Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 94
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Life Sci ; 335: 122278, 2023 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-37981227

RESUMEN

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.


Asunto(s)
Dictyostelium , Molécula 1 de Adhesión Celular Vascular , Ratones , Animales , Masculino , Humanos , Molécula 1 de Adhesión Celular Vascular/metabolismo , Lipopolisacáridos/farmacología , Dictyostelium/metabolismo , Ratones Endogámicos C57BL , Proteínas , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina , Diferenciación Celular , Adhesión Celular , Mamíferos/metabolismo
2.
J Pharmacol Sci ; 152(1): 39-49, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-37059490

RESUMEN

Differentiation-inducing factor 1 (DIF-1) is a morphogen produced by Dictyostelium discoideum that inhibits the proliferation and migration of both D. discoideum and most mammalian cells. Herein, we assessed the effect of DIF-1 on mitochondria, because DIF-3, which is similar to DIF-1, reportedly localizes in the mitochondria when added exogenously, however the significance of this localization remains unclear. Cofilin is an actin depolymerization factor that is activated by dephosphorylation at Ser-3. By regulating the actin cytoskeleton, cofilin induces mitochondrial fission, the first step in mitophagy. Here, we report that DIF-1 activates cofilin and induces mitochondrial fission and mitophagy mainly using human umbilical vein endothelial cells (HUVECs). AMP-activated kinase (AMPK), a downstream molecule of DIF-1 signaling, is required for cofilin activation. Pyridoxal phosphatase (PDXP)-known to directly dephosphorylate cofilin-is also required for the effect of DIF-1 on cofilin, indicating that DIF-1 activates cofilin through AMPK and PDXP. Cofilin knockdown inhibits mitochondrial fission and decreases mitofusin 2 (Mfn2) protein levels, a hallmark of mitophagy. Taken together, these results indicate that cofilin is required for DIF-1- induced mitochondrial fission and mitophagy.


Asunto(s)
Dictyostelium , Hexanonas , Animales , Humanos , Proteínas Quinasas Activadas por AMP , Factores Despolimerizantes de la Actina/metabolismo , Factores Despolimerizantes de la Actina/farmacología , Dinámicas Mitocondriales , Dictyostelium/metabolismo , Células Endoteliales/metabolismo , Diferenciación Celular , Monoéster Fosfórico Hidrolasas , Piridoxal/farmacología , Hexanonas/farmacología , Mamíferos/metabolismo
3.
Nihon Yakurigaku Zasshi ; 158(2): 111, 2023.
Artículo en Japonés | MEDLINE | ID: mdl-36858487
4.
Nihon Yakurigaku Zasshi ; 158(2): 112-118, 2023.
Artículo en Japonés | MEDLINE | ID: mdl-36858488

RESUMEN

A variety of new methods are being tried in education of pharmacology for medical students, to make pharmacology be directly oriented to practical medical treatment. Among them, thinking that the method of "selection of personal drug (P-drug)" is suitable for learning "evidence-based medicine (EBM)", I have been engaged in "P-drug education" in Kyushu university for many years. If doctors carefully select medicines that are indispensable for their medical treatment based on clinical evidence, are made familiar with how to use them, and in principle perform daily medical treatment using only those medicines, EBM can be really practiced. And moreover, it may also lead to the suppression of medical errors and adverse drug reactions. Such essential medicines for an individual doctor are called P-drug. Since 2003, I have adopted "P-drug selection" in the education of pharmacology for upper grade medical students. After more than 15 years of trial and error, I have been able to create an educational model using "P-drug selection" that I think could be easily adopted at any medical school. At this symposium, I talked about the relationship between "P-drug selection" and EBM and demonstrate the "P-drug education" model.


Asunto(s)
Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Estudiantes de Medicina , Humanos , Aprendizaje , Medicina Basada en la Evidencia , Facultades de Medicina
5.
Nihon Yakurigaku Zasshi ; 158(2): 128-133, 2023.
Artículo en Japonés | MEDLINE | ID: mdl-36858490

RESUMEN

Pharmacology role-play works well for students playing the role of the medical doctor or patient, but students without any roles behave just like observers, resulting in a relatively low learning effectiveness. To improve this issue, a personal drug (P-drug) report was introduced to the role-play program. To examine to what extent the P-drug report affected the learning effectiveness of role-play, we performed questionnaire surveys for players and audiences and subsequent nominal logistic regression analysis. The questionnaire topics were (1) understanding of medical treatment, (2) understanding patient's feelings, (3) improvement of awareness and motivation as a medical doctor, and (4) positive influence upon study attitude. In the topics (1) and (2), the statistical analyses in audiences showed significant relationship between the introduction of the report and observer's recognition of the learning effectiveness, indicating the improvement of learning effectiveness after the introduction of the P-drug report. In players, the percentage of high marks was higher than that in audiences, and no significant differences were found between before and after the introduction of the report. In addition, in the free description, many students realized the importance of selection of therapeutic drugs based on P-drug methods. These results suggest that the introduction of the P-drug report seems useful to make all students participate in the activity of role-play with understanding the selection process of therapeutic drugs, and improves the learning effectiveness of role-play especially in observers. It may be useful to combine P-drug with pharmacology role-play in practical pharmacotherapy education.


Asunto(s)
Aprendizaje , Proyectos de Investigación , Humanos
6.
Int Immunopharmacol ; 117: 109913, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-36812674

RESUMEN

The tumor microenvironment (TME), largely composed of tumor-associated macrophages (TAMs) and cancer-associated fibroblasts (CAFs), plays a key role in cancer progression. A small molecule, differentiation-inducing factor-1 (DIF-1) secreted by Dictyostelium discoideum, is known to exhibit anticancer activity; however, its effect on the TME remains unknown. In this study, we investigated the effect of DIF-1 on the TME using mouse triple-negative breast cancer 4T1-GFP cells, mouse macrophage RAW 264.7 cells, and mouse primary dermal fibroblasts (DFBs). Polarization of 4T1 cell-conditioned medium-induced macrophage into TAMs was not affected by DIF-1. In contrast, DIF-1 decreased 4T1 cell co-culturing-induced C-X-C motif chemokine ligand 1 (CXCL1), CXCL5, and CXCL7 expression in DFBs and suppressed DFB differentiation into CAF-like cells. Additionally, DIF-1 inhibited C-X-C motif chemokine receptor 2 (CXCR2) expression in 4T1 cells. Immunohistochemical analyses of tumor tissue samples excised from breast cancer-bearing mice showed that DIF-1 did not affect the number of CD206-positive TAMs; however, it decreased the number of α-smooth muscle actin-positive CAFs and CXCR2 expression. These results indicated that the anticancer effect of DIF-1 was partially attributed to the inhibition of CXCLs/CXCR2 axis-mediated communication between breast cancer cells and CAFs.


Asunto(s)
Fibroblastos Asociados al Cáncer , Dictyostelium , Neoplasias , Animales , Ratones , Fibroblastos Asociados al Cáncer/metabolismo , Neoplasias/metabolismo , Macrófagos/metabolismo , Fibroblastos , Comunicación , Microambiente Tumoral , Línea Celular Tumoral
7.
Hypertens Res ; 45(12): 1869-1881, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36171325

RESUMEN

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.


Asunto(s)
Neoplasias Renales , Neoplasias Pancreáticas , Humanos , Antihipertensivos/uso terapéutico , Antagonistas de Receptores de Angiotensina/uso terapéutico , Antagonistas de Receptores de Angiotensina/farmacología , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Sistema Renina-Angiotensina , Estudios Retrospectivos , Registros Electrónicos de Salud , Inhibidores Enzimáticos/farmacología , Neoplasias Pancreáticas/inducido químicamente , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Renales/tratamiento farmacológico
8.
Eur J Pharmacol ; 909: 174415, 2021 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-34375673

RESUMEN

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.


Asunto(s)
Angiotensina II/metabolismo , Fibroblastos Asociados al Cáncer/patología , Neoplasias Pulmonares/secundario , Neoplasias de la Mama Triple Negativas/patología , Animales , Fibroblastos Asociados al Cáncer/metabolismo , Línea Celular Tumoral , Modelos Animales de Enfermedad , Transición Epitelial-Mesenquimal , Femenino , Humanos , Pulmón/patología , Glándulas Mamarias Animales/patología , Ratones , Microambiente Tumoral
9.
Oncogene ; 40(37): 5579-5589, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34304250

RESUMEN

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.


Asunto(s)
Diana Mecanicista del Complejo 1 de la Rapamicina , Neoplasias de la Mama Triple Negativas , Proteínas Quinasas Activadas por AMP , Animales , Humanos , Ratones , Proteínas Quinasas S6 Ribosómicas 70-kDa , Transducción de Señal
10.
Low Urin Tract Symptoms ; 13(2): 299-307, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33089671

RESUMEN

OBJECTIVE: To develop a new mouse model of underactive bladder (UAB) caused by chronic bladder outlet obstruction (BOO). METHODS: BOO was created in 6-week-old male C57BL/6 mice using surgery to loosely place a silver jump ring around the bladder neck of each mouse. Micturition behavior (assessed with a metabolic cage) and cystometry were used to evaluate bladder function at 8 and 16 weeks after BOO. Following completion of the functional studies, the bladders of the mice were excised, weighed, and subjected to histological analysis. RESULTS: Micturition behavior analysis showed that mice subjected to BOO for 16 weeks had a lower frequency of micturition (7.3 ± 1.1 vs 12.5 ± 3.0 times/d, P < .05) and volume per void (106.0 ± 0.1 vs 133.9 ± 3.2 µL, P < .05) than mice subjected to BOO for 8 weeks. Cystometry revealed that mice subjected to BOO for 16 weeks had lower baseline pressure (8.4 ± 0.6 vs 14.0 ± 0.7 cmH2 O, P < .01) and micturition pressure (13.9 ± 1.1 vs 42.8 ± 1.7 cmH2 O, P < .05) than mice subjected to BOO for 8 weeks. BOO caused progressive increases in bladder mass and collagen deposition over time. CONCLUSIONS: We successfully established a novel mouse model of UAB using surgery to place a silver jump ring loosely on the bladder neck. BOO initially induced bladder overactivity but subsequently resulted in UAB due to deterioration of detrusor smooth muscle contractility and progressive deposition of collagen in the bladder wall.


Asunto(s)
Obstrucción del Cuello de la Vejiga Urinaria , Vejiga Urinaria de Baja Actividad , Animales , Masculino , Ratones , Ratones Endogámicos C57BL , Obstrucción del Cuello de la Vejiga Urinaria/etiología , Micción
11.
J Hypertens ; 39(5): 892-903, 2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-33252422

RESUMEN

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.


Asunto(s)
Angiotensina II , Hipertensión , Animales , Humanos , Hipertensión/inducido químicamente , Riñón , Ratones , Pirazoles , Sulfonamidas
12.
Sci Rep ; 10(1): 2891, 2020 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-32076019

RESUMEN

This study aimed to examine the effect of pool walking on renal function in pregnant women. Fifteen pregnant women (mean gestational age, 37.8 weeks) walked in a pool (depth 1.3 m) for 1 h. A few days later, they walked on a street for 1 h. Within each activity, the starting and ending levels of plasma renin activity were measured. The total urine volume, creatinine clearance, and change in plasma renin activity levels between each activity were compared by Wilcoxon rank-sum test. The renin-angiotensin-aldosterone level was suppressed during pool walking: the mean starting and ending values of plasma renin activity and serum aldosterone were 6.8 vs. 5.5 ng/mL/h (p = 0.002) and 654 vs. 473 pg/mL (p = 0.01), respectively. The decreases in plasma renin activity and serum aldosterone levels were more evident in pool walking than in land walking (plasma renin activity, -1.27 vs. 0.81 ng/mL/h, p = 0.002; serum aldosterone, -180.9 vs. 3.1 ng/mL/h, p = 0.03), resulting in higher total urine volume (299 vs. 80 mL, p < 0.001) and creatinine clearance (161.4 vs. 123.4 mL/min, p = 0.03) in pool walking. Pool walking may improve renal function in pregnant women partly through the suppressed renin-angiotensin-aldosterone system.


Asunto(s)
Pruebas de Función Renal , Riñón/fisiología , Sistema Renina-Angiotensina/fisiología , Caminata/fisiología , Adulto , Femenino , Humanos , Embarazo
13.
Hypertens Res ; 43(3): 220-226, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31748705

RESUMEN

We investigated changes in blood pressure (BP) and metabolic adverse effects, especially elevation of uric acid (UA), after treatment with a thiazide-like diuretic (TD) in patients with essential hypertension. Furthermore, the role of genetic factors in the elevation of UA by TD was assessed by a 500 K SNP DNA microarray. The subjects included 126 hypertensive patients (57 women and 69 men, mean age 59 ± 12 years) who registered for the GEANE (Gene Evaluation for ANtihypertensive Effects) study. After one month of the nontreatment period, TD, indapamide, angiotensin II receptor antagonist valsartan, and Ca channel blocker amlodipine were administered to all patients for 3 months each in a randomized crossover manner. BP, renal function, serum UA level, and electrolytes were measured at baseline and at the end of each treatment period. Single nucleotide polymorphisms (SNPs) associated with UA elevation after treatment with indapamide were investigated by a genome-wide association study (GWAS). Indapamide significantly decreased both office and home BP levels. Treatment with indapamide also significantly reduced the estimated glomerular filtration rate and serum potassium and increased serum UA. Patients whose UA level increased more than 1 mg/dl showed significantly higher baseline office SBP and plasma glucose and showed greater decline in renal function compared with those who showed less UA increase (<1 mg/dl). Some SNPs strongly associated with an increase in UA after treatment with indapamide were identified. This study is the first report on SNPs associated with UA elevation after TD treatment. This information may be useful for the prevention of adverse effects after treatment with TD.


Asunto(s)
Diuréticos/uso terapéutico , Hipertensión Esencial/genética , Indapamida/uso terapéutico , Polimorfismo de Nucleótido Simple , Ácido Úrico/sangre , Anciano , Amlodipino/farmacología , Amlodipino/uso terapéutico , Antagonistas de Receptores de Angiotensina/farmacología , Antagonistas de Receptores de Angiotensina/uso terapéutico , Presión Sanguínea/efectos de los fármacos , Bloqueadores de los Canales de Calcio/farmacología , Bloqueadores de los Canales de Calcio/uso terapéutico , Estudios Cruzados , Diuréticos/farmacología , Hipertensión Esencial/sangre , Hipertensión Esencial/tratamiento farmacológico , Femenino , Estudio de Asociación del Genoma Completo , Humanos , Indapamida/farmacología , Masculino , Persona de Mediana Edad , Valsartán/farmacología , Valsartán/uso terapéutico
14.
Cancer Sci ; 110(12): 3761-3772, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31553107

RESUMEN

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.


Asunto(s)
Ciclina D1/antagonistas & inhibidores , Hexanonas/farmacología , Hidrocarburos Clorados/farmacología , Proteínas Quinasas S6 Ribosómicas/fisiología , Factor de Transcripción STAT3/antagonistas & inhibidores , Animales , Proliferación Celular/efectos de los fármacos , Ciclina D1/análisis , Femenino , Glucógeno Sintasa Quinasa 3/metabolismo , Humanos , Células MCF-7 , Ratones , Ratones Endogámicos BALB C , Fosforilación , Proteínas Quinasas S6 Ribosómicas 70-kDa , Factor de Transcripción STAT3/biosíntesis
15.
Biochem Pharmacol ; 168: 82-90, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31229551

RESUMEN

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.


Asunto(s)
Cardiomegalia/inducido químicamente , Cardiomegalia/tratamiento farmacológico , Fibroblastos/efectos de los fármacos , Glucógeno Sintasa Quinasa 3/metabolismo , Isoproterenol/farmacología , Miocitos Cardíacos/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Pirazoles/uso terapéutico , Sulfonamidas/uso terapéutico , Animales , Animales Recién Nacidos , Modelos Animales de Enfermedad , Fibroblastos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Miocitos Cardíacos/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Pirazoles/farmacología , Ratas , Ratas Sprague-Dawley , Sulfonamidas/farmacología , Remodelación Ventricular/efectos de los fármacos
16.
J Pharmacol Sci ; 139(3): 209-214, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30773300

RESUMEN

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.


Asunto(s)
Presión Sanguínea/efectos de los fármacos , Cafeína/farmacología , Citocromo P-450 CYP1A2/genética , Frecuencia Cardíaca/efectos de los fármacos , Café , Método Doble Ciego , Femenino , Humanos , Masculino , Matemática , Polimorfismo Genético , Estudios Prospectivos , Receptor de Adenosina A2A/genética , Adulto Joven
17.
JA Clin Rep ; 4(1): 59, 2018 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-32025958

RESUMEN

INTRODUCTION: To examine whether sex and polymorphisms of cytochrome P450 (CYP) 2B6 and UDP-glucuronosyltransferase (UGT) 1A9 affect the difference between predicted and measured plasma propofol concentration during continuous infusion by target-controlled infusion. RESULTS: Blood samples of 69 patients (48 men and 21 women) were obtained at 4 h after initial propofol infusion. Percentage performance error (PE) was calculated to assess the difference between measured and predicted propofol concentration. Regression coefficients (ß) and 95% confidence intervals (CI) of sex and the polymorphisms of CYP2B6 and UGT1A9 for PE were, separately and mutually, estimated with linear regression. Covariates included age and body mass index in the minimal adjusted model, and additionally included clinical factors (mean blood pressure, heart rate, volume of intravenous fluid, surgical site, surgical position, and pneumoperitoneum) in the full adjusted model. PE was higher in men than in women (28.7% versus 10.5%, p = 0.015). Female sex was inversely associated with PE: the minimal adjusted ß = - 8.84 (95% CI, - 16.26 to - 1.43); however, the fully adjusted ß with clinical factors became not significant. The average of PE did not differ between polymorphisms of CYP2B6 and UGT1A9, and ß of CYP2B6 516G>T polymorphisms mutually adjusted with female sex was not significant. Mean blood pressure, heart rate, and volume of intravenous fluid were independently associated with PE in the full adjusted model. CONCLUSIONS: Under 4 h anesthesia with propofol target-controlled infusion in our population, sex differences appeared to exist in the propofol concentration, which might be largely mediated by clinical factors, such as hemodynamic status. TRIAL REGISTRATION: UMIN-CTR UMIN000009015 , Registered 1 October 2012.

18.
Arch Biochem Biophys ; 637: 40-47, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29169900

RESUMEN

The plant-derived flavonoid, quercetin (QCT), has many biological actions, including cardioprotective actions, resulting from its antioxidant and anti-inflammatory effects. In this study, effects of QCT and its metabolites on the contraction and Ca2+ transients (CaT) of mouse single cardiomyocytes were simultaneously measured and compared with those of isoproterenol and digoxin. Furthermore, cardiac function and plasma concentrations were analyzed after bolus intravenous administration of QCT in mice. QCT and its metabolite, tamarixetin, as well as isoproterenol and digoxin, enhanced the contraction and CaT of cardiomyocytes. The inotropic action of isoproterenol was accompanied by an increase in the velocities of sarcomere shortening and relengthening and CaT decay through activation of cAMP-dependent protein kinase; however, no such lusitropic effects accompanied the inotropic action of QCT, tamarixetin or digoxin. Intravenous administration of QCT to mice resulted in a sustained increase in cardiac systolic function; QCT was rapidly metabolized to tamarixetin and its plasma concentration was maintained at high levels over a similar time frame as the enhancement of cardiac systolic function. These results suggest that QCT exerts a cardiotonic action in vivo at least, in part, through digitalis-like enhancement of CaT by itself and its metabolite tamarixetin.


Asunto(s)
Señalización del Calcio/efectos de los fármacos , Cardiotónicos/farmacología , Disacáridos/farmacología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/fisiología , Quercetina/análogos & derivados , Quercetina/farmacología , Animales , Cardiotónicos/metabolismo , Glicósidos Digitálicos/farmacología , Digoxina/farmacología , Disacáridos/metabolismo , Técnicas In Vitro , Isoproterenol/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Contracción Miocárdica/efectos de los fármacos , Quercetina/metabolismo
19.
Biochem Biophys Res Commun ; 493(1): 9-13, 2017 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-28939042

RESUMEN

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.


Asunto(s)
Fusión Celular , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Osteoclastos/metabolismo , Osteogénesis/fisiología , Fosfatos/administración & dosificación , Ligando RANK/metabolismo , Factor de Transcripción AP-1/metabolismo , Animales , Diferenciación Celular , Línea Celular , Relación Dosis-Respuesta a Droga , Ratones , Osteoclastos/citología
20.
Biochem Pharmacol ; 138: 31-48, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28501501

RESUMEN

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.


Asunto(s)
Antineoplásicos/uso terapéutico , Glucógeno Sintasa Quinasa 3/metabolismo , Hexanonas/uso terapéutico , Hidrocarburos Clorados/uso terapéutico , Melanoma/tratamiento farmacológico , Proteínas de Neoplasias/metabolismo , Vía de Señalización Wnt/efectos de los fármacos , Animales , Antineoplásicos/efectos adversos , Antineoplásicos/farmacología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Femenino , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3/química , Glucógeno Sintasa Quinasa 3/genética , Hexanonas/efectos adversos , Hexanonas/farmacología , Humanos , Hidrocarburos Clorados/efectos adversos , Hidrocarburos Clorados/farmacología , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/patología , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Melanoma/metabolismo , Melanoma/patología , Melanoma Experimental/tratamiento farmacológico , Melanoma Experimental/metabolismo , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Invasividad Neoplásica/patología , Invasividad Neoplásica/prevención & control , Proteínas de Neoplasias/agonistas , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Interferencia de ARN , Distribución Aleatoria , Carga Tumoral/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...