Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Cell Biol Int ; 47(4): 787-795, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36626273

RESUMO

Tribbles homolog 2 (TRIB2) functions as an adapter protein that regulates signal transductions involved in a variety of cellular functions, including tumorigenesis. However, the role of TRIB2 in the proliferation of vascular smooth muscle cells (VSMCs) and the underlying expression mechanisms remain unclear. The present study investigated the role of TRIB2 in VSMC proliferation and revealed that TRIB2 expression increases following vascular injury and platelet-derived growth factor (PDGF)-BB-stimulated VSMCs. We found that pretreatment with diphenyleneiodonium (a nicotinamide adenine dinucleotide phosphate oxidase inhibitor), U0126 (an inhibitor of mitogen-activated protein kinase kinase 1 [MEK1]), or siRNA targeting the gene encoding early growth response 1 (EGR-1) significantly inhibits PDGF-BB-induced TRIB2 expression in VSMCs. Furthermore, TRIB2 knockdown significantly inhibits PDGF-BB-induced proliferation of VSMCs but does not affect the phosphorylation of AKT. However, phosphorylation of ERK1 and expression of proliferating cell nuclear antibody are significantly suppressed in VSMCs by PDGF-BB stimulation. Thus, PDGF-BB-induced TRIB2 expression is mediated by ROS/ERK/EGR-1 pathways and plays a critical role in VSMC proliferation via modulation of ERK activity. We propose TRIB2 as a promising therapeutic target for the prevention of neointima formation and vascular disease.


Assuntos
Proliferação de Células , Músculo Liso Vascular , Transdução de Sinais , Animais , Ratos , Becaplermina/metabolismo , Becaplermina/farmacologia , Movimento Celular , Proliferação de Células/genética , Células Cultivadas , Sistema de Sinalização das MAP Quinases , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo
2.
Cardiovasc Drugs Ther ; 37(6): 1175-1192, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-35150385

RESUMO

Erythropoiesis-stimulating agents (ESAs) have markedly reduced the need for blood transfusion for renal anemia and are included in standard therapies for patients with chronic kidney disease (CKD). Various protective effects of ESAs on the cardiovascular system have been discovered through basic research, and the effects have received much attention because the rates of cardiovascular events and mortality are high in CKD patients. However, randomized clinical trials did not provide strong evidence that ESAs exert cardioprotection in humans, including CKD patients. It is difficult to assess the cardioprotective effects of ESAs in CKD patients through the clinical data that has been reported to date because the relationship between hemoglobin level rather than ESA dose and cardiovascular event rates was examined in most studies. Interestingly, recent studies using a rat model of CKD showed that the infarct size-limiting effect of an ESA was lost when its dose was increased to a level that normalized blood hemoglobin levels, suggesting that the optimal dose of an ESA for myocardial protection is less than the dose required to normalize hemoglobin levels. Furthermore, animal models of traditional coronary risk factors or comorbidities were resistant to the cardioprotective effects of ESAs because of interruptions in signal-mediated mechanisms downstream of erythropoietin receptors. In this review, we briefly discuss basic and clinical data on the impact of anemia on coronary and systemic circulation, the effects of CKD on the cardiovascular system, and the multiple pharmacological actions of ESAs to examine whether the ESAs that are prescribed for renal anemia exert any cardioprotection in patients with CKD.


Assuntos
Anemia , Sistema Cardiovascular , Eritropoetina , Hematínicos , Insuficiência Renal Crônica , Humanos , Animais , Ratos , Hematínicos/efeitos adversos , Eritropoetina/farmacologia , Eritropoese , Insuficiência Renal Crônica/complicações , Insuficiência Renal Crônica/diagnóstico , Insuficiência Renal Crônica/tratamento farmacológico , Anemia/tratamento farmacológico , Anemia/etiologia , Doença Crônica , Hemoglobinas/farmacologia , Hemoglobinas/uso terapêutico
3.
J Nat Med ; 77(2): 276-283, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36477683

RESUMO

Although various diterpenoid alkaloids have been evaluated recently for antiproliferative activity against human tumor cell lines, little information is available regarding the antiproliferative effects of C20-diterpenoid alkaloids against MCF-7 cells. Six new diterpenoid alkaloid derivatives (13, 14, 22, 23, 25, 26) were prepared by C-11 and 15 esterification of kobusine (6). The natural parent alkaloid 6 and all synthesized derivatives (7 - 27, 12a, 15a, 15b, 18a, 18b) were evaluated for antiproliferative activity against MCF-7 cells. The structure-based design strategy resulted in an initial lead derivative, 11,15-dibenzoylkobusine (7; IC50 8.6 µM). Subsequent synthesized 11,15-diacylkobusine derivatives (9, 16, 20, 21, 23, 25, and 26) showed substantially increased suppressive effects against the MCF-7 cell line (IC50 2.3-4.4 µM). In contrast, parent alkaloid 6, two 11-acylkobusine derivatives (15a, 18a), and two 15-acylkobusine derivatives (15b, 18b) showed no effect. 11,15-Diacylation appears to be critical for producing antiproliferative activity in this alkaloid class and could introduce a new avenue in overcoming breast cancer cell proliferation using natural product derivatives. In a preliminary mechanism of action study, representative derivatives (5, 8, 9, and 17) decreased cyclin D1 mRNA expression.


Assuntos
Alcaloides , Antineoplásicos , Neoplasias da Mama , Diterpenos , Humanos , Feminino , Células MCF-7 , Alcaloides/farmacologia , Relação Estrutura-Atividade , Diterpenos/farmacologia , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Proliferação de Células , Estrutura Molecular
4.
Clin Nutr ; 41(12): 2691-2698, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36343560

RESUMO

BACKGROUND & AIMS: Milk-derived miR-148a-3p (miR-148a), which is abundant in breast milk, has been shown to be associated with the development of infants' intestines. Although it is well known that stress during lactation changes milk constituents in terms of lipid and protein, no studies have examined the influence of stress on miR-148a expression in breast milk. The objective of this study is to investigate the relationship between stress and miR-148a expression in milk, and to evaluate whether the changes in milk-derived miR-148a expression-caused by the mother's exposure to stress-influence intestinal ZO-1 expression in infants. METHODS: The participants of this study were healthy Japanese women who were nursing. Psychological stress evaluation of the subjects was conducted using a short form of the Profile of Mood State Second Edition-Adult (POMS-2). Additionally, miR-148a expressions in restraint stressed nursing mice were investigated using quantitative real-time PCR. The levels of a tight junction protein zonula occludens-1 (ZO-1) and DNA methyltransferase 1 (DNMT1), which is a direct target of miR-148a, in ileum in neonatal mice breastfed by stressed nursing mice were investigated using Western blot. Furthermore, to investigate the influence of miR-148a on ZO-1 expression within the intestine, the levels of ZO-1 and DNMT1 in human intestinal epithelial Caco-2 cells with lentivirus-mediated miR-148a overexpression were evaluated. RESULTS: A significantly negative correlation was observed between relative miR-148a expression in breast milk and the total mood disturbance T-score. Each T-score on negative mood subscales of anger-hostility, confusion-bewilderment, depression-dejection, fatigue-inertia, and tension-anxiety was significantly negatively correlated with relative miR-148a expression in breast milk: a positive mood subscale vigor-activity T-score was significantly positively correlated with relative miR-148a expression in breast milk. A positive mood friendliness T-score, estimated separately from other scores, was significantly positively correlated with relative miR-148a expression in breast milk. Additionally, the relative expression of miR-148a in the milk obtained from stressed mice was significantly lower than that of control mice. The relative level of ZO-1 in ileum of neonatal mice nursed by stressed mice was significantly lower than that of neonatal mice nursed by control mice. Additionally, the relative level of DNMT1 in ileum of neonatal mice nursed by stressed mice was significantly higher than that of neonatal mice nursed by control mice. Furthermore, the relative level of ZO-1 in miR-148a-overexpressed Caco-2 cells was significantly higher than that in control cells. The relative level of DNMT1 in miR-148a-overexpressed Caco-2 cells was significantly lower than that in control cells. CONCLUSIONS: Mothers' exposure to stress during lactation may cause miR-148a expression in breast milk. Additionally, stressed-induced suppression of miR-148a expression in breast milk may cause a decrease in intestinal ZO-1 level via the increase in DNMT1 in infants' intestines. These observations are beneficial information for breastfeeding mothers and their families and perinatal medical professionals. Our findings encourage monitoring maternal psychological stress during lactation to promote breastfeeding and adequate infant nutrition.


Assuntos
MicroRNAs , Junções Íntimas , Adulto , Animais , Feminino , Humanos , Lactente , Camundongos , Gravidez , Células CACO-2 , Intestinos , MicroRNAs/genética , Leite Humano , Junções Íntimas/metabolismo
5.
Int Breastfeed J ; 17(1): 1, 2022 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-34980190

RESUMO

BACKGROUND: Milk-derived microRNAs (miRNAs), including hsa-miR-148a-3p (miR-148a) and hsa-miR-125b-5p (miR-125b), have been shown to be beneficial to the gastrointestinal function in infants. Here, we investigated their expression during lactation in humans and determined whether the infant formulae available in Japan contain these miRNAs. METHODS: Healthy Japanese women (n = 16) who gave birth vaginally or by cesarean section at the Teine Keijinkai Hospital between 1 September 2020, and 31 April 2021 were included in this study. Breast milk was collected by nurses on days 4 or 5 after delivery (hereinafter, transition milk) and on day 30 of postpartum (hereinafter, mature milk). The levels of miR-148a and miR-125b in breastmilk and six commercially available infant formulae were compared and evaluated using quantitative reverse transcription-polymerase chain reaction. RESULTS: In all participants, the miR-148a level in mature breastmilk was significantly lower than that in the transition milk. The changes in miR-125b expression during lactation showed similar trends to the changes in miR-148a expression. The miR-148a and miR-125b levels in all analyzed infant formulae were lower than 1/500th and 1/100th of those in mature breastmilk, respectively. CONCLUSIONS: The levels of both miR-148a and miR-125b in human breast milk decreased on day 30 postpartum compared with those in the transition milk. Additionally, the expression of these miRNAs in infant formulae available in Japan was very low. Further studies with larger populations are required to understand precisely the lactational changes in the expression of miR148a and miR-125b in breast milk.


Assuntos
MicroRNAs , Leite Humano , Aleitamento Materno , Cesárea , Feminino , Humanos , Lactação , MicroRNAs/genética , Gravidez
6.
Biochem Biophys Res Commun ; 545: 1-7, 2021 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-33529804

RESUMO

Mammary epithelial cells synthesize and secrete norepinephrine (NE) into breast milk to regulate ß-casein expression through the adrenergic ß2 receptor. We investigated the expression, localization, and roles of NE transporter (NET) in the mammary epithelium during lactation. mRNA and protein levels of NET were determined in primary normal human mammary epithelial cells (pHMECs) and non-malignant human mammary epithelial MCF-12A cells. In nursing CD1 mice, NET localized to the apical membranes of the mammary epithelium. The intracellular NE content of pHMECs incubated with NE increased. Although the ß-casein concentration in milk was slightly higher at day 10 than at day 2 of lactation, the NE concentration and lactation-related proteins were only slightly changed on days 2-10. Restraint stress increased the NE concentration in milk from nursing mice and NET protein levels were significantly higher than in non-stressed nursing mice. NET is expressed on the apical membrane of mammary epithelial cells and incorporates NE in milk into cells, potentially regulating the NE concentration in milk.


Assuntos
Glândulas Mamárias Humanas/metabolismo , Leite Humano/metabolismo , Proteínas da Membrana Plasmática de Transporte de Norepinefrina/metabolismo , Norepinefrina/metabolismo , Animais , Transporte Biológico Ativo , Caseínas/metabolismo , Linhagem Celular , Células Cultivadas , Células Epiteliais/metabolismo , Feminino , Humanos , Lactação/genética , Lactação/metabolismo , Glândulas Mamárias Animais/citologia , Glândulas Mamárias Animais/metabolismo , Glândulas Mamárias Humanas/citologia , Camundongos , Leite/metabolismo , Proteínas da Membrana Plasmática de Transporte de Norepinefrina/genética , Gravidez , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Restrição Física/efeitos adversos , Estresse Fisiológico/fisiologia
7.
Eur J Pharmacol ; 872: 172924, 2020 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-31958455

RESUMO

Brain and muscle Arnt-like protein-1 (BMAL1), a component of the molecular clock, is implicated in the development of cardiovascular diseases, including atherosclerosis and abdominal aortic aneurysms. However, the role of BMAL1 in vascular proliferation associated with vascular remodeling is unknown. In the present study, we investigated the mechanisms underlying BMAL1 expression in vascular smooth muscle cells (VSMCs) and the role of BMAL1 in VSMC proliferation. BMAL1 expression significantly increased in injured carotid arteries in C57BL/6J mice and platelet-derived growth factor (PDGF)-BB-stimulated VSMC cultures. Pretreatment with diphenyleneiodonium (an NADPH oxidase inhibitor) and U0126 or PD98059 (MEK Inhibitors) inhibited PDGF-BB-induced BMAL1 expression in a dose-dependent manner in VSMCs. In addition, the knockdown of early growth factor protein-1 (Egr-1) significantly inhibited PDGF-BB-induced BMAL1 mRNA or protein expression in VSMCs, and the knockdown of BMAL1 significantly decreased PDGF-BB-induced cell proliferation and extracellular signal-regulated kinase (ERK) phosphorylation but not Akt phosphorylation in VSMCs. The results demonstrate that PDGF-BB up-regulates BMAL1 expression through reactive oxygen species/ERK/Egr-1 pathways and that BMAL1 is involved in PDGF-BB-induced cell proliferation partially through ERK in VSMCs. Thus, BMAL1 may be a novel therapeutic target for the treatment of atherosclerosis including vascular remodeling.


Assuntos
Fatores de Transcrição ARNTL/metabolismo , Lesões das Artérias Carótidas/patologia , Proliferação de Células/fisiologia , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/patologia , Fatores de Transcrição ARNTL/antagonistas & inibidores , Fatores de Transcrição ARNTL/genética , Animais , Aterosclerose/tratamento farmacológico , Aterosclerose/patologia , Becaplermina/farmacologia , Artérias Carótidas/patologia , Lesões das Artérias Carótidas/fisiopatologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Ritmo Circadiano/fisiologia , Modelos Animais de Doenças , Técnicas de Silenciamento de Genes , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Camundongos , Músculo Liso Vascular/citologia , Cultura Primária de Células , Ratos , Espécies Reativas de Oxigênio/metabolismo , Remodelação Vascular/efeitos dos fármacos , Remodelação Vascular/fisiologia
8.
Cell Biol Int ; 44(3): 873-881, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31833156

RESUMO

Cardiac cell death is one of the major events implicated in doxorubicin-induced cardiotoxicity, which leads to heart failure. We recently reported that Yes-associated protein 1 (YAP1) regulates cell survival and apoptosis. However, it is unclear whether YAP1 regulates doxorubicin-induced cell death in cardiomyocytes. We investigated whether YAP1 is involved in doxorubicin-induced cell death using H9c2 cardiac cells and mouse heart. In an in vivo study, YAP1 protein expression was significantly decreased in hearts of doxorubicin-treated mice with increased caspase-3 activation. Doxorubicin also caused cell death by increasing caspase-3 activation in H9c2 cells. Doxorubicin reduced YAP1 protein expression and messenger RNA expression accompanied by increased phosphorylation of YAP1 at Ser127. Doxorubicin further increased cell death with increased caspase-3/7 activation in the absence of YAP1 when compared with doxorubicin or siYAP1 treatment alone. Overexpression of constitutively active YAP1 (YAP1-5SA) using an adenovirus gene transfer technique significantly reversed doxorubicin-induced cell death by decreasing caspase-3/7 activation in H9c2 cells. Akt, a potential prosurvival factor, decreased in doxorubicin- and YAP1 short interfering RNA (siRNA)-treated cells. Doxorubicin further significantly decreased Akt protein expression when YAP1 was silenced. Overexpression of YAP1 canceled decreased Akt protein expression induced by doxorubicin treatment in H9c2 cells. In conclusion, these results suggest that doxorubicin-induced cardiac cell death is mediated in part by down-regulation of YAP1 and YAP1-targeted gene, Akt. Modulating YAP1 and its related Hippo pathway on local cardiomyocytes may be a promising therapeutic approach for doxorubicin-induced cardiotoxicity.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Proteínas de Ciclo Celular/fisiologia , Morte Celular/efeitos dos fármacos , Doxorrubicina/toxicidade , Miócitos Cardíacos/efeitos dos fármacos , Animais , Cardiotoxicidade , Caspase 3/metabolismo , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Miócitos Cardíacos/citologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Proteínas de Sinalização YAP
9.
Yakugaku Zasshi ; 138(11): 1329-1334, 2018.
Artigo em Japonês | MEDLINE | ID: mdl-30381640

RESUMO

Impaired insulin signaling in adipose tissue and skeletal muscle causes insulin resistance associated with the development of type 2 diabetes. However, the molecular mechanisms underlying insulin resistance remain to be elucidated. In this review, we describe the current understanding of the effects of 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitors (statins) and tumor necrosis factor (TNF)-α on insulin signal transduction in adipocytes. First, we determined that atorvastatin inhibits the tyrosine phosphorylation of insulin receptor substrate (IRS)-1 through a decrease in the RhoA-Rho-kinase pathway, resulting in the inhibition of glucose uptake. Second, we found that TNF-α induces IRS-1 phosphorylation at serine residues 636/639 and inhibits the tyrosine phosphorylation of IRS-1 through the increase in both extracellular signal-regulated kinase (ERK) and c-jun N-terminal kinase (JNK) phosphorylation. Interestingly, 5-aminoimidazole-4-carboxamide-1-ß-D-ribofuranoside, an AMP-activated protein kinase activator, suppresses TNF-α-induced IRS-1 serine phosphorylation at 636/639 and the phosphorylation of ERK by enhancing interactions between ERK and dual-specificity phosphatase-9. These results may be helpful in understanding the mechanisms underlying insulin resistance.


Assuntos
Atorvastatina/efeitos adversos , Diabetes Mellitus/etiologia , Inibidores de Hidroximetilglutaril-CoA Redutases/efeitos adversos , Mediadores da Inflamação/fisiologia , Resistência à Insulina , Insulina/fisiologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Fator de Necrose Tumoral alfa/fisiologia , Proteínas Quinases Ativadas por AMP/fisiologia , Adipócitos/metabolismo , Animais , Fosfatases de Especificidade Dupla/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Proteínas Substratos do Receptor de Insulina/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Camundongos , Fosfatases da Proteína Quinase Ativada por Mitógeno/metabolismo , Fosforilação/efeitos dos fármacos , Serina/metabolismo , Tirosina/metabolismo , Quinases Associadas a rho/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo
10.
Anticancer Res ; 38(7): 4305-4311, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-29970566

RESUMO

BACKGROUND/AIM: Mulberry (Morus alba L.) leaves (ML) contain many functional components, such as 1-deoxynojirimycin, flavonoids (rutin, quercetin, kaempferol). It is well known that 1-deoxynojirimycin functions to suppress increases in blood glucose level by α-glucosidase inhibitory activity. Thus, the molecular mechanism underlying the protective and therapeutic effects of ML supplementation was investigated on a mouse model of high-calorie diet (Western diet: WD)-induced hepatic steatosis (HS). MATERIALS AND METHODS: The C57BL/6J mouse was used for the HS model. The mice were divided into three groups: control (normal diet: ND), WD, and WD + 1% ML groups. The WD group was fed a high-calorie (high carbohydrate and high fat) diet for 12 weeks to develop HS. At week 12, all mice were sacrificed, blood was collected for biochemical tests, and the liver was obtained for histological examination and RNA sequencing (RNA-Seq). RESULTS: Liver weight, plasma triglycerides (TG), alanine aminotransferase (ALT), and alanine aminotransferase (AST) levels of both ML groups were significantly lower than those of the WD group. On histological examination of the liver, the area of fatty deposits was found to be suppressed by ML administration. In the gene expression analysis of the liver of WD- versus ML-fed mice by RNA-Seq, 722/45,706 genes exhibited a significant change in expression (corrected p-value<0.05). Gene network analysis of these genes showed that genes related to liver inflammation were inactivated and those related to regeneration of liver were activated in the ML group. CONCLUSION: ML functions to suppress HS in WD-fed mice and regulates genes related to inflammation and regeneration of liver cells.


Assuntos
Fígado/efeitos dos fármacos , Morus , Hepatopatia Gordurosa não Alcoólica , Extratos Vegetais/farmacologia , Transcriptoma/efeitos dos fármacos , Animais , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Folhas de Planta , Pós
11.
Eur J Pharmacol ; 815: 470-477, 2017 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-28951205

RESUMO

Apoptosis of vascular smooth muscle cells (VSMCs) has been implicated in the progression of atherosclerosis, especially in vascular remodelling and plaque rupture. Although it is known that Yes-associated protein 1 (YAP1) is a critical molecule that regulates cell proliferation, differentiation and apoptosis, the role of YAP1 in VSMCs apoptosis remains unknown. In this study, we investigated whether YAP1 modulates VSMC apoptosis induced by endoplasmic reticulum (ER) stress. In cultured VSMC, tunicamycin caused cell death accompanied by an increase in caspase-3 processing and C/EBP homologous protein (CHOP) expression. YAP1 protein expression was downregulated by tunicamycin and the phosphorylation of YAP1 at the Ser127 site was significantly increased by tunicamycin. Tunicamycin further decreased cell viability followed by an increase in caspase-3 processing in the absence of YAP1 when compared with treatment only with tunicamycin or siYAP1. On the other hand, overexpression of a constitutively active YAP1 (YAP1-5SA), which lacks five serine phosphorylation sites, significantly prevented the caspase-3 processing and restored the decrease in cell viability induced by tunicamycin. Overexpression of YAP1-5SA significantly inhibited tunicamycin-induced caspase-8 processing without affecting phosphorylation of p-38 and Akt. Furthermore, the overexpression of YAP1-5SA significantly restored the decrease in ANKRD1 expression induced by tunicamycin. The inhibition of tunicamycin-induced caspase-3 cleavage by YAP1-5SA was markedly attenuated in ANKRD1-knockdown cells. These results demonstrate that ER stress can alter intracellular YAP1 protein expression in VSMCs and that YAP1 is protective against VSMC apoptosis induced by ER stress through inhibiting caspase8/3 activation mediated in part by upregulation of ANKRD1.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Estresse do Retículo Endoplasmático , Músculo Liso Vascular/citologia , Animais , Proteínas Reguladoras de Apoptose/deficiência , Proteínas Reguladoras de Apoptose/genética , Caspase 3/metabolismo , Morte Celular/efeitos dos fármacos , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Masculino , Proteínas Musculares/metabolismo , Músculo Liso Vascular/efeitos dos fármacos , Proteínas Nucleares/metabolismo , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Sprague-Dawley , Proteínas Repressoras/metabolismo , Tunicamicina/farmacologia , Proteínas de Sinalização YAP , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
12.
In Vivo ; 31(4): 599-607, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28652426

RESUMO

BACKGROUND/AIM: Senescence marker protein-30/gluconolactonase knockout mice (SMP-30/GNL-KO) are a very useful model for clarifying the involvement of vitamin C (VC) in aging-related diseases. In this study, the effects of VC deficiency on skin and hair growth were investigated using SMP-30/GNL-KO mice by RNA sequencing. MATERIALS AND METHODS: SMP-30/GNL-KO mice were given water containing 1.5 g/l VC until up to 8 weeks after birth to maintain a VC concentration in their organs and plasma equivalent to that in wild-type mice. The mice were then divided into two groups: a VC(+) group, where VC was administered, and a VC(-) group, where VC was not administered. Skin samples were collected at 4 and 8 weeks after the treatment. RNA was extracted from each skin sample, followed by cDNA synthesis and RNA-seq. In addition, hair growth was compared between the VC(-) and VC(+) groups after shaving. Skin samples were collected from the shaved area for histological examination by hematoxylin & eosin (HE) staining. RESULTS: RNA-seq revealed that there were 1,736 (FDR<0.001) differentially expressed genes in the VC(-) and VC(+) groups. From the functional analysis of the differentially expressed genes in the VC(-) and VC(+) groups, predicted functionalities including cell death and cytotoxicity increased in the VC(+) group. Furthermore, it was predicted that the difference in hair growth between the VC(-) and VC(+) groups was caused by the expression of genes including keratin-related genes and the Sonic hedgehog gene. It was confirmed that hair growth was significantly promoted; hair growth from hair papilla cells was also confirmed by HE staining of the shaved backs of SMP-30/GNL-KO mice in the VC(+) group. CONCLUSION: RNA-seq of the skin from VC-deficient mice showed the effects of VC deficiency on the expression of genes involved in cell growth and the hair cycle. Visual inspection suggested that changes in the expression of the genes are involved in delaying hair growth in the VC(-) group. Further research on the relationship among VC deficiency, the hair cycle, and skin cell growth may contribute to research on hair restoration and skin aging.


Assuntos
Envelhecimento/genética , Proteínas de Ligação ao Cálcio/genética , Hidrolases de Éster Carboxílico/genética , Peptídeos e Proteínas de Sinalização Intracelular/genética , Pele/metabolismo , Envelhecimento/metabolismo , Envelhecimento/patologia , Animais , Ácido Ascórbico/genética , Ácido Ascórbico/metabolismo , Deficiência de Ácido Ascórbico/genética , Deficiência de Ácido Ascórbico/patologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento/genética , Cabelo/crescimento & desenvolvimento , Cabelo/metabolismo , Cabelo/patologia , Humanos , Camundongos , Camundongos Knockout , Pele/crescimento & desenvolvimento , Pele/patologia
13.
Cell Biol Int ; 40(11): 1204-1211, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27569904

RESUMO

Impaired insulin signaling in skeletal muscle cells causes insulin resistance associated with the onset of type 2 diabetes. Although interleukin (IL)-1ß has been considered to be implicated in the pathogenesis of type 2 diabetes, the action of prolonged stimulation with IL-1ß on the insulin signaling pathway in skeletal muscle cells remains poorly understood. In the current study, we investigated the effect of IL-1ß stimulation on insulin signal transduction from the insulin receptor (IR), resulting in glucose transporter 4 (GLUT4) translocation in skeletal muscle cells. In L6-GLUT4myc cells, stimulation with IL-1ß for 24 h promoted GLUT4 translocation to the plasma membrane and increased glucose uptake in a concentration-dependent manner, whereas short-term stimulation with IL-1 for up to 6 h did not affect that. In addition, stimulation with IL-1ß for 24 h further increased insulin-stimulated GLUT4 translocation. Interestingly, stimulation with IL-1ß for 24 h did not cause any change in the phosphorylation of insulin signal molecules IR, insulin receptor substrate (IRS)-1, Akt, and p21-activated kinase (PAK1). Stimulation with IL-1ß for 24 h significantly increased AMP-activated protein kinase (AMPK) phosphorylation and GLUT4 protein expression. Small interfering RNA (siRNA) targeting AMPK1/2 significantly inhibited IL-1ß-stimulated GLUT4 translocation. These results suggest that prolonged stimulation with IL-1ß positively regulates GLUT4 translocation in skeletal muscle cells. IL-1ß may have a beneficial effect on maintaining glucose homeostasis in skeletal muscle cells in patients with type 2 diabetes. .


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Transportador de Glucose Tipo 4/metabolismo , Interleucina-1beta/farmacologia , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/metabolismo , Animais , Transporte Biológico/efeitos dos fármacos , Linhagem Celular , Ativação Enzimática/efeitos dos fármacos , Glucose/metabolismo , Insulina/metabolismo , Músculo Esquelético/enzimologia , Fosforilação , Transporte Proteico , RNA Interferente Pequeno , Ratos , Receptor de Insulina/metabolismo , Proteínas Recombinantes/farmacologia , Transdução de Sinais/efeitos dos fármacos
14.
Cell Biol Int ; 40(3): 269-76, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26514426

RESUMO

Increased A disintegrin and metalloprotease 17 (ADAM17) expression in vascular smooth muscle cells (VSMC) is implicated in the development of cardiovascular diseases including atherosclerosis and hypertension. Although cilostazol, type III phosphodiesterase (PDE III) inhibitor, has recently been found to inhibit VSMC proliferation, the mechanisms remain largely unclear. Here, we hypothesized that cilostazol regulates the ADAM17 expression in VSMC. In cultured VSMC, interleukin (IL)-1α and IL-1ß significantly increased ADAM17 expression. MEK inhibitor U0126, NF-κB inhibitor BAY-11-7085, and siRNA targeting p65/RelA significantly inhibited IL-1α or IL-ß-induced ADAM17 expression. Cilostazol significantly inhibited IL-1α or IL-1ß-induced extracellular signal-regulated kinase (ERK) phosphorylation and ADAM17 expression. Unexpectedly, cilostamide, dibutryl cAMP, and forskolin did not affect IL-1-induced ADAM17 expression. Our results clearly demonstrated that IL-1 induces ADAM17 expression through ERK/NF-κB activation in VSMCs. Moreover, the inhibitory effects of cilostazol on IL-1-induced ADAM17 expression may be independent of the cAMP signaling pathway in VSMC. These novel findings may provide important clues to understanding the expression mechanisms of ADAM17 and the inhibitory mechanisms of cilostazol in VSMC proliferation.


Assuntos
Proteína ADAM17/metabolismo , AMP Cíclico/metabolismo , Interleucina-1/farmacologia , Transdução de Sinais/efeitos dos fármacos , Tetrazóis/farmacologia , Animais , Células Cultivadas , Cilostazol , MAP Quinases Reguladas por Sinal Extracelular , Masculino , Músculo Liso Vascular/citologia , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Fosforilação/efeitos dos fármacos , Biossíntese de Proteínas/efeitos dos fármacos , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Sprague-Dawley , Fator de Transcrição RelA/antagonistas & inibidores , Fator de Transcrição RelA/genética , Fator de Transcrição RelA/metabolismo
15.
Cell Biol Int ; 39(9): 1053-60, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25858022

RESUMO

Doxorubicin is an anthracycline antibiotic widely used in cancer treatment. Although its antitumor efficacy appears to be dose dependent, its clinical use is greatly restricted by the development of cardiotoxicity associated with apoptosis. Although caveolin-1, the major structural protein in caveolae, can positively or negatively regulate apoptosis depending on the stimulus or cell types, the contribution of caveolin-1 to doxorubicin-induced apoptosis remains unknown. This study was performed to identify the regulatory role of caveolin-1 on doxorubicin-induced apoptosis in H9c2 cardiac cells using a genetic approach. Caveolin-1 knockdown with a short hairpin (sh) RNA adenovirus, but not overexpression of wild-type caveolin-1, resulted in a marked inhibition of doxorubicin-induced caspase-3 cleavage. However, caveolin-1 knockdown tended to protect against doxorubicin-induced decrease in cell viability, but it did not significantly reverse cell death induced by doxorubicin. Doxorubicin stimulated the phosphorylation of p38 and extracellular signal regulated kinase (ERK). Doxorubicin-induced caspase-3 cleavage was inhibited by U0126, a MEK inhibitor or SB203580, a p38 inhibitor. Caveolin-1 knockdown markedly inhibited doxorubicin-induced p-38 phosphorylation but not ERK-mediated p-53 phosphorylation in H9c2 cardiac cells. Our results suggest that reduced caveolin-1 expression plays an anti-apoptotic role in doxorubicin-induced apoptosis but that it is insufficient to prevent such an apoptosis in H9c2 cardiac cells.


Assuntos
Apoptose/efeitos dos fármacos , Caveolina 1/biossíntese , Doxorrubicina/farmacologia , Miócitos Cardíacos/efeitos dos fármacos , Animais , Antibióticos Antineoplásicos/farmacologia , Caspase 3/metabolismo , Cavéolas/metabolismo , Caveolina 1/metabolismo , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Fosforilação/efeitos dos fármacos , Ratos , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
16.
J Pharmacol Sci ; 122(2): 93-102, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23698110

RESUMO

AMP-activated protein kinase (AMPK) contributes to the acceleration of insulin signaling. However, the mechanism by which AMPK regulates insulin signaling remains unclear. Serine phosphorylation of insulin receptor substrate (IRS)-1 negatively regulates insulin signaling. Here we investigated the role of AMPK in serine phosphorylation of IRS-1 at 636/639 and 307, which is induced by tumor necrosis factor (TNF)-α in 3T3L1 adipocytes. We demonstrated that the AMPK activator 5-aminoimidazole-4-carboxamide-1-d-ribofuranoside (AICAR) significantly inhibited the TNF-α-induced serine phosphorylation of IRS-1 at 636/639 and 307 by suppression of extracellular signal-regulated kinase (ERK) phosphorylation but not c-Jun-NH2-terminal kinase (JNK) phosphorylation. In addition, AICAR stimulation resulted in enhanced interaction between ERK and MAP kinase phosphatase-4 (DUSP9/MKP-4) without affecting DUSP9/MPK4 mRNA synthesis. Moreover, intraperitoneal administration (0.25 g/kg) of AICAR to db/db mice improved blood glucose levels and inhibited the phosphorylation of ERK in adipose tissue. In conclusion, we propose a new mechanism in which AICAR suppresses TNF-α-induced serine phosphorylation of IRS-1 at 636/639 and 307 by enhancing the interaction between ERK and DUSP9/MKP-4. Taken together, these findings provide evidence that AMPK plays a crucial role in improving of type 2 diabetes.


Assuntos
Células 3T3-L1/metabolismo , Proteínas Quinases Ativadas por AMP/fisiologia , Aminoimidazol Carboxamida/análogos & derivados , Hipoglicemiantes/farmacologia , Proteínas Substratos do Receptor de Insulina/metabolismo , Ribonucleotídeos/farmacologia , Serina/metabolismo , Fator de Necrose Tumoral alfa/fisiologia , Aminoimidazol Carboxamida/administração & dosagem , Aminoimidazol Carboxamida/farmacologia , Animais , Células Cultivadas , Diabetes Mellitus Tipo 2/tratamento farmacológico , Diabetes Mellitus Tipo 2/etiologia , Fosfatases de Especificidade Dupla/fisiologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Hipoglicemiantes/administração & dosagem , Injeções Intraperitoneais , Resistência à Insulina/fisiologia , Camundongos , Camundongos Endogâmicos , Fosfatases da Proteína Quinase Ativada por Mitógeno/fisiologia , Fosforilação/efeitos dos fármacos , Ribonucleotídeos/administração & dosagem , Transdução de Sinais , Estimulação Química , Fator de Necrose Tumoral alfa/antagonistas & inibidores
17.
J Mol Cell Cardiol ; 62: 1-7, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23688779

RESUMO

Small interfering RNA (siRNA) mediated gene silencing has been utilized as a powerful molecular tool to study the functional significance of a specific protein. However, due to transient gene silencing and insufficient transfection efficiency, this approach can be problematic in primary cell culture such as vascular smooth muscle cells. To overcome this weakness, we utilized an adenoviral-encoded microRNA (miRNA)-embedded siRNA "mi/siRNA"-based RNA interference. Here, we report the results of silencing a disintegrin and metalloprotease 17 (ADAM17) in cultured rat vascular smooth muscle cells and its functional mechanism in angiotensin II signal transduction. 3 distinct mi/siRNA sequences targeting rat ADAM17 were inserted into pAd/CMV/V5-DEST and adenoviral solutions were obtained. Nearly 90% silencing of ADAM17 was achieved when vascular smooth muscle cells were infected with 100 multiplicity of infection of each ADAM17 mi/siRNA encoding adenovirus for 3days. mi/siRNA-ADAM17 but not mi/siRNA-control inhibited angiotensin II-induced epidermal growth factor receptor trans-activation and subsequent extracellular signal-regulated kinase activation and hypertrophic response in the cells. mi/siRNA-ADAM17 also inhibited angiotensin II-induced heparin-binding epidermal growth factor-like factor shedding. This inhibition was rescued with co-infection of adenovirus encoding mouse ADAM17 but not by its cytosolic domain deletion mutant or cytosolic Y702F mutant. As expected, angiotensin II induced tyrosine phosphorylation of ADAM17 in the cells. In conclusion, ADAM17 activation via its tyrosine phosphorylation contributes to heparin-binding epidermal growth factor-like factor shedding and subsequent growth promoting signals induced by angiotensin II in vascular smooth muscle cells. An artificial mi/siRNA-based adenoviral approach appears to be a reliable gene-silencing strategy for signal transduction research in primary cultured vascular cells.


Assuntos
Proteínas ADAM/genética , Adenoviridae/genética , Angiotensina II/genética , MicroRNAs/genética , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/metabolismo , Proteína ADAM17 , Animais , Linhagem Celular , Células Cultivadas , Humanos , Immunoblotting , Imunoprecipitação , Masculino , RNA Interferente Pequeno/genética , Ratos
18.
Atherosclerosis ; 228(1): 90-3, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23535568

RESUMO

OBJECTIVE: The Max-interacting protein Mnt is a transcriptional repressor that can antagonize the transcriptional and proliferation-related activities of Myc. Here, we tested the hypothesis that Mnt is a negative regulator of pathological vascular remodeling. METHODS: Adenovirus encoding Mnt or control GFP was infected to cultured rat vascular smooth muscle cells (VSMC) and carotid arteries after a balloon angioplasty. RESULTS: In VSMC, adenoviral gene transfer of Mnt suppressed angiotensin II-induced protein expression of early growth response protein-1 (Egr1) and its promoter activation. Mnt adenovirus did not interfere with upstream signaling of angiotensin II. Angiotensin II-induced protein accumulation in VSMC was inhibited by Mnt adenovirus. Mnt adenovirus also inhibited platelet-derived growth factor-induced VSMC proliferation. Moreover, Mnt adenovirus prevented neointima formation in response to arterial injury. The adenoviral Mnt gene transfer also prevented Egr1 induction in neointima. CONCLUSION: These data identify Mnt as a previously unrecognized negative regulator of pathological vascular remodeling.


Assuntos
Angiotensina II/metabolismo , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Lesões das Artérias Carótidas/metabolismo , Músculo Liso Vascular/metabolismo , Neointima/metabolismo , Proteínas Repressoras/metabolismo , Adenoviridae/genética , Angioplastia com Balão/efeitos adversos , Animais , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/genética , Lesões das Artérias Carótidas/patologia , Proteínas de Fluorescência Verde/genética , Hiperplasia/metabolismo , Hiperplasia/patologia , Hipertrofia/metabolismo , Hipertrofia/patologia , Masculino , Músculo Liso Vascular/patologia , Neointima/patologia , Cultura Primária de Células , Ratos , Ratos Sprague-Dawley , Proteínas Repressoras/genética , Transdução de Sinais/fisiologia , Transcrição Gênica/fisiologia
19.
Am J Hypertens ; 25(3): 280-3, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22113169

RESUMO

BACKGROUND: To fill the gap between acute and chronic stimulation methods of angiotensin II (Ang II) and obtain relevant signaling information, we have made an adenovirus vector encoding a furin-cleavable Ang II fusion protein. METHODS: Vascular smooth muscle cells (VSMCs) were infected with adenovirus to evaluate Ang II production. Also, expression of early growth response-1 (Egr-1) and hypertrophic responses were examined in VSMCs. RESULTS: Acute stimulation of VSMCs with synthetic Ang II showed the peptide had a half-life of less than 1 h. Infection of VSMCs with Ang II adenovirus showed a time-dependent production of Ang II as early as 2 days and up to 7 days postinfection. The Ang II adenovirus induced VSMC hypertrophy, stimulated Egr-1 expression, and suppressed Ang II type 1 receptor mRNA expression. Chronic Ang II infusion in mice for 2 weeks markedly enhanced Egr-1 immunostaining in carotid artery compared with the control saline infusion. CONCLUSION: Application of the Ang II adenovirus vector to cultured cells will be useful to elucidate molecular and signaling mechanisms of cardiovascular diseases associated with enhanced Ang II production.


Assuntos
Adenoviridae , Angiotensina II/farmacologia , Artéria Carótida Primitiva/efeitos dos fármacos , Proteína 1 de Resposta de Crescimento Precoce/efeitos dos fármacos , Vetores Genéticos/farmacologia , Músculo Liso Vascular/efeitos dos fármacos , Angiotensina II/metabolismo , Animais , Artéria Carótida Primitiva/metabolismo , Células Cultivadas , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Furina/metabolismo , Expressão Gênica/efeitos dos fármacos , Hipertrofia/metabolismo , Camundongos , Músculo Liso Vascular/metabolismo , Ratos , Ratos Sprague-Dawley
20.
Hypertension ; 57(4): 841-5, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21357274

RESUMO

The requirement of a metalloprotease, a disintegrin and metalloprotease 17 (ADAM17) for the growth of cultured vascular smooth muscle cells has been demonstrated in vitro. However, whether this metalloprotease is responsible for vascular remodeling in vivo remains unanswered. Rat carotid arteries were analyzed 2 weeks after a balloon angioplasty. The neointimal cells were strongly positive for ADAM17 immunostaining. Marked inhibition of intimal hyperplasia was observed in a dominant-negative ADAM17 adenovirus-treated carotid artery. Proliferating cell nuclear antigen-positive cells and phospho-epidermal growth factor receptor-positive cells in the neointima were reduced by dominant-negative ADAM17 as well. In contrast, the neointima formation, proliferating cell nuclear antigen-positive cells, and phospho-epidermal growth factor receptor-positive cells were markedly enhanced by wild-type ADAM17 adenovirus. In conclusion, ADAM17 activation is involved in epidermal growth factor receptor activation and subsequent neointimal hyperplasia after vascular injury. ADAM17 could be a novel therapeutic target for pathophysiological vascular remodeling.


Assuntos
Proteínas ADAM/metabolismo , Artérias Carótidas/patologia , Lesões das Artérias Carótidas/metabolismo , Músculo Liso Vascular/metabolismo , Neointima/patologia , Proteína ADAM17 , Angioplastia com Balão , Animais , Artérias Carótidas/metabolismo , Lesões das Artérias Carótidas/patologia , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Receptores ErbB/metabolismo , Hiperplasia/metabolismo , Imuno-Histoquímica , Masculino , Músculo Liso Vascular/citologia , Músculo Liso Vascular/patologia , Neointima/metabolismo , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA