred with typical features for apoptosis in time- and dose-dependent manners, and was observed in SH-SY5Y neuroblastomas, but not in glioblastomas or non-neuronal Chinese hamster ovary cells. Activated, but not native, alpha2-macroglobulin suppressed this ApoE4 toxicity. Suppression by the antisense oligonucleotide to LRP and inhibition by low nanomolar concentrations of LRP-associated protein RAP provided evidence for the involvement of LRP. The involvement of heterotrimeric GTPases was demonstrated by the findings that (1) ApoE4-induced death was suppressed by pertussis toxin (PTX), but not by heat-inactivated PTX; and (2) transfection with PTX-resistant mutant cDNAs of Galpha(i) restored the toxicity of ApoE4 restricted by PTX. We thus conclude that one of the neurotoxic mechanisms triggered by ApoE4 is to activate a cell type-specific apoptogenic program involving LRP and the G(i) class of GTPases and that the apoE4 gene may play a direct role in the pathogenesis of AD and other forms of dementia.


Assuntos
Apolipoproteínas E/metabolismo , Apoptose , Proteínas Heterotriméricas de Ligação ao GTP/metabolismo , Neurônios/metabolismo , Receptores Imunológicos/metabolismo , Animais , Apolipoproteína E4 , Apolipoproteínas E/antagonistas & inibidores , Apolipoproteínas E/farmacologia , Apoptose/efeitos dos fármacos , Células CHO , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Cricetinae , Relação Dose-Resposta a Droga , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/antagonistas & inibidores , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/genética , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Glioblastoma , Complexo Antigênico da Nefrite de Heymann , Humanos , Células Híbridas/citologia , Células Híbridas/efeitos dos fármacos , Células Híbridas/metabolismo , Marcação In Situ das Extremidades Cortadas , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/farmacologia , Camundongos , Neuroblastoma , Neurônios/citologia , Neurônios/efeitos dos fármacos , Oligonucleotídeos Antissenso/farmacologia , Toxina Pertussis , Ratos , Receptores Imunológicos/antagonistas & inibidores , Receptores de LDL/metabolismo , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Transfecção , Fatores de Virulência de Bordetella/farmacologia , alfa-Macroglobulinas/metabolismo , alfa-Macroglobulinas/farmacologia
19.
Biochim Biophys Acta ; 1166(2-3): 188-92, 1993 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-8443236

RESUMO

Tyrosine kinase inhibitors such as erbstatin and lavendustin derivative inhibited platelet-derived growth factor (PDGF)- and bombesin-induced inositol phosphate formation and phospholipase C (PLC) activation in quiescent NIH3T3 cells. However, bombesin-induced PLC activation was only partially inhibited by tyrosine kinase inhibitors, whereas PDGF-induced activation was completely. Moreover, although bombesin-induced PLC activation was partially inhibited by pertussis toxin alone, this toxin inhibited almost completely in the presence of tyrosine kinase inhibitors. Thus, tyrosine kinase was suggested to be involved in PDGF- and bombesin-induced PLC activation in a different manner.


Assuntos
Bombesina/farmacologia , Hidroquinonas/farmacologia , Fenóis/farmacologia , Fator de Crescimento Derivado de Plaquetas/farmacologia , Proteínas Tirosina Quinases/metabolismo , Fosfolipases Tipo C/metabolismo , Células 3T3/efeitos dos fármacos , Células 3T3/metabolismo , Animais , Bombesina/antagonistas & inibidores , Células Cultivadas , Interações Medicamentosas , Ativação Enzimática , Fosfatos de Inositol/biossíntese , Camundongos , Toxina Pertussis , Fator de Crescimento Derivado de Plaquetas/antagonistas & inibidores , Proteínas Tirosina Quinases/antagonistas & inibidores , Fatores de Virulência de Bordetella/farmacologia
20.
Biochim Biophys Acta ; 1179(2): 213-23, 1993 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-8218364

RESUMO

Human neutrophils maximally stimulated with the optimal concentration (100 ng/ml) of phorbol myristate acetate (PMA), a direct activator of protein kinase C (PKC), for 5 min at 37 degrees C did not respond with superoxide (O2-) release to the later addition of PMA itself or the Ca2+ ionophore ionomycin. However, these cells did respond with enhanced release of O2- to the later addition of N-formyl-methionyl-leucyl-phenylalanine (FMLP) or concanavalin A (Con A). In these PMA-pretreated cells, an increase in cytoplasmic free Ca2+ ([Ca2+]i) induced by ionomycin was unaffected, whereas that induced by FMLP was inhibited by 50-60% and that induced by Con A was completely abolished. A 42-kDa protein was predominantly and consistently tyrosine-phosphorylated by FMLP, PMA and ionomycin with the different kinetics according to the stimuli. The dose-response curves showed that tyrosine phosphorylation and O2- release were stimulated in parallel by PMA, whereas tyrosine phosphorylation and an increase in [Ca2+]i, but not O2- release, were stimulated in parallel by FMLP or ionomycin. The potency of inducing tyrosine phosphorylation was ionomycin > FMLP = PMA, whereas the potency of triggering of O2- release was PMA > ionomycin = FMLP. UCN-01, a PKC inhibitor, inhibited O2- release and tyrosine phosphorylation induced by PMA, but not by FMLP or ionomycin. In contrast, pertussis toxin inhibited O2- release and tyrosine phosphorylation induced by FMLP, but not by PMA. Tyrosine kinase inhibitors (erbstatin and genistein) inhibited O2- release induced by FMLP, but not by PMA. However, both tyrosine kinase inhibitors did not impair FMLP- or PMA-induced tyrosine phosphorylation of a 42-kDa protein. Increased tyrosine phosphorylation of a 42-kDa protein was also detected in immature myeloid cells (HL-60 cells) stimulated by PMA, but not by ionomycin. These findings suggest that FMLP and Con A trigger the respiratory burst in human neutrophils by activating the definite pathway which include other signals than activation of PKC and an increase in [Ca2+]i; tyrosine phosphorylation of a 42-kDa protein is induced by the PKC-dependent and independent mechanisms according to the stimuli, and the PKC-independent and ionomycin-sensitive mechanism is inoperative in HL-60 cells; and tyrosine phosphorylation of a 42-kDa protein is unlikely to be causally related to activation of the respiratory burst.


Assuntos
Neutrófilos/metabolismo , Proteína Quinase C/metabolismo , Explosão Respiratória , Tirosina/metabolismo , Concanavalina A/farmacologia , Genisteína , Humanos , Hidroquinonas/farmacologia , Ionomicina/farmacologia , Isoflavonas/farmacologia , N-Formilmetionina Leucil-Fenilalanina/farmacologia , Neutrófilos/efeitos dos fármacos , Toxina Pertussis , Fosforilação , Transdução de Sinais , Superóxidos/metabolismo , Acetato de Tetradecanoilforbol/farmacologia , Fatores de Virulência de Bordetella/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA