Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biomedicines ; 11(11)2023 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-38001945

RESUMO

Aging is a growing problem worldwide, and the prevalence and mortality of arterial and venous thromboembolism (VTE) are higher in the elderly than in the young population. To address this issue, various anticoagulants have been used. However, no evidence can confirm that antithrombotic agents are suitable for the elderly. Therefore, this study aims to investigate the platelet proteome of aged mice and identify antithrombotic drug targets specific to the elderly. Based on the proteome analysis of platelets from aged mice, 308 increased or decreased proteins were identified. Among these proteins, three targets were selected as potential antithrombotic drug targets. These targets are membrane proteins or related to platelet function and include beta-2-glycoprotein 1 (ß2GP1, ApolipoproteinH (ApoH)), alpha-1-acid glycoprotein2 (AGP2, Orosomucoid-2 (Orm2)), and Ras-related protein (Rab11a).

2.
Sci Transl Med ; 15(711): eabh3489, 2023 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-37647389

RESUMO

Peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α) is a master regulator of mitochondrial biogenesis. Reduced PGC-1α abundance is linked to skeletal muscle weakness in aging or pathological conditions, such as neurodegenerative diseases and diabetes; thus, elevating PGC-1α abundance might be a promising strategy to treat muscle aging. Here, we performed high-throughput screening and identified a natural compound, farnesol, as a potent inducer of PGC-1α. Farnesol administration enhanced oxidative muscle capacity and muscle strength, leading to metabolic rejuvenation in aged mice. Moreover, farnesol treatment accelerated the recovery of muscle injury associated with enhanced muscle stem cell function. The protein expression of Parkin-interacting substrate (PARIS/Zfp746), a transcriptional repressor of PGC-1α, was elevated in aged muscles, likely contributing to PGC-1α reduction. The beneficial effect of farnesol on aged muscle was mediated through enhanced PARIS farnesylation, thereby relieving PARIS-mediated PGC-1α suppression. Furthermore, short-term exercise increased PARIS farnesylation in the muscles of young and aged mice, whereas long-term exercise decreased PARIS expression in the muscles of aged mice, leading to the elevation of PGC-1α. Collectively, the current study demonstrated that the PARIS-PGC-1α pathway is linked to muscle aging and that farnesol treatment can restore muscle functionality in aged mice through increased farnesylation of PARIS.


Assuntos
Farneseno Álcool , Debilidade Muscular , Animais , Camundongos , Farneseno Álcool/farmacologia , Envelhecimento , Prenilação , Ubiquitina-Proteína Ligases
3.
Cell Mol Life Sci ; 79(2): 99, 2022 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-35089423

RESUMO

Angiotensin II (AngII) has potent cardiac hypertrophic effects mediated through activation of hypertrophic signaling like Wnt/ß-Catenin signaling. In the current study, we examined the role of protein arginine methyltransferase 7 (PRMT7) in cardiac function. PRMT7 was greatly decreased in hypertrophic hearts chronically infused with AngII and cardiomyocytes treated with AngII. PRMT7 depletion in rat cardiomyocytes resulted in hypertrophic responses. Consistently, mice lacking PRMT7 exhibited the cardiac hypertrophy and fibrosis. PRMT7 overexpression abrogated the cellular hypertrophy elicited by AngII, while PRMT7 depletion exacerbated the hypertrophic response caused by AngII. Similar with AngII treatment, the cardiac transcriptome analysis of PRMT7-deficient hearts revealed the alteration in gene expression profile related to Wnt signaling pathway. Inhibition of PRMT7 by gene deletion or an inhibitor treatment enhanced the activity of ß-catenin. PRMT7 deficiency decreases symmetric dimethylation of ß-catenin. Mechanistic studies reveal that methylation of arginine residue 93 in ß-catenin decreases the activity of ß-catenin. Taken together, our data suggest that PRMT7 is important for normal cardiac function through suppression of ß-catenin activity.


Assuntos
Cardiomegalia/genética , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Proteína-Arginina N-Metiltransferases/genética , beta Catenina/genética , Angiotensinas , Animais , Cardiomegalia/induzido quimicamente , Cardiomegalia/metabolismo , Fibrose , Perfilação da Expressão Gênica/métodos , Humanos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Miocárdio/patologia , Proteína-Arginina N-Metiltransferases/deficiência , RNA-Seq/métodos , Via de Sinalização Wnt/genética , beta Catenina/metabolismo
4.
Biochem Biophys Rep ; 29: 101190, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-34988296

RESUMO

Particulate matter (PM) causes several diseases, including cardiovascular diseases (CVDs). Previous studies compared the gene expression patterns in airway epithelial cells and keratinocytes exposed to PM. However, analysis of differentially expressed gene (DEGs) in endothelial cells exposed to PM2.5 (diameter less than 2.5 µm) from fossil fuel combustion has been limited. Here, we exposed human umbilical vein endothelial cells (HUVECs) to PM2.5 from combustion of gasoline, performed RNA-seq analysis, and identified DEGs. Exposure to the IC50 concentrations of gasoline engine exhaust PM2.5 (GPM) for 24 h yielded 1081 (up-regulation: 446, down-regulation: 635) DEGs. The most highly up-regulated gene is NGFR followed by ADM2 and NUPR1. The most highly down-regulated gene is TNFSF10 followed by GDF3 and EDN1. Gene Ontology enrichment analysis revealed that GPM regulated genes involved in cardiovascular system development, tube development and circulatory system development. Kyoto Encyclopedia of Genes and Genomes and Reactome pathway analyses showed that genes related to cytokine-cytokine receptor interactions and cytokine signaling in the immune system were significantly affected by GPM. We confirmed the RNA-seq data of some highly altered genes by qRT-PCR and showed the induction of NGFR, ADM2 and IL-11 at a protein level, indicating that the observed gene expression patterns were reliable. Given the adverse effects of PM2.5 on CVDs, our findings provide new insight into the importance of several DEGs and pathways in GPM-induced CVDs.

5.
Cell Death Dis ; 11(3): 203, 2020 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-32251281

RESUMO

A correction to this paper has been published and can be accessed via a link at the top of the paper.

6.
J Cachexia Sarcopenia Muscle ; 11(4): 1070-1088, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32096917

RESUMO

BACKGROUND: Muscle wasting, resulting from aging or pathological conditions, leads to reduced quality of life, increased morbidity, and increased mortality. Much research effort has been focused on the development of exercise mimetics to prevent muscle atrophy and weakness. In this study, we identified indoprofen from a screen for peroxisome proliferator-activated receptor γ coactivator α (PGC-1α) inducers and report its potential as a drug for muscle wasting. METHODS: The effects of indoprofen treatment on dexamethasone-induced atrophy in mice and in 3-phosphoinositide-dependent protein kinase-1 (PDK1)-deleted C2C12 myotubes were evaluated by immunoblotting to determine the expression levels of myosin heavy chain and anabolic-related and oxidative metabolism-related proteins. Young, old, and disuse-induced muscle atrophic mice were administered indoprofen (2 mg/kg body weight) by gavage. Body weight, muscle weight, grip strength, isometric force, and muscle histology were assessed. The expression levels of muscle mass-related and function-related proteins were analysed by immunoblotting or immunostaining. RESULTS: In young (3-month-old) and aged (22-month-old) mice, indoprofen treatment activated oxidative metabolism-related enzymes and led to increased muscle mass. Mechanistic analysis using animal models and muscle cells revealed that indoprofen treatment induced the sequential activation of AKT/p70S6 kinase (S6K) and AMP-activated protein kinase (AMPK), which in turn can augment protein synthesis and PGC-1α induction, respectively. Structural prediction analysis identified PDK1 as a target of indoprofen and, indeed, short-term treatment with indoprofen activated the PDK1/AKT/S6K pathway in muscle cells. Consistent with this finding, PDK1 inhibition abrogated indoprofen-induced AKT/S6K activation and hypertrophic response. CONCLUSIONS: Our findings demonstrate the effects of indoprofen in boosting skeletal muscle mass through the sequential activation of PDK1/AKT/S6K and AMPK/PGC-1α. Taken together, our results suggest that indoprofen represents a potential drug to prevent muscle wasting and weakness related to aging or muscle diseases.


Assuntos
Inibidores de Ciclo-Oxigenase/uso terapêutico , Indoprofen/uso terapêutico , Atrofia Muscular/tratamento farmacológico , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Inibidores de Ciclo-Oxigenase/farmacologia , Humanos , Indoprofen/farmacologia , Masculino , Camundongos
7.
Cell Death Dis ; 10(12): 903, 2019 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-31787756

RESUMO

Endoplasmic reticulum (ER) stress signaling plays a critical role in the control of cell survival or death. Persistent ER stress activates proapoptotic pathway involving the ATF4/CHOP axis. Although accumulating evidences support its important contribution to cardiovascular diseases, but its mechanism is not well characterized. Here, we demonstrate a critical role for PRMT1 in the control of ER stress in cardiomyocytes. The inhibition of PRMT1 augments tunicamycin (TN)-triggered ER stress response in cardiomyocytes while PRMT1 overexpression attenuates it. Consistently, PRMT1 null hearts show exacerbated ER stress and cell death in response to TN treatment. Interestingly, ATF4 depletion attenuates the ER stress response induced by PRMT1 inhibition. The methylation-deficient mutant of ATF4 with the switch of arginine 239 to lysine exacerbates ER stress accompanied by enhanced levels of proapoptotic cleaved Caspase3 and phosphorylated-γH2AX in response to TN. The mechanistic study shows that PRMT1 modulates the protein stability of ATF4 through methylation. Taken together, our data suggest that ATF4 methylation on arginine 239 by PRMT1 is a novel regulatory mechanism for protection of cardiomyocytes from ER stress-induced cell death.


Assuntos
Fator 4 Ativador da Transcrição/metabolismo , Estresse do Retículo Endoplasmático , Miócitos Cardíacos/metabolismo , Proteína-Arginina N-Metiltransferases/metabolismo , Fator 4 Ativador da Transcrição/química , Fator 4 Ativador da Transcrição/genética , Sequência de Aminoácidos , Animais , Animais Recém-Nascidos , Apoptose/efeitos dos fármacos , Linhagem Celular , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Humanos , Metilação/efeitos dos fármacos , Mutação/genética , Miócitos Cardíacos/efeitos dos fármacos , Especificidade de Órgãos , Ligação Proteica/efeitos dos fármacos , Proteína-Arginina N-Metiltransferases/antagonistas & inibidores , Ratos , Fator de Transcrição CHOP/metabolismo , Tunicamicina/farmacologia , Regulação para Cima/efeitos dos fármacos
8.
Nat Commun ; 9(1): 5107, 2018 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-30504773

RESUMO

Dysregulation of Ca2+/calmodulin-dependent protein kinase (CaMK)II is closely linked with myocardial hypertrophy and heart failure. However, the mechanisms that regulate CaMKII activity are incompletely understood. Here we show that protein arginine methyltransferase 1 (PRMT1) is essential for preventing cardiac CaMKII hyperactivation. Mice null for cardiac PRMT1 exhibit a rapid progression to dilated cardiomyopathy and heart failure within 2 months, accompanied by cardiomyocyte hypertrophy and fibrosis. Consistently, PRMT1 is downregulated in heart failure patients. PRMT1 depletion in isolated cardiomyocytes evokes hypertrophic responses with elevated remodeling gene expression, while PRMT1 overexpression protects against pathological responses to neurohormones. The level of active CaMKII is significantly elevated in PRMT1-deficient hearts or cardiomyocytes. PRMT1 interacts with and methylates CaMKII at arginine residues 9 and 275, leading to its inhibition. Accordingly, pharmacological inhibition of CaMKII restores contractile function in PRMT1-deficient mice. Thus, our data suggest that PRMT1 is a critical regulator of CaMKII to maintain cardiac function.


Assuntos
Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Insuficiência Cardíaca/etiologia , Insuficiência Cardíaca/metabolismo , Miocárdio/metabolismo , Proteína-Arginina N-Metiltransferases/metabolismo , Animais , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/genética , Linhagem Celular , Ecocardiografia , Eletrocardiografia , Eletrofisiologia , Insuficiência Cardíaca/genética , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Miocárdio/patologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Proteína-Arginina N-Metiltransferases/genética
9.
Cell Rep ; 22(2): 471-481, 2018 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-29320741

RESUMO

Insulin secretion from pancreatic ß cells in response to high glucose (HG) critically depends on the inhibition of KATP channel activity in HG. It is generally believed that HG-induced effects are mediated by the increase in intracellular ATP, but here, we showed that, in INS-1 cells, endocytosis of KATP channel plays a major role. Upon HG stimulation, resting membrane potential depolarized by 30.6 mV (from -69.2 to -38.6 mV) and KATP conductance decreased by 91% (from 0.243 to 0.022 nS/pF), whereas intracellular ATP was increased by only 47%. HG stimulation induced internalization of KATP channels, causing a significant decrease in surface channel density, and this decrease was completely abolished by inhibiting endocytosis using dynasore, a dynamin inhibitor, or a PKC inhibitor. These drugs profoundly inhibited HG-induced depolarization. Our results suggest that the control of KATP channel surface density plays a greater role than ATP-dependent gating in regulating ß cell excitability.


Assuntos
Endocitose/fisiologia , Glucose/metabolismo , Células Secretoras de Insulina/metabolismo , Canais de Potássio/metabolismo , Humanos
10.
Proc Natl Acad Sci U S A ; 114(8): E1345-E1354, 2017 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-28154134

RESUMO

On pathological stress, Wnt signaling is reactivated and induces genes associated with cardiac remodeling and fibrosis. We have previously shown that a cell surface receptor Cdon (cell-adhesion associated, oncogene regulated) suppresses Wnt signaling to promote neuronal differentiation however its role in heart is unknown. Here, we demonstrate a critical role of Cdon in cardiac function and remodeling. Cdon is expressed and predominantly localized at intercalated disk in both mouse and human hearts. Cdon-deficient mice develop cardiac dysfunction including reduced ejection fraction and ECG abnormalities. Cdon-/- hearts exhibit increased fibrosis and up-regulation of genes associated with cardiac remodeling and fibrosis. Electrical remodeling was demonstrated by up-regulation and mislocalization of the gap junction protein, Connexin 43 (Cx43) in Cdon-/- hearts. In agreement with altered Cx43 expression, functional analysis both using Cdon-/- cardiomyocytes and shRNA-mediated knockdown in rat cardiomyocytes shows aberrant gap junction activities. Analysis of the underlying mechanism reveals that Cdon-/- hearts exhibit hyperactive Wnt signaling as evident by ß-catenin accumulation and Axin2 up-regulation. On the other hand, the treatment of rat cardiomyocytes with a Wnt activator TWS119 reduces Cdon levels and aberrant Cx43 activities, similarly to Cdon-deficient cardiomyocytes, suggesting a negative feedback between Cdon and Wnt signaling. Finally, inhibition of Wnt/ß-catenin signaling by XAV939, IWP2 or dickkopf (DKK)1 prevented Cdon depletion-induced up-regulation of collagen 1a and Cx43. Taken together, these results demonstrate that Cdon deficiency causes hyperactive Wnt signaling leading to aberrant intercellular coupling and cardiac fibrosis. Cdon exhibits great potential as a target for the treatment of cardiac fibrosis and cardiomyopathy.


Assuntos
Moléculas de Adesão Celular/deficiência , Coração/fisiologia , Remodelação Ventricular/fisiologia , Via de Sinalização Wnt/fisiologia , beta Catenina/metabolismo , Animais , Moléculas de Adesão Celular/metabolismo , Conexina 43/metabolismo , Conexinas/metabolismo , Fibrose/metabolismo , Junções Comunicantes/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Pirimidinas/metabolismo , Pirróis/metabolismo , Ratos , Ratos Sprague-Dawley , Regulação para Cima/fisiologia
11.
Elife ; 52016 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-27466704

RESUMO

KCNQ channels are critical determinants of neuronal excitability, thus emerging as a novel target of anti-epileptic drugs. To date, the mechanisms of KCNQ channel modulation have been mostly characterized to be inhibitory via Gq-coupled receptors, Ca(2+)/CaM, and protein kinase C. Here we demonstrate that methylation of KCNQ by protein arginine methyltransferase 1 (Prmt1) positively regulates KCNQ channel activity, thereby preventing neuronal hyperexcitability. Prmt1+/- mice exhibit epileptic seizures. Methylation of KCNQ2 channels at 4 arginine residues by Prmt1 enhances PIP2 binding, and Prmt1 depletion lowers PIP2 affinity of KCNQ2 channels and thereby the channel activities. Consistently, exogenous PIP2 addition to Prmt1+/- neurons restores KCNQ currents and neuronal excitability to the WT level. Collectively, we propose that Prmt1-dependent facilitation of KCNQ-PIP2 interaction underlies the positive regulation of KCNQ activity by arginine methylation, which may serve as a key target for prevention of neuronal hyperexcitability and seizures.


Assuntos
Arginina/metabolismo , Epilepsia/fisiopatologia , Canais de Potássio KCNQ/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Processamento de Proteína Pós-Traducional , Proteína-Arginina N-Metiltransferases/metabolismo , Animais , Modelos Animais de Doenças , Metilação , Camundongos Endogâmicos C57BL , Ligação Proteica
12.
J Mol Cell Cardiol ; 93: 57-66, 2016 04.
Artigo em Inglês | MEDLINE | ID: mdl-26906632

RESUMO

Sonic hedgehog (Shh) signaling plays an important role for early heart development, such as heart looping and cardiomyogenesis of pluripotent stem cells. A multifunctional receptor Cdo functions as a Shh coreceptor together with Boc and Gas1 to activate Shh signaling and these coreceptors seem to play compensatory roles in early heart development. Thus in this study, we examined the role of Cdo in cardiomyogenesis by utilizing an in vitro differentiation of pluripotent stem cells. Here we show that Cdo is required for efficient cardiomyogenesis of pluripotent stem cells by activation of Shh signaling. Cdo is induced concurrently with Shh signaling activation upon induction of cardiomyogenesis of P19 embryonal carcinoma (EC) cells. Cdo-depleted P19 EC and Cdo(-/-) mouse embryonic stem (ES) cells display decreased expression of key cardiac regulators, including Gata4, Nkx2.5 and Mef2c and this decrease coincides with reduced Shh signaling activities. Furthermore Cdo deficiency causes a stark reduction in formation of mature contractile cardiomyocytes. This defect in cardiomyogenesis is overcome by reactivation of Shh signaling at the early specification stage of cardiomyogenesis. The Shh agonist treatment restores differentiation capacities of Cdo-deficient ES cells into contractile cardiomyocytes by recovering both the expression of early cardiac regulators and structural genes such as cardiac troponin T and Connexin 43. Therefore Cdo is required for efficient cardiomyogenesis of pluripotent stem cells and an excellent target to improve the differentiation potential of stem cells for generation of transplantable cells to treat cardiomyopathies.


Assuntos
Moléculas de Adesão Celular/genética , Coração/embriologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Organogênese , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Animais , Moléculas de Adesão Celular/metabolismo , Diferenciação Celular , Conexina 43/metabolismo , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Técnicas de Inativação de Genes , Proteínas Hedgehog/agonistas , Proteínas Hedgehog/metabolismo , Camundongos , Contração Miocárdica/genética , Organogênese/genética , Transdução de Sinais
13.
Nat Commun ; 5: 5455, 2014 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-25406935

RESUMO

Canonical Wnt signalling regulates expansion of neural progenitors and functions as a dorsalizing signal in the developing forebrain. In contrast, the multifunctional co-receptor Cdo promotes neuronal differentiation and is important for the function of the ventralizing signal, Shh. Here we show that Cdo negatively regulates Wnt signalling during neurogenesis. Wnt signalling is enhanced in Cdo-deficient cells, leading to impaired neuronal differentiation. The ectodomains of Cdo and Lrp6 interact via the Ig2 repeat of Cdo and the LDLR repeats of Lrp6, and the Cdo Ig2 repeat is necessary for Cdo-dependent Wnt inhibition. Furthermore, the Cdo-deficient dorsal forebrain displays stronger Wnt signalling activity, increased cell proliferation and enhanced expression of the dorsal markers and Wnt targets, Pax6, Gli3, Axin2. Therefore, in addition to promoting ventral central nervous system cell fates with Shh, Cdo promotes neuronal differentiation by suppression of Wnt signalling and provides a direct link between two major dorsoventral morphogenetic signalling pathways.


Assuntos
Cisteína Dioxigenase/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Neurogênese , Neurônios/metabolismo , Via de Sinalização Wnt , Animais , Proteína Axina/metabolismo , Diferenciação Celular , Embrião de Mamíferos , Proteínas do Olho/metabolismo , Proteínas Hedgehog/metabolismo , Proteínas de Homeodomínio/metabolismo , Fatores de Transcrição Kruppel-Like/metabolismo , Camundongos , Proteínas do Tecido Nervoso/metabolismo , Neurônios/citologia , Fator de Transcrição PAX6 , Fatores de Transcrição Box Pareados/metabolismo , Proteínas Repressoras/metabolismo , Proteínas Wnt/metabolismo , Proteína Gli3 com Dedos de Zinco , beta Catenina/metabolismo
14.
Stem Cell Res ; 13(2): 262-74, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25117422

RESUMO

Sonic hedgehog (Shh) signaling is required for numerous developmental processes including specification of ventral cell types in the central nervous system such as midbrain dopaminergic (DA) neurons. The multifunctional coreceptor Cdo increases the signaling activity of Shh which is crucial for development of forebrain and neural tube. In this study, we investigated the role of Cdo in midbrain DA neurogenesis. Cdo and Shh signaling components are induced during neurogenesis of embryonic stem (ES) cells. Cdo(-/-) ES cells show reduced neuronal differentiation accompanied by increased cell death upon neuronal induction. In addition, Cdo(-/-) ES cells form fewer tyrosine hydroxylase (TH) and microtubule associated protein 2 (MAP2)-positive DA neurons correlating with the decreased expression of key regulators of DA neurogenesis, such as Shh, Neurogenin2, Mash1, Foxa2, Lmx1a, Nurr1 and Pitx3, relative to the Cdo(+/+) ES cells. Consistently, the Cdo(-/-) embryonic midbrain displays a reduction in expression of TH and Nurr1. Furthermore, activation of Shh signaling by treatment with Purmorphamine (Pur) restores the DA neurogenesis of Cdo(-/-) ES cells, suggesting that Cdo is required for the full Shh signaling activation to induce efficient DA neurogenesis.


Assuntos
Moléculas de Adesão Celular/metabolismo , Neurônios Dopaminérgicos/metabolismo , Proteínas Hedgehog/metabolismo , Mesencéfalo/metabolismo , Neurogênese , Animais , Padronização Corporal , Moléculas de Adesão Celular/deficiência , Moléculas de Adesão Celular/genética , Linhagem da Célula , Técnicas de Cocultura , Neurônios Dopaminérgicos/efeitos dos fármacos , Células Alimentadoras , Regulação da Expressão Gênica no Desenvolvimento , Genótipo , Mesencéfalo/efeitos dos fármacos , Mesencéfalo/embriologia , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Camundongos Knockout , Morfolinas/farmacologia , Neurogênese/efeitos dos fármacos , Fenótipo , Purinas/farmacologia , Transdução de Sinais , Fatores de Tempo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
15.
ScientificWorldJournal ; 2014: 592450, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24600331

RESUMO

The p104 protein inhibits cellular proliferation when overexpressed in NIH3T3 cells and has been shown to associate with p85α, Grb2, and PLCγ1. In order to isolate other proteins that interact with p104, yeast two-hybrid screening was performed. Rac1 was identified as a binding partner of p104 and the interaction between p104 and Rac1 was confirmed by immunoprecipitation. Using a glutathione S-transferase (GST) pull-down assay with various p104 fragments, the 814-848 amino acid residue at the carboxyl-terminal region of p104 was identified as the key component to interact with Rac1. The CrkII which is involved in the Rac1-mediated cellular response was also found to interact with p104 protein. NIH3T3 cells which overexpressed p104 showed a decrease of Rac1 activity. However, neither the proline-rich domain mutant, which is unable to interact with CrkII, nor the carboxy-terminal deletion mutant could attenuate Rac1 activity. During the differentiation of myoblasts, the amount of p104 protein as well as transcript level was increased. The overexpression of p104 enhanced myotube differentiation, whereas siRNA of p104 reversed this process. In this process, more Rac1 and CrkII were bound to increased p104. Based on these results, we conclude that p104 is involved in muscle cell differentiation by modulating the Rac1 activity.


Assuntos
Proteínas de Transporte/metabolismo , Diferenciação Celular , Mioblastos/metabolismo , Neuropeptídeos/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Animais , Sítios de Ligação , Proteínas de Transporte/química , Proteínas de Transporte/genética , Deleção de Genes , Camundongos , Mioblastos/citologia , Células NIH 3T3 , Ligação Proteica , Proteínas Proto-Oncogênicas c-crk/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
16.
Proc Natl Acad Sci U S A ; 111(5): 1778-83, 2014 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-24434556

RESUMO

The potential for pluripotent cells to differentiate into diverse specialized cell types has given much hope to the field of regenerative medicine. Nevertheless, the low efficiency of cell commitment has been a major bottleneck in this field. Here we provide a strategy to enhance the efficiency of early differentiation of pluripotent cells. We hypothesized that the initial phase of differentiation can be enhanced if the transcriptional activity of master regulators of stemness is suppressed, blocking the formation of functional transcriptomes. However, an obstacle is the lack of an efficient strategy to block protein-protein interactions. In this work, we take advantage of the biochemical property of seventeen kilodalton protein (Skp), a bacterial molecular chaperone that binds directly to sex determining region Y-box 2 (Sox2). The small angle X-ray scattering analyses provided a low resolution model of the complex and suggested that the transactivation domain of Sox2 is probably wrapped in a cleft on Skp trimer. Upon the transduction of Skp into pluripotent cells, the transcriptional activity of Sox2 was inhibited and the expression of Sox2 and octamer-binding transcription factor 4 was reduced, which resulted in the expression of early differentiation markers and appearance of early neuronal and cardiac progenitors. These results suggest that the initial stage of differentiation can be accelerated by inhibiting master transcription factors of stemness. This strategy can possibly be applied to increase the efficiency of stem cell differentiation into various cell types and also provides a clue to understanding the mechanism of early differentiation.


Assuntos
Diferenciação Celular , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Fatores de Transcrição/metabolismo , Animais , Proteínas de Escherichia coli/metabolismo , Camundongos , Modelos Biológicos , Modelos Moleculares , Fatores de Transcrição SOXB1/metabolismo , Espalhamento a Baixo Ângulo , Soluções , Transdução Genética , Difração de Raios X , Produtos do Gene tat do Vírus da Imunodeficiência Humana/metabolismo
17.
Biochem Biophys Res Commun ; 425(2): 250-5, 2012 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-22828506

RESUMO

Unlike epidermal growth factor-like protein 7 (EGFL7), which is a secreted protein implicated in the regulation of blood vessel formation and cell migration, little is known about the physiological function of EGFL8. Thymic epithelial cells (TECs) play a pivotal role in T-cell development by regulating cellular interactions and expression of growth factors, cytokines, and chemokines. In order to investigate the functional role of EGFL8 in TECs, we transfected TECs with an EGFL8-expressing vector to overexpress EGFL8 protein and with an EGFL8 siRNA to knockdown EGFL8 expression. EGFL8-silenced TECs showed significant increase in the number of adherent thymocytes by enhancing the expression of intercellular adhesion molecule-1 (ICAM-1), while the overexpression of EGFL8 inhibited the adherence of TECs to thymocytes by suppressing ICAM-1 expression. Furthermore, in vitro co-culture study revealed that knockdown of EGFL8 facilitated the maturation of thymocytes to CD4(+) and CD8(+) single-positive populations. These regulatory effects of EGFL8 in T-cell development were further confirmed by the results that knockdown of EGFL8 enhanced the expression of genes involved in thymopoiesis, such as interleukin-7 (IL-7), granulocyte/macrophage-colony stimulating factor (GM-CSF), and thymus-expressed chemokine (TECK). Our data show that EGFL8 exerts inhibitory effects on TECs and thymocytes, suggesting that EGFL8 acts as a negative regulatory molecule in the development of T cells in the mouse thymus.


Assuntos
Proteínas/fisiologia , Linfócitos T/citologia , Timo/citologia , Animais , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/metabolismo , Proteínas de Ligação ao Cálcio , Adesão Celular , Linhagem Celular , Família de Proteínas EGF , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Técnicas de Silenciamento de Genes , Camundongos , Camundongos Endogâmicos C57BL , Proteínas/genética , Linfócitos T/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...