Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
1.
Mediators Inflamm ; 2019: 7651470, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31182933

RESUMO

Lumbar spinal stenosis (LSS) is a major cause of chronic low back pain; however, only a few therapies which have been used in clinics still have limited effects on functional recovery. SHINBARO2 is a refined traditional formulation for inflamed lesions and relieve pain of muscular skeletal disease. This study aimed at investigating the effects of SHINBARO2 on LSS and at determining its underlying molecular mechanism in rat models. The LSS rat models were set up by surgical operations in 6-week-old male Sprague-Dawley rats. SHINBARO2 was orally or intraperitoneally administered for 14 days. The motor and sensory ability of rats were evaluated using the activity cage and hot plate method. On the termination day, total vertebrae including the disc and spinal cord were excised for ex vivo study. SHINBARO2 improved locomotor functions and pain sensitivity in LSS rat models. Mechanism study suggested that SHINBARO2 inhibited the production of nitric oxide and prostaglandin E2 in tissues from LSS-induced rats. SHINBARO2 also suppressed the expression of proinflammatory cytokines including tumor necrosis factor-α and interleukin-1ß. The activation of NF-κB by LSS surgery was effectively reduced by SHINBARO2, which coincided with the inhibition of IκB degradation. In addition, brain-derived neurotrophic factor (BDNF), a potent promoter of neurite growth, and its downstream ERK signaling were also regulated by SHINBARO2. These findings suggest that the effect of SHINBARO2 might be associated in part with the anti-inflammation and pain control in LSS rat models.


Assuntos
Anti-Inflamatórios/uso terapêutico , Estenose Espinal/tratamento farmacológico , Animais , Anti-Inflamatórios/química , Western Blotting , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Imuno-Histoquímica , Interleucina-1beta/metabolismo , Locomoção/fisiologia , Masculino , NF-kappa B/metabolismo , Ratos , Ratos Sprague-Dawley , Estenose Espinal/imunologia , Estenose Espinal/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
2.
Mediators Inflamm ; 2018: 4514329, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29849490

RESUMO

Shinbaro3, a formulation derived from the hydrolysed roots of Harpagophytum procumbens var. sublobatum (Engl.) Stapf, has been clinically used in the pharamacopuncture treatment of arthritis in Korea. In the present study, Shinbaro3 inhibited NO generation in LPS-induced RAW 264.7 cells in a dose-dependent manner. Shinbaro3 also downregulated the mRNA and protein expression of inflammatory mediators in a dose-dependent manner. Three mechanisms explaining the effects of Shinbaro3 in RAW 264.7 cells were identified as follows: (1) inhibition of the extracellular signal-regulated kinase 1 and 2 (ERK1/2), stress-activated protein kinase (SAPK)/c-Jun N-terminal protein kinase (JNK), and p38 mitogen-activated protein kinase (MAPK) pathways; (2) suppression of IκB kinase-α/ß (IKK-α/ß) phosphorylation and nuclear factor-kappa B (NF-κB) subunits in the NF-κB pathway, which are involved in MyD88-dependent signalling; and (3) downregulation of IFN-ß mRNA expression via inhibition of interferon regulatory factor 3 (IRF3) and Janus-activated kinase 1 (JAK1)/signal transducer and activator of transcription 1 (STAT1) phosphorylation, which is involved in TRIF-dependent signalling. Shinbaro3 exerted anti-inflammatory effects in LPS-stimulated RAW 264.7 macrophage cells through modulation of the TLR4/MyD88 pathways, suggesting that Shinbaro3 is a novel anti-inflammatory therapeutic candidate in the field of pharmacopuncture.


Assuntos
Anti-Inflamatórios/química , Anti-Inflamatórios/farmacologia , Harpagophytum/química , Lipopolissacarídeos/toxicidade , Extratos Vegetais/farmacologia , Receptor 4 Toll-Like/metabolismo , Animais , Fator Regulador 3 de Interferon/metabolismo , Camundongos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , NF-kappa B/metabolismo , Extratos Vegetais/química , Células RAW 264.7 , Fator de Transcrição STAT1/metabolismo , Transdução de Sinais/efeitos dos fármacos
3.
J Nat Prod ; 80(2): 434-442, 2017 02 24.
Artigo em Inglês | MEDLINE | ID: mdl-28106392

RESUMO

Harpagoside (1) is an iridoid glycoside isolated from the radix of Harpagophytum procumbens var. sublobatum, commonly called Devil's claw. The anti-osteoporotic effect of 1 was investigated in both in vitro cell cultures and in vivo using an ovariectomized (OVX) mouse model. Compound 1 induced bone formation by stimulating osteoblast proliferation, alkaline phosphatase activity, and mineralization in osteoblastic MC3T3-E1 cells. Treatment with 1 increased the mRNA and protein expression of bone formation biomarkers through regulation of the BMP2 and Wnt signaling pathway in MC3T3-E1 cells. Compound 1 also suppressed the RANKL-induced osteoclastogenesis of cultured mouse bone marrow cells. Oral administration of 1 restored the OVX-induced destruction of trabecular bone. The bone mineral density of the femur was also increased significantly by 1. The elevated serum levels of osteocalcin, C-terminal telopeptide, and tartrate-resistant acid phosphatase in the OVX mice were decreased by treatment with 1. These findings suggest that compound 1 may protect against bone loss induced by OVX in mice by regulating stimulation of osteoblast differentiation and inhibition of osteoclast resorption. Therefore, harpagoside (1) is a potential candidate for management of postmenopausal osteoporosis.


Assuntos
Glicosídeos/farmacologia , Harpagophytum/química , Osteoblastos/efeitos dos fármacos , Osteoclastos/efeitos dos fármacos , Osteoporose Pós-Menopausa/tratamento farmacológico , Piranos/farmacologia , Via de Sinalização Wnt/fisiologia , Animais , Densidade Óssea/efeitos dos fármacos , Proteína Morfogenética Óssea 2/metabolismo , Reabsorção Óssea/metabolismo , Diferenciação Celular/efeitos dos fármacos , Feminino , Fêmur/efeitos dos fármacos , Glicosídeos/química , Humanos , Camundongos , Estrutura Molecular , Osteoblastos/metabolismo , Osteocalcina/análise , Osteocalcina/sangue , Osteoclastos/metabolismo , Osteogênese/efeitos dos fármacos , Piranos/química , Ligante RANK/metabolismo , Ligante RANK/farmacologia , República da Coreia , Ativação Transcricional , Regulação para Cima
4.
Invest New Drugs ; 34(1): 1-14, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26581399

RESUMO

Arsenic compounds have been used in traditional medicine for several centuries. KML001 (sodium metaarsenite; NaAsO2) is an orally bio-available arsenic compound with potential anti-cancer activity. However, the effect of KML001 has not been studied in lymphoid neoplasms. The aim of this study is to evaluate the anti-proliferative effect of KML001 in non-Hodgkin's lymphoma and to compare its efficacy with As2O3. KML001 inhibited cellular proliferation in all tested lymphoma cell lines as well as JurkatR cells (adriamycin-resistant Jurkat cells) in a dose-dependent manner, while As2O3 was not effective. Cell cycle regulatory protein studies have suggested that KML001 induces G1 arrest via p27-induced inhibition of the kinase activities of CDK2, 4, and 6. Treatment of KML001 induced apoptosis in Jurkat and JurkatR cells. The apoptotic process was associated with down-regulation of Bcl-2 (antiapoptotic molecule), up-regulation of Bax (proapoptotic molecule), and inhibition of caspase-3, -8, and -9. In addition, cell signaling including the STAT, PI3K/Akt, MAPK, and NF-κB signal pathways were inhibited in KML001-treated Jurkat and JurkatR cells. Furthermore, targeting the telomere by KML001 was observed in the Jurkat and JurkatR cells. The In vivo anti-tumoral activity of KML001 was confirmed in a xenograft murine model. Interestingly, partial responses were seen in two lymphoma patients treated with 10 mg/day (follicular lymphoma for 16 weeks and mantle cell lymphoma for 24 weeks) without severe toxicities. These findings suggest that KML001 may be a candidate agent for the treatment of de novo, refractory, and relapsed non-Hodgkin's lymphoma patients.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Arsenitos/farmacologia , Linfoma não Hodgkin/tratamento farmacológico , Compostos de Sódio/farmacologia , Idoso , Animais , Antineoplásicos/administração & dosagem , Trióxido de Arsênio , Arsenicais/farmacologia , Arsenitos/administração & dosagem , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Regulação para Baixo/efeitos dos fármacos , Feminino , Humanos , Células Jurkat , Linfoma não Hodgkin/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Pessoa de Meia-Idade , Óxidos/farmacologia , Projetos Piloto , Transdução de Sinais/efeitos dos fármacos , Compostos de Sódio/administração & dosagem , Regulação para Cima/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
5.
J Nat Prod ; 79(4): 1097-104, 2016 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-27064730

RESUMO

The antitumor activity of spicatoside A (1), a steroidal saponin isolated from the tuber of Liriope platyphylla, and its underlying mechanisms were investigated in HCT116 human colorectal cancer cells. Compound 1 induced autophagy and apoptotic cell death and inhibited tumor growth in a nude mouse xenograft model implanted with HCT116 cells. Treatment with 1 for 24 h enhanced the formation of acidic vesicular organelles in the cytoplasm, indicating the induction of the onset of autophagy. This event was associated with the regulation of autophagic markers including microtubule-associated protein 1 light chain 3 (LC3)-II, p62, beclin 1, lysosomal-associated membrane protein 1 (LAMP 1), and cathepsin D by inhibiting the PI3K/Akt/mTOR signaling pathway, regulating mitogen-activated protein kinase (MAPK) signaling, and increasing p53 levels. However, a prolonged exposure to 1 resulted in apoptosis characterized by the accumulation of a sub-G1 cell population and an annexin V/propidium iodide (PI)-positive cell population. Apoptosis induced by 1 was associated with the regulation of apoptotic proteins including Bcl-2, Bax, and Bid, the release of cytochrome c into the cytosol, and the accumulation of cleaved poly-ADP-ribose polymerase (PARP). Further study revealed that cleavage of beclin 1 by caspases plays a critical role in the 1-mediated switch from autophagy to apoptosis. Taken together, these findings highlight the significance of 1 in the modulation of crosstalk between autophagy and apoptosis, as well as the potential use of 1 as a novel candidate in the treatment of human colorectal cancer cells.


Assuntos
Autofagia/efeitos dos fármacos , Liliaceae/química , Saponinas/farmacologia , Animais , Apoptose/efeitos dos fármacos , Proteínas Reguladoras de Apoptose , Proteína Beclina-1 , Proliferação de Células/efeitos dos fármacos , Neoplasias Colorretais , Células HCT116 , Humanos , Proteínas de Membrana , Camundongos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Estrutura Molecular , Fosfatidilinositol 3-Quinases/metabolismo , Saponinas/química , Saponinas/isolamento & purificação , Transdução de Sinais/efeitos dos fármacos
7.
J Nat Prod ; 77(4): 917-24, 2014 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-24689881

RESUMO

The anti-inflammatory activity of handelin (1), a guaianolide dimer from Chrysanthemum boreale flowers, was evaluated in vivo, and the effects on mediators nitric oxide (NO), prostaglandin E2 (PGE2), tumor necrosis factor-α (TNF-α), and interleukin-1ß (IL-1ß) and the nuclear factor-κB (NF-κB) and ERK/JNK signaling pathways were investigated in vitro. Compound 1 inhibited lipopolysaccharide (LPS)-induced production of NO and PGE2 in cultured mouse macrophage RAW 264.7 cells. The suppression of NO and PGE2 production by 1 was correlated with the downregulation of mRNA and protein expression of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2). Compound 1 also suppressed the induction of pro-inflammatory cytokines TNF-α and IL-1ß in LPS-stimulated RAW 264.7 cells. To further clarify the transcriptional regulatory pathway in the expression of iNOS and COX-2 by 1, the role of NF-κB was determined in RAW 264.7 cells. Compound 1 inhibits the binding activity of NF-κB into the nuclear proteins. The transcriptional activity of NF-κB stimulated with LPS was also suppressed by 1, which coincided with the inhibition of IκB degradation. Compound 1 also suppressed the activation of mitogen-activated protein kinases, including ERK and JNK signaling. In addition, the LPS-stimulated upregulation of miRNA-155 expression was suppressed by 1. The oral administration of 1 inhibited acute inflammation in carrageenan-induced paw and 12-O-tetradecanoylphorbol 13-acetate (TPA)-induced ear edema models. The serum level of IL-1ß was also inhibited by 1 in a carrageenan-induced paw edema model. These findings suggest that the suppression of NF-κB activation and pro-inflammatory cytokine production may be a plausible mechanism of action for the anti-inflammatory activity of handelin.


Assuntos
Anti-Inflamatórios/farmacologia , Chrysanthemum/química , Citocinas/metabolismo , Proteínas I-kappa B/metabolismo , NF-kappa B/efeitos dos fármacos , Terpenos/farmacologia , Animais , Anti-Inflamatórios/química , Ciclo-Oxigenase 2 , Dinoprostona/metabolismo , Regulação para Baixo , Edema/induzido quimicamente , Edema/tratamento farmacológico , Proteínas I-kappa B/efeitos dos fármacos , Interleucina-1beta/efeitos dos fármacos , Lipopolissacarídeos/farmacologia , Macrófagos/efeitos dos fármacos , Camundongos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Modelos Animais , Estrutura Molecular , Inibidor de NF-kappaB alfa , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo II/efeitos dos fármacos , Óxido Nítrico Sintase Tipo II/metabolismo , Fitoterapia , Transdução de Sinais/efeitos dos fármacos , Terpenos/química , Fator de Necrose Tumoral alfa/efeitos dos fármacos
8.
BMC Complement Altern Med ; 14: 184, 2014 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-24903150

RESUMO

BACKGROUND: JSOG-6 is used as a traditional medicine to relieve the symptoms associated with inflammation, rheumatism, and osteoporosis in Korea. In the present study, we investigated the effects of JSOG-6 on bone loss prevention both in in vitro and in vivo as well as its underlying mechanism of action. METHODS: Protection against bone loss was assessed in an ovariectomized (OVX) mouse model. Bone microarchitecture was measured using a micro-computed tomography to detect the parameters of three-dimensional structure of a trabecular bone. Serum biomarkers were also evaluated in an OVX-induced model. Osteoclasts derived from mouse bone marrow cells (BMCs) and osteoblastic MC3T3-E1 cells were also employed to investigate the mechanism of action. RESULTS: Oral administration of JSOG-6 significantly increased the bone mineral density (BMD) of the femur in OVX mice in vivo. Especially, the reduced Tb.No (trabecular bone number) in the OVX group was significantly recovered by JSOG-6 treatment. The serum levels of alkaline phosphatase (ALP), osteocalcin, C-terminal telopeptide, and tartrate-resistant acid phosphatase, biomarkers of bone resorption, were significantly elevated in OVX mice, but JSOG-6 effectively inhibited the increase in OVX mice. JSOG-6 was also found to enhance the osteoblastic differentiation and maturation with the increase of the density and ALP activity, a marker of osteoblastic differentiation, as well as calcium deposition, a marker of osteoblastic maturation in MC3T3-E1 cells. The effects of JSOG-6 on osteoblastic differentiation were also associated in part with the increase of ALP and OPN mRNA expressions and the decrease of RANKL mRNA expression in MC3T3-E1 cells. CONCLUSIONS: The findings demonstrate that JSOG-6 induced protection against bone loss in OVX mice, and its anti-osteoporotic property might be, in part, a function of the stimulation of osteoblast differentiation and the inhibition of osteoclast formation. These findings suggest that JSOG-6 might be an applicable therapeutic traditional medicine for the regulation of the osteoporotic response.


Assuntos
Reabsorção Óssea/prevenção & controle , Osteoblastos/efeitos dos fármacos , Osteoclastos/efeitos dos fármacos , Osteoporose/tratamento farmacológico , Extratos Vegetais/uso terapêutico , Fosfatase Ácida , Fosfatase Alcalina/sangue , Animais , Densidade Óssea/efeitos dos fármacos , Osso e Ossos/metabolismo , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Feminino , Fêmur , Expressão Gênica/efeitos dos fármacos , Humanos , Isoenzimas , Camundongos , Camundongos Endogâmicos ICR , Osteoblastos/metabolismo , Osteocalcina/sangue , Osteogênese/efeitos dos fármacos , Osteoporose/sangue , Ovariectomia , Fitoterapia , Extratos Vegetais/farmacologia , Ligante RANK/metabolismo , República da Coreia , Fosfatase Ácida Resistente a Tartarato , Microtomografia por Raio-X
9.
Chem Biodivers ; 11(5): 749-59, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24827684

RESUMO

Anemarrhena asphodeloides is widely used in traditional Chinese medicine, and is known to possess antidiabetic and anti-inflammatory properties. Because inducible nitric oxide synthase (iNOS) plays an important role in inflammation, we investigated the inhibitory effects of two known phenolic compounds, nyasol (1) and broussonin A (2), from A. asphodeloides, on iNOS and its plausible mechanism of action. Compounds 1 and 2 exhibited inhibitory effects on nitric oxide (NO) production in lipopolysaccharide (LPS)-stimulated RAW 264.7 macrophage cells. Compounds 1 and 2 also suppressed the expressions of iNOS protein and mRNA. Moreover, compounds 1 and 2 suppressed the expression of inflammatory cytokines such as interleukin-1ß (IL-1ß) and interferon-ß (IFN-ß). They also inhibited the transcriptional activity of NF-κB and degradation of IκB-α, as well as the activation of Akt and ERK in LPS-stimulated RAW 264.7 cells. In in vivo animal model, compounds 1 and 2 significantly inhibited TPA-induced mouse ear edema. These results suggest that 1 and 2 suppress LPS-stimulated iNOS expression at the transcriptional level through modulating NF-κB and down-regulation of the Akt and ERK signaling pathways. Taken together, these findings indicate that the suppressive effects of 1 and 2 on iNOS expression might provide one possible mechanism for their anti-inflammatory activities.


Assuntos
Anemarrhena/química , Anti-Inflamatórios/farmacologia , Proteínas I-kappa B/genética , Lignanas/farmacologia , Macrófagos/efeitos dos fármacos , Óxido Nítrico Sintase Tipo II/genética , Fenóis/farmacologia , Animais , Anti-Inflamatórios/isolamento & purificação , Anti-Inflamatórios/uso terapêutico , Linhagem Celular , Regulação para Baixo/efeitos dos fármacos , Edema/induzido quimicamente , Edema/tratamento farmacológico , Proteínas I-kappa B/imunologia , Lignanas/isolamento & purificação , Lignanas/uso terapêutico , Lipopolissacarídeos/imunologia , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Inibidor de NF-kappaB alfa , Óxido Nítrico/imunologia , Óxido Nítrico Sintase Tipo II/imunologia , Fenóis/isolamento & purificação , Fenóis/uso terapêutico , Ativação Transcricional/efeitos dos fármacos
10.
Bioorg Med Chem ; 21(4): 1006-17, 2013 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-23294831

RESUMO

New water soluble antofine C-13a analogues were designed, synthesized, and evaluated for antiproliferative activity against cancer cells. Particularly, (-)-(R)-13a-hydroxymethylantofine ((-)-(R)-4b) demonstrated notable growth inhibition against a panel of human cancer cell lines. This growth inhibition was associated with the arrest of the cell cycle in the G0/G1 phases and suppression of mTOR signaling in human lung A549 cancer cells. Compound (-)-(R)-4b also overcame paclitaxel-resistance in human lung cancer cells (A549-Pa) by suppressing P-glycoprotein expression. Furthermore, compound (-)-(R)-4b significantly inhibited the tumor growth of A549 and A549-Pa xenografts in a nude mouse model, which suggests it is a promising novel antitumor agent with sufficient aqueous solubility.


Assuntos
Antineoplásicos/síntese química , Desenho de Fármacos , Indóis/química , Fenantrolinas/química , Animais , Antineoplásicos/uso terapêutico , Antineoplásicos/toxicidade , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Pontos de Checagem da Fase G1 do Ciclo Celular/efeitos dos fármacos , Células HCT116 , Células HEK293 , Humanos , Indóis/uso terapêutico , Indóis/toxicidade , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Nus , Fenantrolinas/uso terapêutico , Fenantrolinas/toxicidade , Transdução de Sinais/efeitos dos fármacos , Estereoisomerismo , Serina-Treonina Quinases TOR/antagonistas & inibidores , Serina-Treonina Quinases TOR/metabolismo , Transplante Heterólogo , Água/química
11.
J Nat Prod ; 76(7): 1278-84, 2013 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-23855266

RESUMO

The antiproliferative and antitumor activities of 2-hydroxycinnamaldehyde (1), a phenylpropanoid isolated from the bark of Cinnamomum cassia, were investigated using human colorectal cancer cells. Compound 1 exhibited antiproliferative effects in HCT116 colon cancer cells, accompanied by modulation of the Wnt/ß-catenin cell signaling pathway. This substance was found also to inhibit ß-catenin/T-cell factor (TCF) transcriptional activity in HEK293 cells and HCT116 colon cancer cells. Further mechanistic investigations in human colon cancer cells with aberrantly activated Wnt/ß-catenin signaling showed that 1 significantly suppressed the binding of ß-catenin/TCF complexes to their specific genomic targets in the nucleus and led to the down-regulation of Wnt target genes such as c-myc and cyclin D1. In an in vivo xenograft model, the intraperitoneal administration of 1 (10 or 20 mg/kg body weight, three times/week) for four weeks suppressed tumor growth in athymic nude mice implanted with HCT116 colon cancer cells significantly, without any apparent toxicity. In an ex vivo biochemical analysis of the tumors, compound 1 was also found to suppress Wnt target genes associated with tumor growth including ß-catenin, c-myc, cyclin D1, and survivin. The suppression of the Wnt/ß-catenin signaling pathway is a plausible mechanism of action underlying the antiproliferative and antitumor activity of 1 in human colorectal cancer cells.


Assuntos
Antineoplásicos/farmacologia , Cinamatos/farmacologia , Animais , Antineoplásicos/química , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Cinamatos/química , Cinnamomum aromaticum/química , Neoplasias do Colo/genética , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Ciclina D1/metabolismo , Células HCT116 , Humanos , Camundongos , Estrutura Molecular , Casca de Planta/química , Transdução de Sinais/efeitos dos fármacos , Fatores de Transcrição TCF/metabolismo , Via de Sinalização Wnt/efeitos dos fármacos , beta Catenina/genética , beta Catenina/fisiologia
12.
Mol Pharmacol ; 82(2): 168-77, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22550094

RESUMO

Abnormal activation of the canonical Wnt/ß-catenin pathway and up-regulation of the ß-catenin/T-cell factor (TCF) response to transcriptional signaling play a critical role early in colorectal carcinogenesis. Therefore, Wnt/ß-catenin signaling is considered an attractive target for cancer chemotherapeutic or chemopreventive agents. Small molecules derived from the natural products were used in our cell-based reporter gene assay to identify potential inhibitors of Wnt/ß-catenin signaling. Magnolol, a neolignan from the cortex of Magnolia obovata, was identified as a promising candidate because it effectively inhibited ß-catenin/TCF reporter gene (TOPflash) activity. Magnolol also suppressed Wnt3a-induced ß-catenin translocation and subsequent target gene expression in human embryonic kidney 293 cells. To further investigate the precise mechanisms of action in the regulation of Wnt/ß-catenin signaling by magnolol, we performed Western blot analysis, real-time reverse transcriptase-polymerase chain reactions, and an electrophoretic mobility shift assay in human colon cancer cells with aberrantly activated Wnt/ß-catenin signaling. Magnolol inhibited the nuclear translocation of ß-catenin and significantly suppressed the binding of ß-catenin/TCF complexes onto their specific DNA-binding sites in the nucleus. These events led to the down-regulation of ß-catenin/TCF-targeted downstream genes such as c-myc, matrix metalloproteinase-7, and urokinase-type plasminogen activator in SW480 and HCT116 human colon cancer cells. In addition, magnolol inhibited the invasion and motility of tumor cells and exhibited antitumor activity in a xenograft nude mouse model bearing HCT116 cells. These findings suggest that the growth inhibition of magnolol against human colon cancer cells can be partly attributed to the regulation of the Wnt/ß-catenin signaling pathway.


Assuntos
Antineoplásicos Fitogênicos/uso terapêutico , Compostos de Bifenilo/uso terapêutico , Neoplasias Colorretais , Lignanas/uso terapêutico , Transdução de Sinais/fisiologia , Proteína Wnt3A/fisiologia , beta Catenina/fisiologia , Animais , Antineoplásicos Fitogênicos/farmacologia , Compostos de Bifenilo/farmacologia , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/patologia , Feminino , Células HCT116 , Células HEK293 , Humanos , Lignanas/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Transdução de Sinais/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
13.
Bioorg Med Chem ; 20(13): 4109-16, 2012 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-22633122

RESUMO

A chlorinated methyl jasmonate analog (J7) was elaborated as an in vitro anti-inflammatory lead. However, its in vitro efficacy profile was not reproduced in a subsequent in vivo evaluation, presumably due to its rapid enzymatic hydrolysis in a biological system. In an attempt to improve the metabolic stability of the lead J7 by replacement of its labile methyl ester with reasonable ester groups, several analogs resistant to enzymatic hydrolysis were synthesized. In vivo evaluation of the stability-improved analogs showed that these compounds displayed higher efficacy than the lead J7, suggesting that these new jasmonate analogs may serve as potential anti-inflammatory leads.


Assuntos
Anti-Inflamatórios/síntese química , Ciclopentanos/química , Ciclopentanos/síntese química , Ácidos Graxos Insaturados/síntese química , Oxilipinas/química , Animais , Anti-Inflamatórios/química , Anti-Inflamatórios/farmacologia , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Ciclopentanos/farmacologia , Ácidos Graxos Insaturados/química , Ácidos Graxos Insaturados/farmacologia , Camundongos , Óxido Nítrico/metabolismo , Oxilipinas/síntese química , Oxilipinas/farmacologia , Relação Estrutura-Atividade
14.
Cell Physiol Biochem ; 27(3-4): 353-62, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21471724

RESUMO

Since inhibitors of inducible nitric oxide synthase (iNOS) have been considered as potential anti-inflammatory and cancer chemopreventive agents, we have evaluated the inhibitory effects on the production of nitric oxide (NO) in lipopolysaccharide (LPS)-stimulated murine macrophage RAW 264.7 cells with natural and synthetic compounds. Pinosylvin (3,5-dihydroxy-trans-stilbene), a stilbenoid mainly found in heartwood of Pinus sylvestris, exhibited the inhibition of iNOS protein and mRNA expression. The plausible mechanisms of pinosylvin on the suppression of iNOS gene expression were found to be associated with the downregulation of interferon regulatory factor 3 (IRF-3) and interferon-ß (IFN-ß) expression, which are related to Toll/IL-1 receptor domain-containing adapter inducing interferon-ß (TRIF)-mediated signaling. Decreased IFN-ß expression suppressed a phosphorylation of JAK kinase, and subsequently, the phosphorylation of signal transducer and activator of transcription-1, one of the iNOS transcriptional activators, was inhibited by pinosylvin. In addition, the suppression of poly(I:C)-induced iNOS expression, and the attenuation of iNOS expression under the IRF-3 gene knock-down condition also confirmed that pinosylvin affects TRIF pathway. These findings demonstrate that the suppression of LPS-induced iNOS expression by pinosylvin is associated with the regulation of MyD88-independent, but TRIF-mediated signaling pathway.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Fator Regulador 3 de Interferon/metabolismo , Macrófagos/efeitos dos fármacos , Fator 88 de Diferenciação Mieloide/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Estilbenos/farmacologia , Animais , Células Cultivadas , Regulação para Baixo , Fator Regulador 3 de Interferon/antagonistas & inibidores , Fator Regulador 3 de Interferon/genética , Interferon beta/metabolismo , Janus Quinases/metabolismo , Lipopolissacarídeos/farmacologia , Macrófagos/imunologia , Camundongos , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo II/genética , Fosforilação , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Estilbenos/química
15.
J Pharmacol Sci ; 116(1): 89-96, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21512304

RESUMO

NAD(P)H:quinone oxidoreductase 1 (NQO1), a phase II enzyme, plays an important role in the detoxification or chemoprotection of carcinogens, and induction of this enzyme is a target for the prevention of carcinogenesis. Natural stilbenoids have potential cancer chemopreventive activities, potentially through affecting NQO1 activity. Along this line, several stilbenoids were evaluated to procure more potent compounds for inducing NQO1 activity in cultured murine Hepa 1c1c7 cells. As a result, we found that 3,5-dimethoxy-trans-stilbene (DMS) possesses potent NQO1 induction activity through up-regulation of both protein and mRNA expression of NQO1 as determined by Western blot and reverse transcription-polymerase chain reaction analysis, respectively. DMS also increased protein expression of heme oxygenase-1 (HO-1), another phase II enzyme. This induction of NQO1 and HO-1 by DMS was closely related to the regulation of nuclear factor E2-related factor 2 (Nrf2). The translocation and activation of Nrf2 by DMS was also involved in the modulation of the upstream signal transduction molecule, protein kinase C δ. These findings suggest that DMS might have a cancer chemopreventive activity by inducing detoxifying enzymes such as NQO1 and HO-1.


Assuntos
Anticarcinógenos/farmacologia , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/metabolismo , NAD(P)H Desidrogenase (Quinona)/biossíntese , Fator 2 Relacionado a NF-E2/metabolismo , Estilbenos/farmacologia , Animais , Antioxidantes/farmacologia , Carcinoma Hepatocelular/prevenção & controle , Linhagem Celular Tumoral , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Proteínas de Ligação a DNA/metabolismo , Indução Enzimática/efeitos dos fármacos , Genes Reporter/efeitos dos fármacos , Heme Oxigenase-1/genética , Heme Oxigenase-1/metabolismo , Neoplasias Hepáticas/prevenção & controle , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , NAD(P)H Desidrogenase (Quinona)/genética , NAD(P)H Desidrogenase (Quinona)/metabolismo , Regiões Promotoras Genéticas/efeitos dos fármacos , Proteína Quinase C-delta/antagonistas & inibidores , Proteína Quinase C-delta/metabolismo , Transporte Proteico/efeitos dos fármacos , RNA Mensageiro/metabolismo , Elementos de Resposta/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
16.
J Nat Prod ; 74(10): 2102-8, 2011 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-21916433

RESUMO

The growth inhibition and antitumor activities of yuanhuadine (1), a daphnane diterpenoid from the flowers of Daphne genkwa, were investigated in human lung cancer cells. Compound 1 exhibited a relatively selective growth inhibition against human lung cancer cells compared to other solid human cancer cell lines. The potent antiproliferative activity by 1 was associated with cell-cycle arrest and modulation of cell-signaling pathways. Cell-cycle arrest in the G0/G1 and G2/M phase was induced by 1 in A549 human non-small-cell lung cancer cells, and these events were correlated with the expression of checkpoint proteins including the up-regulation of p21 and down-regulation of cyclins, cyclin-dependent kinases 2 (CDK2) and 4 (CDK4), and c-Myc. Compound 1 also suppressed the expression of the Akt/mammalian target of rapamycin (mTOR) and its downstream effector molecules including p70 S6 kinase (p70S6K) and eukaryotic initiation factor 4E-binding protein 1 (4EBP1). Ligand-induced epidermal growth factor receptor (EGFR) and c-Met signaling were also inhibited by 1. The oral administration of 1 (0.5 mg/kg body weight, daily) for 14 days significantly inhibited tumor growth in athymic xenograft nude mouse model bearing human lung A549 cells, without any overt toxicity. Synergistic antiproliferative effects of compound 1 were also found in combination with the EGFR inhibitor gefitinib. Cell-cycle arrest and suppression of Akt/mTOR and EGFR signaling pathways might be plausible mechanisms of actions for the antiproliferative and antitumor activity of 1 in human non-small-cell lung cancer cells.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Daphne/química , Diterpenos/isolamento & purificação , Diterpenos/farmacologia , Receptores ErbB/metabolismo , Terpenos/farmacologia , Animais , Antineoplásicos Fitogênicos/química , Antineoplásicos Fitogênicos/isolamento & purificação , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Ciclo Celular/efeitos dos fármacos , Modelos Animais de Doenças , Diterpenos/química , Regulação para Baixo/efeitos dos fármacos , Receptores ErbB/efeitos dos fármacos , Feminino , Flores/química , Humanos , Camundongos , Camundongos Nus , Estrutura Molecular , Terpenos/química , Terpenos/isolamento & purificação
17.
J Nat Prod ; 74(4): 701-6, 2011 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-21370894

RESUMO

The potential antitumor activity of timosaponin A-III (1), a steroidal saponin from the rhizomes of Anemarrhena asphodeloides, was investigated in human colorectal cancer HCT-15 cells both in cell culture and in an in vivo murine xenograft model. Compound 1 inhibited the proliferation of cancer cells with cell-cycle arrest and induction of apoptosis. Cell-cycle arrest in the G0/G1 and G2/M phase by 1 was correlated with the down-regulation of cyclin A, cyclin B1, cyclin-dependent kinase 2 (CDK2), CDK4, proliferating cell nuclear antigen, and c-Myc. The increase of the sub-G1 peak by 1 was also closely related to the induction of apoptosis, which was evidenced by the induction of DNA fragmentation, activation of caspases, induction of cleaved poly-(ADP ribose) polymerase, and suppression of Bcl-xL and Bcl-2 expression. In an in vivo xenograft model, treatment with 1 (2 or 5 mg/kg body weight, three times/week, ip administration) for four weeks significantly suppressed tumor growth in athymic nude mice bearing HCT-15 cells, without any overt toxicity. Cell-cycle arrest and induction of apoptosis might be plausible mechanisms of actions for the observed antineoplastic activity of 1.


Assuntos
Anemarrhena/química , Antineoplásicos Fitogênicos/química , Antineoplásicos Fitogênicos/farmacologia , Ciclo Celular/efeitos dos fármacos , Neoplasias do Colo/tratamento farmacológico , Quinase 2 Dependente de Ciclina/metabolismo , Saponinas/química , Saponinas/farmacologia , Esteroides/química , Esteroides/farmacologia , Animais , Antineoplásicos Fitogênicos/uso terapêutico , Apoptose/efeitos dos fármacos , Relação Dose-Resposta a Droga , Humanos , Camundongos , Camundongos Nus , Estrutura Molecular , Poli(ADP-Ribose) Polimerases/metabolismo , Rizoma/química , Saponinas/uso terapêutico , Esteroides/uso terapêutico , Ensaios Antitumorais Modelo de Xenoenxerto
18.
Bioorg Med Chem Lett ; 20(17): 5277-81, 2010 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-20667733

RESUMO

In the search for potent water-soluble 3-arylisoquinolines, several 3-arylisoquinolinamines were designed and synthesized. Various substituted 3-arylisoquinolinamines exhibited strong cytotoxic activity against eight different human cancer cell lines. In particular, C-6 or C-7 dimethylamino-substituted 3-arylisoquinolinamines displayed stronger potency than the lead compound 7a. Interestingly, compounds 7b and 7c showed more effective activity against paclitaxel-resistant HCT-15 human colorectal cancer cell lines when compared to the original cytotoxic cancer drug, paclitaxel. We analyzed the cell cycle dynamics by flow cytometry and found that treatment of human HCT-15 cells with 3-arylisoquinolinamine 7b blocked or delayed the progression of cells from G0/G1 phase into S phase, and induced cell death. Treatment with compound 7b also significantly inhibited the growth of tumors and enhanced tumor regression in a paclitaxel-resistant HCT-15 xenograft model.


Assuntos
Antineoplásicos/síntese química , Antineoplásicos/farmacologia , Quinolinas/síntese química , Quinolinas/farmacologia , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Técnicas In Vitro
19.
Nat Prod Res ; 33(24): 3582-3586, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29897257

RESUMO

Radiotherapy is routinely used in the treatment of lung cancer patients. However, it often causes malignant effects, such as promoting cancer cell migration and invasion. Previous studies demonstrated that ionizing radiation (IR) promotes cancer cell invasion by stimulating the ß-catenin, IL-6, STAT3, and Bcl-XL signaling pathway or the PI3K, Akt, and NF-κB signaling pathway. Both Bcl-XL and NF-κB stimulate the secretion of matrix metalloproteases (MMPs), including MMP-2 and MMP-9. In the present study, linarin isolated from Chrysanthemum morifolium flowers significantly decreased the IR-induced cell migration and invasion at a concentration of 5 µM in A549 cells. This effect was mediated via MMP-9 downregulation and the suppression of NF-κB activation by inhibiting NF-κB and IκB-α phosphorylation. However, linarin did not affect the STAT3/Bcl-XL pathway or the stabilization of ß-catenin. Overall, these results suggest that linarin repressed the MMP-9-dependent invasion pathway by regulating NF-κB activity, thereby inhibiting IR-induced cancer metastasis.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Movimento Celular , Regulação para Baixo/efeitos dos fármacos , Glicosídeos/farmacologia , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Induzidas por Radiação/prevenção & controle , Células A549 , Carcinoma Pulmonar de Células não Pequenas/patologia , Movimento Celular/efeitos dos fármacos , Chrysanthemum/química , Glicosídeos/isolamento & purificação , Glicosídeos/uso terapêutico , Humanos , Neoplasias Pulmonares/patologia , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , NF-kappa B/antagonistas & inibidores , NF-kappa B/efeitos dos fármacos , NF-kappa B/metabolismo , Invasividade Neoplásica/prevenção & controle , Fosforilação/efeitos dos fármacos
20.
Mol Ther Oncolytics ; 15: 140-152, 2019 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-31720371

RESUMO

Triple-negative breast cancer (TNBC) is the most intractable cancer in women with a high risk of metastasis. While hyper-methylation of histone H3 catalyzed by disruptor of telomeric silencing 1-like (DOT1L), a specific methyltransferase for histone H3 at lysine residue 79 (H3K79), is reported as a potential target for TNBCs, early developed nucleoside-type DOT1L inhibitors are not sufficient for effective inhibition of growth and metastasis of TNBC cells. We found that TNBC cells had a high expression level of DOT1L and a low expression level of E-cadherin compared to normal breast epithelial cells and non-TNBC cells. Here, a novel psammaplin A analog (PsA-3091) exhibited a potent inhibitory effect of DOT1L-mediated H3K79 methylation. Consistently, PsA-3091 also significantly inhibited the proliferation, migration, and invasion of TNBC cells along with the augmented expression of E-cadherin and the suppression of N-cadherin, ZEB1, and vimentin expression. In an orthotopic mouse model, PsA-3091 effectively inhibited lung metastasis and tumor growth by the regulation of DOT1L activity and EMT biomarkers. Together, we report here a new template of DOT1L inhibitor and suggest that targeting DOT1L-mediated H3K79 methylation by a novel PsA analog may be a promising strategy for the treatment of metastatic breast cancer patients.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA