Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Pharmacol Sci ; 154(2): 97-107, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38246733

RESUMO

In our previous study, we reported that 2, 5-dimethyl-celecoxib (DM-C), a derivative of celecoxib, prevents cardiac remodeling in different mouse models of heart failure, including myocardial infarction (MI). The inflammatory response after MI affects the progression of cardiac remodeling, wherein the immune cells, mainly macrophages, play crucial roles. Therefore, we evaluated the effect of DM-C on macrophages in a cryoinjury-induced myocardial infarction (CMI) mouse model. We observed that DM-C attenuated the deterioration of left ventricular ejection fraction and cardiac fibrosis 14 d after CMI. Gene expression of pro-inflammatory cytokines at the infarct site was reduced by DM-C treatment. Analysis of macrophage surface antigens revealed that DM-C induced transient accumulation of macrophages at the infarct site without affecting their polarization. In vitro experiments using peritoneal monocytes/macrophages revealed that DM-C did not directly increase the phagocytic ability of the macrophages but increased their number, thereby upregulating the clearance capacity. Moreover, DM-C rapidly excluded the cells expressing necrotic cell marker from the infarct site. These results suggested that DM-C enhanced the clearance capacity of macrophages by transiently increasing their number at the infarct site, and terminated the escape from the inflammatory phase earlier, thereby suppressing excessive cardiac remodeling and ameliorating cardiac dysfunction.


Assuntos
Infarto do Miocárdio , Pirazóis , Sulfonamidas , Remodelação Ventricular , Animais , Camundongos , Celecoxib/farmacologia , Celecoxib/uso terapêutico , Volume Sistólico , Função Ventricular Esquerda , Infarto do Miocárdio/tratamento farmacológico , Macrófagos , Modelos Animais de Doenças
2.
Life Sci ; 335: 122278, 2023 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-37981227

RESUMO

AIMS: Differentiation-inducing factor-1 (DIF-1), a compound in Dictyostelium discoideum, exhibits anti-cancer effects by inhibiting cell proliferation and motility of various mammalian cancer cells in vitro and in vivo. In addition, DIF-1 suppresses lung colony formation in a mouse model, thus impeding cancer metastasis. However, the precise mechanism underlying its anti-metastatic effect remains unclear. In the present study, we aim to elucidate this mechanism by investigating the adhesion of circulating tumor cells to blood vessels using in vitro and in vivo systems. MAIN METHODS: Melanoma cells (1.0 × 105 cells) were injected into the tail vein of 8-week-old male C57BL/6 mice after administration of DIF-1 (300 mg/kg per day) and/or lipopolysaccharide (LPS: 2.5 mg/kg per day). To investigate cell adhesion and molecular mechanisms, cell adhesion assay, western blotting, immunofluorescence staining, and flow cytometry were performed. KEY FINDINGS: Intragastric administration of DIF-1 suppressed lung colony formation. DIF-1 also substantially inhibited the adhesion of cancer cells to human umbilical vein endothelial cells. Notably, DIF-1 did not affect the expression level of adhesion-related proteins in cancer cells, but it did decrease the expression of vascular cell adhesion molecule-1 (VCAM-1) in human umbilical vein endothelial cells by suppressing its mRNA-to-protein translation through inhibition of mTORC1-p70 S6 kinase signaling. SIGNIFICANCE: DIF-1 reduced tumor cell adhesion to blood vessels by inhibiting mTORC1-S6K signaling and decreasing the expression of adhesion molecule VCAM-1 on vascular endothelial cells. These findings highlight the potential of DIF-1 as a promising compound for the development of anti-cancer drugs with anti-metastatic properties.


Assuntos
Dictyostelium , Molécula 1 de Adesão de Célula Vascular , Camundongos , Animais , Masculino , Humanos , Molécula 1 de Adesão de Célula Vascular/metabolismo , Lipopolissacarídeos/farmacologia , Dictyostelium/metabolismo , Camundongos Endogâmicos C57BL , Proteínas , Células Endoteliais da Veia Umbilical Humana/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina , Diferenciação Celular , Adesão Celular , Mamíferos/metabolismo
3.
Biochem Pharmacol ; 214: 115663, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37336252

RESUMO

Fibrosis occurs in all organs and tissues except the brain, and its progression leads to dysfunction of affected organs. Fibrosis-induced organ dysfunction results from the loss of elasticity, strength, and functionality of tissues due to the extracellular matrix secreted by myofibroblasts that express smooth muscle-type actin as a marker. Myofibroblasts, which play a major role in fibrosis, were once thought to originate exclusively from activated fibroblasts; however, it is now clear that myofibroblasts are diverse in origin, from epithelial cells, endothelial cells, adipocytes, macrophages, and other cells. Fibrosis of vital organs, such as the heart, lungs, kidneys, and liver, is a serious chronic disease that ultimately leads to death. Currently, anti-cancer drugs have made remarkable progress, as evidenced by the development of many molecular-targeted drugs, and are making a significant contribution to improving the prognosis of cancer treatment. However, the development of anti-fibrotic agents, which also play an important role in prognosis, has lagged. In this review, the current knowledge regarding myofibroblasts is summarized, with particular attention given to their origin and transdifferentiation signaling pathways (e.g., TGF-ß, Wnt/ß-catenin, YAP/TAZ and AMPK signaling pathways). The development of new small molecule anti-fibrotic agents and the repositioning of existing drugs targeting myofibroblast transdifferentiation are discussed.


Assuntos
Transdiferenciação Celular , Miofibroblastos , Humanos , Miofibroblastos/metabolismo , Antifibróticos , Células Endoteliais , Fibroblastos/metabolismo , Fibrose
4.
Hypertens Res ; 45(12): 1869-1881, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36171325

RESUMO

Renin-angiotensin system inhibitors have been shown to prevent cancer metastasis in experimental models, but there are limited data in clinical studies. We aimed to explore whether renin-angiotensin system inhibitors administered during the period of cancer resection can influence the subsequent development of metastasis by analyzing multiple individual types of primary cancers. A total of 4927 patients who had undergone resection of primary cancers at Kyushu University Hospital from 2009 to 2014 were enrolled and categorized into 3 groups based on the use of antihypertensive drugs: renin-angiotensin system inhibitors, other drugs, and none. Cumulative incidence functions of metastasis, treating death as a competing risk, were calculated, and the difference was examined among groups by Gray's test. Fine and Gray's model was employed to evaluate multivariate-adjusted hazards of incidental metastasis. In the multivariate-adjusted analysis, patients with skin and renal cancers showed statistically higher risks of metastasis with the use of renin-angiotensin system inhibitors (hazard ratio [95% confidence interval], 5.81 [1.07-31.57] and 4.24 [1.71-10.53], respectively). Regarding pancreatic cancer, patients treated with antihypertensive drugs other than renin-angiotensin system inhibitors had a significantly increased risk of metastasis (hazard ratio [95% confidence interval], 3.31 [1.43-7.69]). Future larger studies are needed to ascertain whether renin-angiotensin system inhibitors can increase the risk of metastasis in skin and renal cancers, focusing on specific tissue types and potential factors associated with renin-angiotensin system inhibitor use.


Assuntos
Neoplasias Renais , Neoplasias Pancreáticas , Humanos , Anti-Hipertensivos/uso terapêutico , Antagonistas de Receptores de Angiotensina/uso terapêutico , Antagonistas de Receptores de Angiotensina/farmacologia , Inibidores da Enzima Conversora de Angiotensina/farmacologia , Sistema Renina-Angiotensina , Estudos Retrospectivos , Registros Eletrônicos de Saúde , Inibidores Enzimáticos/farmacologia , Neoplasias Pancreáticas/induzido quimicamente , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Renais/tratamento farmacológico
5.
Biochem Pharmacol ; 197: 114950, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35143754

RESUMO

We previously reported that 2,5-dimethylcelecoxib (DM-C), a derivative of celecoxib, lacks cyclooxygenase-2 inhibitory effects and suppresses cardiac remodeling by activating glycogen synthase kinase-3 (GSK-3). However, it remains unclear whether DM-C attenuates fibroblast-to-myofibroblast transformation (FMT), which plays a key role in cardiac fibrosis. Therefore, we evaluated the effect of DM-C on FMT using a cryoinjury-induced myocardial infarction (CMI) mouse model. We found that DM-C attenuated the deterioration of left ventricular ejection fraction after CMI by decreasing cardiac fibrosis. Analysis of the expression level of α-smooth muscle actin (α-SMA), a marker for myofibroblasts, indicated that DM-C decreased FMT at the cardiac injury site. To investigate the mechanism by which DM-C attenuated FMT, fibroblasts obtained from the heart were stimulated with TGF-ß to induce FMT, and the effect of DM-C was analyzed. DM-C suppressed the expression of α-SMA and the phosphorylation levels of Smad 2/3 and GSK-3, indicating that DM-C suppressed α-SMA expression by inhibiting the transforming growth factor (TGF)-ß signaling pathway via activation of GSK-3. DM-C decreased the expression of collagen, connective tissue growth factor (CTGF) and Snail, which are also known to accelerate cardiac fibrosis. These results suggested that DM-C attenuated cardiac fibrosis by suppressing FMT at the injured site after CMI by inhibiting the TGF-ß signaling pathway via activation of GSK-3. Thus, DM-C has potential against cardiac disease as a novel anti-fibrotic agent.


Assuntos
Fibroblastos/efeitos dos fármacos , Congelamento/efeitos adversos , Infarto do Miocárdio/tratamento farmacológico , Miofibroblastos/efeitos dos fármacos , Pirazóis/uso terapêutico , Transdução de Sinais/efeitos dos fármacos , Sulfonamidas/uso terapêutico , Animais , Células Cultivadas , Fibroblastos/enzimologia , Fibroblastos/patologia , Fibrose , Quinase 3 da Glicogênio Sintase/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Infarto do Miocárdio/enzimologia , Infarto do Miocárdio/etiologia , Infarto do Miocárdio/patologia , Miofibroblastos/enzimologia , Miofibroblastos/patologia , Nitrogênio/toxicidade , Pirazóis/farmacologia , Ratos , Ratos Endogâmicos Lew , Transdução de Sinais/fisiologia , Sulfonamidas/farmacologia
6.
Eur J Pharmacol ; 909: 174415, 2021 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-34375673

RESUMO

Angiotensin II (Ang II) reportedly facilitates primary tumor growth and distal hematogenous metastasis formation in various murine intravenous metastasis models. However, it is unclear whether Ang II accelerates the initial processes of metastasis formation that begins in primary tumors surrounded by tumor microenvironment. We examined the effects of Ang II on primary tumors and lung metastasis lesions using a murine spontaneous metastasis model, in which triple negative breast cancer 4T1 cells constitutively expressing luciferase (4T1-Luc cells) were injected into the mammary fat pad of BALB/c mice. Subcutaneous injection of Ang II significantly accelerated primary tumor growth and lung metastasis formation. Ang II increased the protein expression levels of c-Myc, cyclin D1, fibronectin, vimentin, αSMA and Snail, and the treatment with the Ang II type 1 receptor blocker valsartan significantly suppressed the Ang II-induced increases of fibronectin and vimentin. Valsartan also significantly reduced lung metastatic lesions. However, Ang II did not have significant effects on 4T1-Luc cells including the proliferation, migration, invasion, or the expressions of proteins related to cell proliferation and epithelial-to-mesenchymal transition. In contrast, when 4T1-Luc cells were co-cultured with dermal fibroblasts, Ang II significantly accelerated cell migration and increased the expressions of fibronectin, vimentin, αSMA and Snail in 4T1-Luc cells. And moreover, Ang II significantly increased the mRNA expression of IL-6 in fibroblasts co-cultured with 4T1-Luc cells. These results suggested that Ang II accelerates surrounding fibroblasts by soluble factors such as IL-6 to promote epithelial-to-mesenchymal transition, which result in the initiation of cancer metastasis.


Assuntos
Angiotensina II/metabolismo , Fibroblastos Associados a Câncer/patologia , Neoplasias Pulmonares/secundário , Neoplasias de Mama Triplo Negativas/patologia , Animais , Fibroblastos Associados a Câncer/metabolismo , Linhagem Celular Tumoral , Modelos Animais de Doenças , Transição Epitelial-Mesenquimal , Feminino , Humanos , Pulmão/patologia , Glândulas Mamárias Animais/patologia , Camundongos , Microambiente Tumoral
7.
Oncogene ; 40(37): 5579-5589, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34304250

RESUMO

We have previously reported that the differentiation-inducing factor-1 (DIF-1), a compound identified in Dictyostelium discoideum, suppresses the growth of MCF-7 breast cancer cells by inactivating p70 ribosomal protein S6 kinase (p70S6K). Therefore, we first examined whether the same mechanism operates in other breast cancer cells, especially triple-negative breast cancer (TNBC), the most aggressive and refractory phenotype of breast cancer. We also investigated the mechanism by which DIF-1 suppresses p70S6K by focusing on the AMPK-mTORC1 system. We found that DIF-1 induces phosphorylation of AMPK and Raptor and dephosphorylation of p70S6K in multiple TNBC cell lines. Next, we examined whether AMPK-mediated inhibition of p70S6K leads to the suppression of proliferation and migration/infiltration of TNBC cells. DIF-1 significantly reduced the expression levels of cyclin D1 by suppressing the translation of STAT3 and strongly suppressed the expression levels of Snail, which led to the suppression of growth and motility, respectively. Finally, we investigated whether DIF-1 exerts anticancer effects on TNBC in vivo. Intragastric administration of DIF-1 suppressed tumor growth and spontaneous lung metastasis of 4T1-Luc cells injected into the mammary fat pad of BALB/c mice. DIF-1 is expected to lead to the development of anticancer drugs, including anti-TNBC, by a novel mechanism.


Assuntos
Alvo Mecanístico do Complexo 1 de Rapamicina , Neoplasias de Mama Triplo Negativas , Proteínas Quinases Ativadas por AMP , Animais , Humanos , Camundongos , Proteínas Quinases S6 Ribossômicas 70-kDa , Transdução de Sinais
8.
J Hypertens ; 39(5): 892-903, 2021 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-33252422

RESUMO

BACKGROUND: We reported that 2,5-dimethylcelecoxib (DM-celecoxib), a celecoxib derivative that is unable to inhibit cyclooxygenase-2, prevented cardiac remodeling induced by sarcomeric gene mutation, left ventricular pressure overload, or ß-adrenergic receptor stimulation. This effect seemed to be mediated by the inhibition of the canonical Wnt/ß-catenin signaling pathway, which has been suggested to play a key role in the development of chronic kidney disease and chronic heart failure. METHOD: We investigated the effect of DM-celecoxib on cardiac remodeling and kidney injury in hypertension model mice induced by angiotensin II infusion in the absence or presence of high-salt load. RESULTS: DM-celecoxib prevented cardiac remodeling and markedly reduced urinary albumin excretion without altering blood pressure in those mice. Moreover, DM-celecoxib prevented podocyte injury, glomerulosclerosis, and interstitial fibrosis in the kidney of mice loaded with angiotensin II and high-salt load. DM-celecoxib reduced the phosphorylation level of Akt and activated glycogen synthase kinase-3, which led to the suppression of the Wnt/ß-catenin signal in the heart and kidney. DM-celecoxib also reduced the expression level of snail, a key transcription factor for the epithelial-mesenchymal transition and of which gene is a target of the Wnt/ß-catenin signal. CONCLUSION: Results of the current study suggested that DM-celecoxib could be beneficial for patients with hypertensive heart and kidney diseases.


Assuntos
Angiotensina II , Hipertensão , Animais , Humanos , Hipertensão/induzido quimicamente , Rim , Camundongos , Pirazóis , Sulfonamidas
9.
Biochim Biophys Acta Mol Cell Res ; 1868(2): 118909, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33189784

RESUMO

Mesenchymal stem cells (MSCs) are an attractive cell source for tissue regeneration and repair. However, their low differentiation efficacy currently impedes the development of MSC therapy. Therefore, in this study, we investigated the effects of differentiation-inducing factor-1 (DIF-1) on the differentiation efficacy of bone marrow-derived MSCs (BM-MSCs) into adipogenic or osteogenic lineages. BM-MSCs, which were obtained from Sprague-Dawley rats, were positive for the MSC markers (CD29, CD73, and CD90). DIF-1 alone neither affected cell surface antigen expression nor induced adipogenic or osteogenic differentiation. However, DIF-1 significantly enhanced the effects of adipogenic differentiation stimuli, which were evaluated as the number of oil red-O positive cells and the expression of adipocyte differentiation markers (peroxisome proliferator-activated receptor gamma, adipocyte fatty acid-binding protein, and adiponectin). In contrast, DIF-1 significantly attenuated the effects of osteogenic differentiation stimuli, which were evaluated as alizarin red-S positive calcium deposition, and the expression of osteoblast differentiation markers alkaline phosphatase, runt-related transcription factor 2, and osteopontin. We further investigated the mechanism by which DIF-1 affects MSC differentiation efficacy and found that glycogen synthase kinase-3 was the main factor mediating the action of DIF-1 on the adipogenic differentiation of BM-MSCs, whereas it was only partially involved in osteogenic differentiation. These results suggest that DIF-1 supports MSC differentiation toward the desired cell fate by enhancing the differentiation efficacy.


Assuntos
Adipogenia/efeitos dos fármacos , Hexanonas/farmacologia , Hidrocarbonetos Clorados/farmacologia , Células-Tronco Mesenquimais/metabolismo , Osteogênese/efeitos dos fármacos , Adipócitos/metabolismo , Animais , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Quinase 3 da Glicogênio Sintase/metabolismo , Masculino , Osteoblastos/metabolismo , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos
10.
Cancer Sci ; 110(12): 3761-3772, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31553107

RESUMO

Differentiation-inducing factor-1 (DIF-1) has been reported to inhibit the proliferation of various mammalian cells by unknown means, although some possible mechanisms of its action have been proposed, including the activation of glycogen synthase kinase-3 (GSK-3). Here, we report an alternative mechanism underlying the action of DIF-1 in human breast cancer cell line MCF-7, on which the effects of DIF-1 have not been examined previously. Intragastric administration of DIF-1 reduced the tumor growth from MCF-7 cells injected into a mammary fat pad of nude mice, without causing adverse effects. In cultured MCF-7, DIF-1 arrested the cell cycle in G0 /G1 phase and suppressed cyclin D1 expression, consistent with our previous results obtained in other cell species. However, DIF-1 did not inhibit the phosphorylation of GSK-3. Investigating an alternative mechanism for the reduction of cyclin D1, we found that DIF-1 reduced the protein levels of signal transducer and activator of transcription 3 (STAT3). The STAT3 inhibitor S3I-201 suppressed cyclin D1 expression and cell proliferation and the overexpression of STAT3 enhanced cyclin D1 expression and accelerated proliferation. Differentiation-inducing factor-1 did not reduce STAT3 mRNA or reduce STAT3 protein in the presence of cycloheximide, suggesting that DIF-1 inhibited STAT3 protein synthesis. Seeking its mechanism, we revealed that DIF-1 inhibited the activation of 70 kDa and/or 85 kDa ribosomal protein S6 kinase (p70S6K /p85S6K ). Inhibition of p70S6K /p85S6K by rapamycin also reduced the expressions of STAT3 and cyclin D1. Therefore, DIF-1 suppresses MCF-7 proliferation by inhibiting p70S6K /p85S6K activity and STAT3 protein synthesis followed by reduction of cyclin D1 expression.


Assuntos
Ciclina D1/antagonistas & inibidores , Hexanonas/farmacologia , Hidrocarbonetos Clorados/farmacologia , Proteínas Quinases S6 Ribossômicas/fisiologia , Fator de Transcrição STAT3/antagonistas & inibidores , Animais , Proliferação de Células/efeitos dos fármacos , Ciclina D1/análise , Feminino , Quinase 3 da Glicogênio Sintase/metabolismo , Humanos , Células MCF-7 , Camundongos , Camundongos Endogâmicos BALB C , Fosforilação , Proteínas Quinases S6 Ribossômicas 70-kDa , Fator de Transcrição STAT3/biossíntese
11.
Biochem Pharmacol ; 168: 82-90, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31229551

RESUMO

We previously reported that 2,5-dimethylcelecoxib (DM-celecoxib), a celecoxib derivative that is unable to inhibit cyclooxygenase-2, prevented cardiac remodeling by activating glycogen synthase kinase-3 (GSK-3) and prolonged the lifespan of heart failure mice with genetic dilated cardiomyopathy or transverse aortic constriction-induced left ventricular hypertrophy. However, it remained unclear how DM-celecoxib regulated structure and function of cardiomyocytes and cardiac fibroblasts involved in cardiac remodeling. In the present study, therefore, we investigated the effect of DM-celecoxib on isoprenaline-induced cardiomyocyte hypertrophy and cardiac fibroblast activation, because DM-celecoxib prevented isoprenaline-induced cardiac remodeling in vivo. DM-celecoxib suppressed isoprenaline-induced neonatal rat cardiomyocyte hypertrophy by the inhibition of Akt phosphorylation resulting in the activation of GSK-3 and the inhibition of ß-catenin and mammalian target of rapamycin (mTOR). DM-celecoxib also suppressed the proliferation and the production of matrix metalloproteinase-2 and fibronectin of rat cardiac fibroblasts. Moreover, we found that phosphatase and tensin homolog on chromosome 10 (PTEN) could be a molecule to mediate the effect of DM-celecoxib on Akt. These results suggest that DM-celecoxib directly improves the structure and function of cardiomyocytes and cardiac fibroblasts and that this compound could be clinically useful for the treatment of ß-adrenergic receptor-mediated maladaptive cardiac remodeling.


Assuntos
Cardiomegalia/induzido quimicamente , Cardiomegalia/tratamento farmacológico , Fibroblastos/efeitos dos fármacos , Quinase 3 da Glicogênio Sintase/metabolismo , Isoproterenol/farmacologia , Miócitos Cardíacos/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Pirazóis/uso terapêutico , Sulfonamidas/uso terapêutico , Animais , Animais Recém-Nascidos , Modelos Animais de Doenças , Fibroblastos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Miócitos Cardíacos/metabolismo , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Pirazóis/farmacologia , Ratos , Ratos Sprague-Dawley , Sulfonamidas/farmacologia , Remodelação Ventricular/efeitos dos fármacos
12.
J Pharmacol Sci ; 139(3): 209-214, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30773300

RESUMO

The aim of this study was to investigate the contribution of gene polymorphisms, in combination with habitual caffeine consumption, to the effect of caffeine intake on hemodynamic and psychoactive parameters. A double-blind, prospective study was conducted with 201 healthy volunteers randomly allocated 2:1 to the caffeinated group (150 mL decaffeinated coffee with additional 200 mg caffeine) or decaffeinated group (150 mL decaffeinated coffee). We measured the changes in blood pressure (BP) and calculation speed upon coffee intake, stratifying with gene polymorphisms, e.g., those in adenosine A2A receptor (ADORA2A) and cytochrome P450 (CYP) 1A2, and daily caffeine consumption (≤90 mg/day and >90 mg/day). Overall, caffeine intake independently increased BP and calculation speed (p-values < 0.05), irrespective of the polymorphisms. In stratified analysis, a statistical significance within the caffeinated group was observed for the change in systolic BP in the stratum of CYP1A2 polymorphism with daily caffeine consumption ≤90 mg/day: change in systolic BP in the CYP1A2 rs762551 CC group (mean ± SD = 11.8 ± 5.9) was higher than that in the AA/CA group (4.1 ± 5.5). Gene polymorphisms may limitedly modify the effect of caffeine intake on hemodynamic parameters in combination with habitual caffeine consumption.


Assuntos
Pressão Sanguínea/efeitos dos fármacos , Cafeína/farmacologia , Citocromo P-450 CYP1A2/genética , Frequência Cardíaca/efeitos dos fármacos , Café , Método Duplo-Cego , Feminino , Humanos , Masculino , Matemática , Polimorfismo Genético , Estudos Prospectivos , Receptor A2A de Adenosina/genética , Adulto Jovem
13.
Biochem Pharmacol ; 165: 207-213, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30776323

RESUMO

Rheumatoid arthritis (RA) is a chronic inflammatory joint disease that causes swelling, bone erosion, and joint disorder. Patients with RA therefore suffer from pain and physiological disability, and have a decreased quality of life. During the progression of RA, many different types of cells and inflammatory factors influence each other with an important role. A better understanding of the pathology of RA should therefore lead to the development of effective anti-rheumatoid drugs, such as the anti-TNFα antibody. Glycogen synthase kinase-3 (GSK-3) is a cytoplasmic serine/threonine protein kinase that is involved in a large number of key cellular processes and is dysregulated in a wide variety of diseases, including inflammation and osteoporosis. The accumulated evidence has suggested that GSK-3 could be involved in multiple steps in the progression of RA. In the present review, the mechanisms of the pathogenesis of RA are summarized, and recent developments and potential new drugs targeting GSK-3 are discussed.


Assuntos
Antirreumáticos/farmacologia , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Animais , Regeneração Óssea/fisiologia , Quinase 3 da Glicogênio Sintase/fisiologia , Humanos , Inflamação/etiologia , Osteoclastos/fisiologia , Osteogênese/fisiologia , Células Th17/imunologia
14.
J UOEH ; 40(2): 147-156, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29925734

RESUMO

Glycogen synthase kinase-3 (GSK-3) is a cytoplasmic serine/threonine protein kinase which is known to regulate a variety of cellular processes through a number of signaling pathways important for cell proliferation, stem cell renewal, apoptosis and development. Although GSK-3 exists in a variety of tissues, this kinase plays very important roles in the heart to control its development through the formation of heart and cardiomyocyte proliferation. GSK-3 is also recognized as one of the main molecules that control cardiac hypertrophy and fibrosis. Therefore, GSK-3 could be an attractive target for the development of new drugs to cure cardiac diseases. The present review summarizes the roles of GSK-3 in the signaling pathways and the heart, and discusses the possibility of new drug development targeting this kinase.


Assuntos
Quinase 3 da Glicogênio Sintase/metabolismo , Cardiopatias/enzimologia , Coração/embriologia , Coração/crescimento & desenvolvimento , Miocárdio/enzimologia , Animais , Humanos , Transdução de Sinais
15.
Biochem Pharmacol ; 154: 136-147, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29674000

RESUMO

Hypertension is considered as one of the cancer progressive factors, and often found comorbidity in cancer patients. Renin-angiotensin system (RAS) plays an important role in the regulation of blood pressure, and angiotensin II (Ang II) is well known pressor peptide associated with RAS. Ang II has been reported to accelerate progression and metastasis of cancer cells. However, its precise mechanisms have not been fully understood. In this study, we sought to elucidate the mechanisms by which Ang II exacerbates hematogenous metastasis in mouse melanoma cells, focusing the adhesion pathway in vascular endothelial cells. For this purpose, B16/F10 mouse melanoma cells, which do not express the Ang II type 1 receptor (AT1R), were intravenously injected into C57BL/6 mice. Two weeks after cell injection, the number of lung metastatic colonies was significantly higher in the Ang II-treated group (1 µg/kg/min) than in the vehicle-treated group. The AT1R blocker valsartan (40 mg/kg/day), but not the calcium channel blocker amlodipine (5 or 10 mg/kg/day), significantly suppressed the effect of Ang II. In endothelium-specific Agtr1a knockout mice, Ang II-mediated acceleration of lung metastases of melanoma cells was significantly diminished. Ang II treatment significantly increased E-selectin mRNA expression in vascular endothelial cells collected from lung tissues, and thus promoted adherence of melanoma cells to the vascular endothelium. Ang II-accelerated lung metastases of melanoma cells were also suppressed by treatment with anti-E-selectin antibody (20 mg/kg). Taken together, Ang II-treatment exacerbates hematogenous cancer metastasis by promoting E-selectin-mediated adhesion of cancer cells to vascular endothelial cells.


Assuntos
Angiotensina II/toxicidade , Moléculas de Adesão Celular/metabolismo , Células Endoteliais/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundário , Melanoma Experimental/metabolismo , Animais , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/fisiologia , Relação Dose-Resposta a Droga , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/patologia , Neoplasias Pulmonares/patologia , Masculino , Melanoma Experimental/induzido quimicamente , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Distribuição Aleatória
16.
Biochem Pharmacol ; 151: 96-103, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29534876

RESUMO

Hypertension, which often exists as a comorbid condition in cancer patients, is considered as a factor affecting cancer progression. The renin-angiotensin system (RAS) plays an important role in the regulation of blood pressure, and angiotensin II (Ang II) is a well-known pressor peptide in RAS. There is also accumulated evidence indicating that Ang II plays a critical role in the metastasis of various cancers by modulating adhesion, migration invasion, proliferation, and angiogenesis. Consistent with this, large epidemiological studies have reported the potential beneficial effects of angiotensin-converting enzyme (ACE) inhibitors and Ang II type 1 receptor blockers (ARBs) against cancer metastasis; however, some of the results remain controversial. Although the precise Ang II-related mechanisms involved in cancer metastasis are not completely clear yet, a number of basic and meta-analytic studies have shown that ACE inhibitors and ARBs reduce the metastatic potential of tumors. In this review, we summarize the relationships among hypertension, RAS, and metastasis as demonstrated in basic and clinical studies. Finally, we discuss the possibility of using RAS inhibitors as anti-metastatic drugs.


Assuntos
Bloqueadores do Receptor Tipo 1 de Angiotensina II/uso terapêutico , Angiotensina II/metabolismo , Inibidores da Enzima Conversora de Angiotensina/uso terapêutico , Metástase Neoplásica/tratamento farmacológico , Sistema Renina-Angiotensina/efeitos dos fármacos , Adesão Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Humanos , Metástase Neoplásica/patologia
17.
Biochem Biophys Res Commun ; 493(1): 9-13, 2017 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-28939042

RESUMO

Chronic kidney disease (CKD) causes hyperphosphatemia and secondary hyperparathyroidism, leading to several disorders of bone metabolism. Although high concentrations of extracellular inorganic phosphate (Pi) inhibit osteoclastogenesis, the molecular mechanism of this effect has not been fully understood. In the present study, therefore, we examined the effect of Pi on the differentiation of the osteoclast precursor RAW-D cells. Treatment with the receptor activator of nuclear factor-kappa B ligand induced the differentiation of RAW-D cells (osteoclastogenesis). However, Pi significantly weakened this effect, assessed by the tartrate-resistant acid phosphatase (TRAP) activity and the number of TRAP-positive multinucleated cells. Pi also reduced the expressions of nuclear factor of activated T-cell (NFAT) c1 and dendritic cell-specific transmembrane protein (DC-STAMP). Interestingly, the Pi-induced reduction of DC-STAMP gene promoter activity was lost when the activator protein 1 (AP-1) binding site was mutated. Since Pi strongly inhibited the expression of c-Fos which is the component of AP-1, the Pi-induced reduction of DC-STAMP expression was proposed to be mediated by the absence of c-Fos. These results suggested that hyperphosphatemia in the patients with CKD suppresses bone resorption by inhibiting osteoclastogenesis, and this impairs the regulation of bone metabolism.


Assuntos
Fusão Celular , Proteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Osteoclastos/metabolismo , Osteogênese/fisiologia , Fosfatos/administração & dosagem , Ligante RANK/metabolismo , Fator de Transcrição AP-1/metabolismo , Animais , Diferenciação Celular , Linhagem Celular , Relação Dose-Resposta a Droga , Camundongos , Osteoclastos/citologia
18.
Biochem Pharmacol ; 138: 31-48, 2017 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-28501501

RESUMO

Differentiation-inducing factor-1 (DIF-1) isolated from Dictyostelium discoideum strongly inhibits the proliferation of various mammalian cells through the activation of glycogen synthase kinase-3 (GSK-3). To evaluate DIF-1 as a novel anti-cancer agent for malignant melanoma, we examined whether DIF-1 has anti-proliferative, anti-migratory, and anti-invasive effects on melanoma cells using in vitro and in vivo systems. DIF-1 reduced the expression levels of cyclin D1 and c-Myc by facilitating their degradation via GSK-3 in mouse (B16BL6) and human (A2058) malignant melanoma cells, and thereby strongly inhibited their proliferation. DIF-1 suppressed the canonical Wnt signaling pathway by lowering the expression levels of transcription factor 7-like 2 and ß-catenin, key transcription factors in this pathway. DIF-1 also inhibited cell migration and invasion, reducing the expression of matrix metalloproteinase-2; however, this effect was not dependent on GSK-3 activity. In a mouse lung tumor formation model, repeated oral administrations of DIF-1 markedly reduced melanoma colony formation in the lung. These results suggest that DIF-1 inhibits cell proliferation by a GSK-3-dependent mechanism and suppresses cell migration and invasion by a GSK-3-independent mechanism. Therefore, DIF-1 may have a potential as a novel anti-cancer agent for the treatment of malignant melanoma.


Assuntos
Antineoplásicos/uso terapêutico , Quinase 3 da Glicogênio Sintase/metabolismo , Hexanonas/uso terapêutico , Hidrocarbonetos Clorados/uso terapêutico , Melanoma/tratamento farmacológico , Proteínas de Neoplasias/metabolismo , Via de Sinalização Wnt/efeitos dos fármacos , Animais , Antineoplásicos/efeitos adversos , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Feminino , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Quinase 3 da Glicogênio Sintase/química , Quinase 3 da Glicogênio Sintase/genética , Hexanonas/efeitos adversos , Hexanonas/farmacologia , Humanos , Hidrocarbonetos Clorados/efeitos adversos , Hidrocarbonetos Clorados/farmacologia , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Pulmão/patologia , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Melanoma/metabolismo , Melanoma/patologia , Melanoma Experimental/tratamento farmacológico , Melanoma Experimental/metabolismo , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Invasividade Neoplásica/patologia , Invasividade Neoplásica/prevenção & controle , Proteínas de Neoplasias/agonistas , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/genética , Interferência de RNA , Distribuição Aleatória , Carga Tumoral/efeitos dos fármacos
19.
Hypertens Res ; 40(2): 130-139, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27628899

RESUMO

Glycogen synthase kinase-3 (GSK-3) is a crucial regulator of cardiac hypertrophy. We previously reported that 2,5-dimethylcelecoxib (DM-celecoxib), a celecoxib derivative unable to inhibit cyclooxygenase-2, prevented cardiac remodeling by activating GSK-3, resulting in lifespan prolongation in a mouse model of genetic dilated cardiomyopathy. In the present study, we investigated whether DM-celecoxib can also prevent pressure-induced cardiac remodeling and heart failure, elicited by transverse aortic constriction (TAC). Before testing the effects of DM-celecoxib, we compared the effects of TAC on the hearts of wild-type and GSK-3ß hetero-deficient (GSK-3ß+/-) mice to determine the role of GSK-3 in cardiac remodeling and heart failure. GSK-3ß+/- mouse hearts exhibited more severe hypertrophy, which was characterized by accelerated interstitial fibrosis, than wild-type mouse hearts after TAC, suggesting that reduced GSK-3ß activity aggravates pressure-induced left ventricular remodeling. We subsequently examined the effects of DM-celecoxib on TAC-induced cardiac remodeling. DM-celecoxib inhibited left ventricular systolic functional deterioration, and prevented left ventricular hypertrophy and fibrosis. It also activated GSK-3α and ß by inhibiting Akt, suppressing the activity of ß-catenin and nuclear factor of activated T-cells and thereby decreasing the expression of the Wnt/ß-catenin target gene products fibronectin and matrix metalloproteinase-2. These results suggest that DM-celecoxib is clinically useful for treating pressure-induced heart diseases.


Assuntos
Cardiomegalia/metabolismo , Cardiomiopatia Dilatada/metabolismo , Quinase 3 da Glicogênio Sintase/metabolismo , Ventrículos do Coração/efeitos dos fármacos , Pirazóis/farmacologia , Sulfonamidas/farmacologia , Remodelação Ventricular/efeitos dos fármacos , Animais , Cardiomegalia/patologia , Cardiomiopatia Dilatada/patologia , Modelos Animais de Doenças , Quinase 3 da Glicogênio Sintase/genética , Ventrículos do Coração/metabolismo , Ventrículos do Coração/patologia , Metaloproteinase 2 da Matriz/metabolismo , Camundongos , Camundongos Knockout , Proteínas Proto-Oncogênicas c-akt/metabolismo
20.
Cancer Sci ; 108(1): 108-115, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27761963

RESUMO

We previously reported that celecoxib, a selective COX-2 inhibitor, strongly inhibited human colon cancer cell proliferation by suppressing the Wnt/ß-catenin signaling pathway. 2,5-Dimethylcelecoxib (DM-celecoxib), a celecoxib analog that does not inhibit COX-2, has also been reported to have an antitumor effect. In the present study, we elucidated whether DM-celecoxib inhibits intestinal cancer growth, and its underlying mechanism of action. First, we compared the effect of DM-celecoxib with that of celecoxib on the human colon cancer cell lines HCT-116 and DLD-1. 2,5-Dimethylcelecoxib suppressed cell proliferation and inhibited T-cell factor 7-like 2 expression with almost the same strength as celecoxib. 2,5-Dimethylcelecoxib also inhibited the T-cell factor-dependent transcription activity and suppressed the expression of Wnt/ß-catenin target gene products cyclin D1 and survivin. Subsequently, we compared the in vivo effects of celecoxib and DM-celecoxib using the Mutyh-/- mouse model, in which oxidative stress induces multiple intestinal carcinomas. Serum concentrations of orally administered celecoxib and DM-celecoxib elevated to the levels enough to suppress cancer cell proliferation. Repeated treatment with celecoxib and DM-celecoxib markedly reduced the number and size of the carcinomas without showing toxicity. These results suggest that the central mechanism for the anticancer effect of celecoxib derivatives is the suppression of the Wnt/ß-catenin signaling pathway but not the inhibition of COX-2, and that DM-celecoxib might be a better lead compound candidate than celecoxib for the development of novel anticancer drugs.


Assuntos
Celecoxib/farmacologia , Neoplasias Intestinais/tratamento farmacológico , Neoplasias Intestinais/patologia , Pirazóis/farmacologia , Sulfonamidas/farmacologia , Proteínas Wnt/metabolismo , Via de Sinalização Wnt/efeitos dos fármacos , beta Catenina/metabolismo , Animais , Contagem de Células Sanguíneas , Peso Corporal/efeitos dos fármacos , Celecoxib/sangue , Celecoxib/uso terapêutico , Linhagem Celular Tumoral , DNA Glicosilases/deficiência , DNA Glicosilases/genética , Feminino , Humanos , Neoplasias Intestinais/metabolismo , Masculino , Camundongos , Estresse Oxidativo/efeitos dos fármacos , Proteólise/efeitos dos fármacos , Pirazóis/sangue , Pirazóis/uso terapêutico , Sulfonamidas/sangue , Sulfonamidas/uso terapêutico , Fatores de Transcrição TCF/metabolismo , Proteína 2 Semelhante ao Fator 7 de Transcrição/metabolismo , Transcrição Gênica/efeitos dos fármacos , beta Catenina/antagonistas & inibidores
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...