Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Med Sci Monit ; 26: e922561, 2020 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-32594094

RESUMO

BACKGROUND This study aimed to investigate the effects of the paeonol-platinum(II) (PL-Pt[II]) complex on SW1736 human anaplastic thyroid carcinoma cell line and the BHP7-13 human thyroid papillary carcinoma cell line in vitro and on mouse SW1736 tumor xenografts in vivo. MATERIAL AND METHODS The cytotoxic effects of the PL-Pt(II) complex on SW1736 cells and BHP7-13 cells was measured using the MTT assay. Western blot measured the expression levels of cyclins, cell apoptotic proteins, and signaling proteins. DNA content and apoptosis were detected by flow cytometry. SW1736 cell thyroid tumor xenografts were established in mice followed by treatment with the PL-Pt(II) complex. RESULTS Treatment of the SW1736 and BHP7-13 cells with the PL-Pt(II) complex reduced cell proliferation in a dose-dependent manner, with an IC50 of 1.25 µM and 1.0 µM, respectively, and increased the cell fraction in G0/G1phase, inhibited p53, cyclin D1, promoted p27 and p21 expression, and significantly increased the sub-G1 fraction. Treatment with the PL-Pt(II) complex increased caspase-3 degradation, reduced the expression of p-4EBP1, p-4E-BP1 and p-S6, and reduced the expression of p-ERK1/2 and p-AKT. Treatment with the PL-Pt(II) complex reduced the volume of the SW1736 mouse tumor xenografts on day 14 and day 21, and reduced AKT phosphorylation and S6 protein expression and increased degradation of caspase-3. CONCLUSIONS The cytotoxic effects of the PL-Pt(II) complex in human thyroid carcinoma cells, including activation of apoptosis and an increased sub-G1 cell fraction of the cell cycle, were mediated by down-regulation of the mTOR pathway.


Assuntos
Acetofenonas/farmacologia , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Compostos de Platina/farmacologia , Serina-Treonina Quinases TOR/efeitos dos fármacos , Câncer Papilífero da Tireoide/genética , Carcinoma Anaplásico da Tireoide/genética , Neoplasias da Glândula Tireoide/genética , Proteínas Adaptadoras de Transdução de Sinal/efeitos dos fármacos , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Apoptose/genética , Western Blotting , Caspase 3/efeitos dos fármacos , Caspase 3/metabolismo , Ciclo Celular/genética , Proteínas de Ciclo Celular/efeitos dos fármacos , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/genética , Ciclina D1/efeitos dos fármacos , Ciclina D1/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/efeitos dos fármacos , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Inibidor de Quinase Dependente de Ciclina p27/efeitos dos fármacos , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Regulação para Baixo , Humanos , Técnicas In Vitro , Camundongos , Proteína Quinase 1 Ativada por Mitógeno/efeitos dos fármacos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/efeitos dos fármacos , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Transplante de Neoplasias , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Quinases S6 Ribossômicas/efeitos dos fármacos , Proteínas Quinases S6 Ribossômicas/metabolismo , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo , Câncer Papilífero da Tireoide/metabolismo , Carcinoma Anaplásico da Tireoide/metabolismo , Neoplasias da Glândula Tireoide/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Exp Neurol ; 333: 113398, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32659382

RESUMO

We investigated the ability of agmatine to potentiate the antidepressant-like and synaptic effects of ketamine in mice. Agmatine (0.1 and 1 mg/kg, p.o.) and ketamine (1 and 10 mg/kg, i.p.) produced an antidepressant-like effect in the tail suspension test. The combination of agmatine (0.01 mg/kg, p.o.) and ketamine (0.1 mg/kg, i.p.), at subthreshold doses, produced an antidepressant-like effect 1 h, 24 h and 7d after treatment. Western blot analysis from prefrontal cortex tissue showed that the combined treatment, after 1 h, increased p70S6K and GluA1, and reduced synapsin 1 phosphorylation. Additionally, after 24 h, Akt, p70S6K, GluA1, and synapsin 1 phosphorylation; and PSD95 immunocontent increased (which persisted for up to 7d). Dendritic architecture analysis of the prefrontal cortex revealed that the combined treatment improved dendritic arbor complexity (after 24 h, up to 7d), and increased spine density (after 1 h, up to 24 h). Morphometric analysis revealed a filopodia-shaped dendrite spine upregulation after 1 h. A predominance of stubby, mushroom, branched and filopodia; and a reduction in thin protrusions were observed after 24 h. Finally, mushroom-shaped dendritic spines predominance increased after 7d. Agmatine potentiated ketamine's antidepressant, and dendritic arbors and spines remodeling effects in a time-dependent manner. Our data indicate Akt/p70S6K signaling as a likely target for these effects.


Assuntos
Agmatina/farmacologia , Antidepressivos/farmacologia , Dendritos/efeitos dos fármacos , Espinhas Dendríticas/efeitos dos fármacos , Ketamina/farmacologia , Proteína Oncogênica v-akt/efeitos dos fármacos , Proteínas Quinases S6 Ribossômicas/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Sinapses/efeitos dos fármacos , Animais , Dendritos/ultraestrutura , Espinhas Dendríticas/ultraestrutura , Sinergismo Farmacológico , Elevação dos Membros Posteriores , Masculino , Camundongos , Atividade Motora/efeitos dos fármacos , Córtex Pré-Frontal/efeitos dos fármacos
3.
Neuropharmacology ; 130: 62-70, 2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29191753

RESUMO

Melanin-Concentrating Hormone (MCH) is one of the most relevant orexigenic factors specifically located in the lateral hypothalamic area (LHA), with its physiological relevance demonstrated in studies using several genetically manipulated mice models. However, the central mechanisms controlling MCH-induced hyperphagia remain largely uncharacterized. Here, we show that central injection of MCH in mice deficient for kappa opoid receptor (k-OR) failed to stimulate feeding. To determine the hypothalamic area responsible for this MCH/k-OR interaction, we performed virogenetic studies and found that downregulation of k-OR by adeno-associated viruses (shOprk1-AAV) in LHA, but not in other hypothalamic nuclei, was sufficient to block MCH-induced food intake. Next, we sought to investigate the molecular signaling pathway within the LHA that mediates acute central MCH stimulation of food intake. We found that MCH activates k-OR and that increased levels of phosphorylated extracellular signal regulated kinase (ERK) are associated with downregulation of phospho-S6 Ribosomal Protein. This effect was prevented when a pharmacological inhibitor of k-OR was co-administered with MCH. Finally, the specific activation of the direct upstream regulator of S6 (p70S6K) in the LHA attenuated MCH-stimulated food consumption. Our results reveal that lateral hypothalamic k-OR system modulates the orexigenic action of MCH via the p70S6K/S6 pathway.


Assuntos
Ingestão de Alimentos/efeitos dos fármacos , Hormônios Hipotalâmicos/administração & dosagem , Melaninas/administração & dosagem , Hormônios Hipofisários/administração & dosagem , Receptores Opioides kappa/antagonistas & inibidores , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Animais , Depressores do Apetite/administração & dosagem , Depressores do Apetite/metabolismo , Dependovirus , Região Hipotalâmica Lateral/efeitos dos fármacos , Região Hipotalâmica Lateral/metabolismo , Hormônios Hipotalâmicos/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Melaninas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Hormônios Hipofisários/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores Opioides kappa/metabolismo , Proteínas Quinases S6 Ribossômicas/efeitos dos fármacos , Proteínas Quinases S6 Ribossômicas/metabolismo
4.
J Orthop Res ; 25(7): 933-40, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17427956

RESUMO

Pulsed electromagnetic field (PEMF) devices are approved for the healing of bone nonunions, but there is a lack of understanding as to their mechanism of action at the cell and molecular level. Intermittent parathyroid hormone (PTH) therapy is currently utilized for treatment of osteoporosis, and is also being investigated for the purpose of augmenting fracture healing. Insulin and IGF-1 are also thought to play important anabolic roles in osteogenesis. In this report, signaling pathways activated by acute PTH or insulin treatments were compared to those activated by PEMF treatment in osteoblast-like cells. Some signaling molecules like the extracellular response kinases 1/2 (Erk1/2) and the cAMP response element binding protein (CREB) were activated by insulin and PTH, respectively, but not by PEMF treatment. Other signaling molecules like the insulin receptor substrate-1 (IRS-1), the S6 ribosomal subunit kinase, and the endothelial nitric oxide synthase (eNOS) were phosphorylated by PTH, insulin, and PEMF to the same relative extent and within the same time frame. IRS-1, eNOS, and S6 have been implicated in bone anabolism, and our results suggest that the anabolic effects of PEMF may be mediated, in part, through the activation of these proteins.


Assuntos
Anabolizantes/farmacologia , Insulina/farmacologia , Osteoblastos/efeitos da radiação , Hormônio Paratireóideo/farmacologia , Radiação , Transdução de Sinais/efeitos da radiação , Animais , Proteína de Ligação a CREB/efeitos dos fármacos , Proteína de Ligação a CREB/metabolismo , Proteína de Ligação a CREB/efeitos da radiação , Linhagem Celular , Proteínas Substratos do Receptor de Insulina , Proteína Quinase 3 Ativada por Mitógeno/efeitos dos fármacos , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/efeitos da radiação , Óxido Nítrico Sintase Tipo III/efeitos dos fármacos , Óxido Nítrico Sintase Tipo III/metabolismo , Óxido Nítrico Sintase Tipo III/efeitos da radiação , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Fosfoproteínas/efeitos dos fármacos , Fosfoproteínas/metabolismo , Fosfoproteínas/efeitos da radiação , Fosforilação , Radioterapia , Ratos , Ratos Sprague-Dawley , Proteínas Quinases S6 Ribossômicas/efeitos dos fármacos , Proteínas Quinases S6 Ribossômicas/metabolismo , Proteínas Quinases S6 Ribossômicas/efeitos da radiação , Transdução de Sinais/efeitos dos fármacos
5.
Mol Cancer Ther ; 5(8): 2051-9, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16928826

RESUMO

Signaling through the receptor for epidermal growth factor receptor (EGFR) is frequently deregulated in solid tumors. Erlotinib (Tarceva, OSI-774, OSI Pharmaceuticals, Inc., Melville, NY) is a low molecular weight, orally bioavailable inhibitor of the EGFR that has been approved for both non-small cell lung cancer and pancreatic cancers. Previous studies have indicated that sensitivity to EGFR antagonists correlated with HER-3 signaling for non-small cell lung cancer. Herein, we have sought to understand the signaling pathways that mediate erlotinib sensitivity for pancreatic and colorectal cancers. In a panel of 12 pancreatic tumor cell lines, we find that EGFR is coexpressed with HER-3 in all cell lines sensitive to erlotinib but not in insensitive cell lines. Erlotinib can block HER-3 phosphorylation in these sensitive cell lines, suggesting that HER-3 is transactivated by EGFR. Knockdown of HER-3 in BxPC3, an erlotinib-sensitive pancreatic tumor cell line, results in inhibition of the phosphorylation for both Akt and S6 and is associated with a decrease in cell proliferation and reduced sensitivity to erlotinib. Therefore, EGFR transactivation of HER-3 mediates Akt signaling and can contribute to erlotinib sensitivity for pancreatic tumors. We extended our analysis to a panel of 13 colorectal tumor cell lines and find that, like pancreatic, HER-3 is coexpressed with EGFR in the most erlotinib-sensitive cell lines but not in erlotinib-insensitive cell lines. These studies suggest that HER-3 could be used as a biomarker to select patients who are most likely to respond to erlotinib therapy.


Assuntos
Neoplasias Colorretais/tratamento farmacológico , Neoplasias Pancreáticas/tratamento farmacológico , Proteínas Proto-Oncogênicas c-akt/efeitos dos fármacos , Quinazolinas/farmacologia , Receptor ErbB-3/metabolismo , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Linhagem Celular Tumoral , Neoplasias Colorretais/metabolismo , Relação Dose-Resposta a Droga , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/metabolismo , Cloridrato de Erlotinib , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/metabolismo , Neoplasias Pancreáticas/metabolismo , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Proteínas Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor ErbB-3/efeitos dos fármacos , Proteínas Quinases S6 Ribossômicas/efeitos dos fármacos , Proteínas Quinases S6 Ribossômicas/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR
6.
Circ Res ; 88(5): 513-9, 2001 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-11249875

RESUMO

Glucose-insulin-potassium solutions exert beneficial effects on the ischemic heart by reducing infarct size and mortality and improving postischemic left ventricular function. Insulin could be the critical protective component of this mixture, although the insulin response of the ischemic and postischemic myocardium has not been systematically investigated. The aim of this work was to study the insulin response during ischemia by analyzing insulin signaling. This was evaluated by measuring changes in activity and/or phosphorylation state of insulin signaling elements in isolated perfused rat hearts submitted to no-flow ischemia. Intracellular pH (pH(i)) was measured by NMR. No-flow ischemia antagonized insulin signaling including insulin receptor, insulin receptor substrate-1, phosphatidylinositol 3-kinase, protein kinase B, p70 ribosomal S6 kinase, and glycogen synthase kinase-3. These changes were concomitant with intracellular acidosis. Perfusing hearts with ouabain and amiloride in normoxic conditions decreased pH(i) and insulin signaling, whereas perfusing at pH 8.2 counteracted the drop in pH(i) and the inhibition of insulin signaling by ischemia. Incubation of cardiomyocytes in normoxic conditions, but at pH values below 6.75, mimicked the effect of ischemia and also inhibited insulin-stimulated glucose uptake. Finally, the in vitro insulin receptor tyrosine kinase activity was progressively inhibited at pH values below physiological pH(i), being abolished at pH 6.0. Therefore, ischemic acidosis decreases kinase activity and tyrosine phosphorylation of the insulin receptor thereby preventing activation of the downstream components of the signaling pathway. We conclude that severe ischemia inhibits insulin signaling by decreasing pH(i).


Assuntos
Coração/efeitos dos fármacos , Insulina/farmacologia , Isquemia Miocárdica/fisiopatologia , Proteínas Serina-Treonina Quinases , Animais , Proteínas Quinases Dependentes de Cálcio-Calmodulina/efeitos dos fármacos , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Relação Dose-Resposta a Droga , Ativação Enzimática/efeitos dos fármacos , Quinase 3 da Glicogênio Sintase , Quinases da Glicogênio Sintase , Coração/fisiologia , Concentração de Íons de Hidrogênio , Proteínas Substratos do Receptor de Insulina , Masculino , Reperfusão Miocárdica , Miocárdio/citologia , Miocárdio/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosfoproteínas/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Ratos , Ratos Wistar , Receptor de Insulina/metabolismo , Proteínas Quinases S6 Ribossômicas/efeitos dos fármacos , Proteínas Quinases S6 Ribossômicas/metabolismo , Transdução de Sinais
7.
Circ Res ; 89(6): 496-502, 2001 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-11557736

RESUMO

Sphingosine 1-phosphate (S1P), a platelet-derived ligand for the EDG-1 family of G protein-coupled receptors (GPCRs), has recently emerged as a regulator of vascular development. Although S1P has potent effects on endothelial cells and vascular smooth muscle cells (VSMCs), the functions of the specific S1P receptors in the latter cell type are not known. Here we show that pup-intimal VSMCs express higher levels of EDG-1 mRNA than adult-medial VSMCs. Stable transfection of EDG-1 into adult-medial VSMCs enhanced their proliferative response to S1P, concomitant with induction of p70 S6 kinase activity and expression of cyclin D1. Pertussis toxin treatment inhibited S1P-induced p70 S6 kinase activation, cyclin D1 expression and proliferation, suggesting that EDG-1-coupling to the G(i) pathway is critical. Furthermore, blocking p70 S6 kinase phosphorylation with rapamycin inhibited cyclin D1 expression and proliferation, suggesting that activation of p70 S6 kinase is critical in EDG-1/G(i)-mediated cell proliferation. EDG-1 expression also profoundly enhanced the migratory response of adult-medial VSMCs to S1P. S1P-induced migration of adult-medial VSMCs expressing exogenous EDG-1 required G(i) activation but not p70 S6 kinase. These results suggest that enhanced expression of EDG-1 in VSMCs dramatically stimulates both the proliferative and migratory responses to S1P. Since EDG-1 is expressed in the pup-intimal phenotype of VSMCs, S1P signaling via EDG-1 may play a role in vascular diseases in which the proliferation and migration of VSMCs are dysregulated.


Assuntos
Proteínas Imediatamente Precoces/fisiologia , Lisofosfolipídeos , Músculo Liso Vascular/citologia , Receptores de Superfície Celular , Receptores Acoplados a Proteínas G , Animais , Animais Recém-Nascidos , Divisão Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Meios de Cultura Livres de Soro/farmacologia , Ciclina D1/efeitos dos fármacos , Ciclina D1/metabolismo , DNA/biossíntese , DNA/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ativação Enzimática/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas Imediatamente Precoces/genética , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Toxina Pertussis , Isoformas de Proteínas/genética , RNA Mensageiro/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Receptores de Lisofosfolipídeos , Proteínas Quinases S6 Ribossômicas/efeitos dos fármacos , Proteínas Quinases S6 Ribossômicas/metabolismo , Sirolimo/farmacologia , Esfingosina/análogos & derivados , Esfingosina/farmacologia , Transfecção , Fatores de Virulência de Bordetella/farmacologia
8.
J Neurosci ; 21(2): 382-91, 2001 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-11160419

RESUMO

The identification of tags that can specifically mark activated synapses is important for understanding how long-term synaptic changes can be restricted to specific synapses. The maintenance of synapse-specific facilitation in Aplysia sensory to motor neuron cultures can be blocked by inhibitors of translation and by the drug rapamycin, which specifically blocks a signaling pathway that regulates phosphorylation of translational regulators. One important target of rapamycin is the phosphorylation and subsequent activation of S6 kinase. To test whether S6 kinase is the target for the ability of rapamycin to block synapse-specific facilitation in Aplysia, we cloned Aplysia S6 kinase, its substrate S6, and the S6 kinase kinase phosphoinositide-dependent kinase 1 (PDK-1). Serotonin, which induces synapse-specific facilitation, increased phosphorylation of Aplysia S6 kinase at threonine 399 in a rapamycin-sensitive manner in Aplysia synaptosomes. The phosphorylation of threonine 399 by 5-HT was independent of phosphoinositide-3 kinase, dependent on PKA and PKC, and occluded by the phosphatase inhibitor calyculin-A. 5-HT also increased S6 kinase activity and led to increased phosphorylation of S6 in synaptosomes. 5-HT increased levels of S6 in synaptosomes because of a rapamycin-sensitive increase in translation-stabilization of S6. Aplysia PDK-1 bound to and phosphorylated Aplysia S6 kinase but only modulated phosphorylation of threonine 399 indirectly. These results suggest a mechanism by which the levels of translation factors can be increased specifically at activated synapses generating a long-lasting synaptic tag.


Assuntos
Proteínas Quinases S6 Ribossômicas/metabolismo , Serotonina/metabolismo , Sirolimo/farmacologia , Sinaptossomos/enzimologia , Proteínas Quinases Dependentes de 3-Fosfoinositídeo , Animais , Antibacterianos/farmacologia , Aplysia , Sítios de Ligação/efeitos dos fármacos , Linhagem Celular , Inibidores Enzimáticos/farmacologia , Potenciação de Longa Duração/efeitos dos fármacos , Toxinas Marinhas , Dados de Sequência Molecular , Plasticidade Neuronal/efeitos dos fármacos , Oxazóis/farmacologia , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteína S6 Ribossômica , Proteínas Quinases S6 Ribossômicas/efeitos dos fármacos , Proteínas Quinases S6 Ribossômicas/genética , Proteínas Ribossômicas/genética , Proteínas Ribossômicas/metabolismo , Serotonina/farmacologia , Spodoptera , Sinapses/metabolismo , Sinaptossomos/efeitos dos fármacos , Treonina/metabolismo
9.
Cell Death Differ ; 8(8): 841-9, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11526437

RESUMO

Treatment of Swiss 3T3 cells with staurosporine resulted in dephosphorylation of two proteins which play key roles in regulating mRNA translation. This occurred before the execution of apoptosis, assessed by caspase-3 activity. These translation regulators are p70 S6 kinase, which phosphorylates ribosomal protein S6, and eukaryotic initiation factor (eIF) 4E binding protein 1 (4E-BP1), which both lie downstream of the mammalian target of rapamycin (mTOR). This resulted in decreased p70 S6 kinase activity, dephosphorylation of ribosomal protein S6, increased binding of 4E-BP1 to eIF4E and a concomitant decrease in eIF4F complexes. Our data show that staurosporine impairs mTOR signalling in vivo but that this not due to direct inhibition of mTOR or to inhibition of protein kinase C. It is becoming clear that agents which cause apoptosis inactivate mTOR signalling as a common early response prior to the execution of apoptosis, i.e., before caspase activation.


Assuntos
Apoptose/genética , Inibidores Enzimáticos/farmacologia , Biossíntese de Proteínas/fisiologia , Proteínas Quinases/metabolismo , RNA Mensageiro/metabolismo , Estaurosporina/farmacologia , Células 3T3 , Proteínas Adaptadoras de Transdução de Sinal , Animais , Apoptose/efeitos dos fármacos , Proteínas de Transporte/efeitos dos fármacos , Proteínas de Transporte/metabolismo , Caspases/efeitos dos fármacos , Caspases/metabolismo , Proteínas de Ciclo Celular , Fator de Iniciação 2 em Eucariotos/efeitos dos fármacos , Fator de Iniciação 2 em Eucariotos/metabolismo , Fatores de Iniciação em Eucariotos , Insulina/farmacologia , Camundongos , Fosfoproteínas/efeitos dos fármacos , Fosfoproteínas/metabolismo , Fosforilação/efeitos dos fármacos , Biossíntese de Proteínas/efeitos dos fármacos , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/metabolismo , Proteínas Quinases/efeitos dos fármacos , Proteínas/antagonistas & inibidores , RNA Mensageiro/efeitos dos fármacos , Proteínas Quinases S6 Ribossômicas/efeitos dos fármacos , Proteínas Quinases S6 Ribossômicas/metabolismo , Sirolimo/farmacologia , Serina-Treonina Quinases TOR
10.
FASEB J ; 15(13): 2539-41, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11641267

RESUMO

Mitogenic stimulation by growth factors may be mediated through intracellular reactive oxygen species (ROS) acting as signaling molecules. Incubation of multicellular prostate tumor spheroids with adenosine 5' triphosphate (ATP) dose-dependently stimulated tumor growth. ATP, uridine 5'-triphosphate (UTP), adenosine 5'-diphosphate (ADP), and 2-methylthio-ATP (2-MeS-ATP) increased intracellular ROS levels significantly. ROS generation by ATP was inhibited by the P2 receptor antagonist suramin, by the reduced nicotinamide adenine dinucleotide phosphate (NADPH) oxidase inhibitors diphenylene iodonium chloride (DPI) and 4-(2-aminoethyl) benzenesulfonylfluoride (AEBSF), as well as by the Ca2+-dependent phospholipase A2 (PLA2) inhibitors indomethacin and methyl arachidonyl fluorophosphonate (MAFP). The generation of ROS was dependent on the intracellular Ca2+ response evoked by ATP. Exogenous ATP activated the extracellular signal-regulated kinase 1/2 (ERK1/2) mitogen-activated protein kinase (MAPK) pathway, which was blunted by the MAPK/ERK kinase 1/2 (MEK1/2) antagonist PD98059. The radical scavengers vitamin E, dimethyl thiourea (DMTU), and N-acetyl cysteine (NAC) failed to inhibit ERK1/2 activation but abolished p90 ribosomal S6 kinase (p90RSK) activation downstream of ERK1/2, as well as the growth stimulation of tumor spheroids. Our data indicate that p90RSK downstream of ERK1/2 is the molecular target for ROS generated through stimulation of purinergic receptors by ATP.


Assuntos
Trifosfato de Adenosina/farmacologia , Agonistas Purinérgicos , Espécies Reativas de Oxigênio/metabolismo , Proteínas Quinases S6 Ribossômicas/efeitos dos fármacos , Esferoides Celulares/efeitos dos fármacos , Tioureia/análogos & derivados , Acetilcisteína/farmacologia , Cálcio/farmacologia , Divisão Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ativação Enzimática/efeitos dos fármacos , Flavonoides/farmacologia , Sequestradores de Radicais Livres/farmacologia , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Proteínas Quinases Ativadas por Mitógeno/metabolismo , NADPH Oxidases/metabolismo , Fosfolipases A/metabolismo , Fosfolipases A2 , Fosforilação/efeitos dos fármacos , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Receptores Purinérgicos/fisiologia , Proteínas Quinases S6 Ribossômicas/metabolismo , Esferoides Celulares/metabolismo , Esferoides Celulares/patologia , Tioureia/farmacologia , Células Tumorais Cultivadas , Vitamina E/farmacologia
11.
FEBS Lett ; 550(1-3): 94-100, 2003 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-12935893

RESUMO

The molecular mechanisms that govern cell movement are the subject of intense study, as they impact biologically and medically important processes such as leukocyte chemotaxis and angiogenesis, among others. We demonstrate that leukocyte chemotaxis is prevented by the macrolide immunosuppressant rapamycin, a specific inhibitor of the mammalian target of rapamycin (mTOR)/ribosomal p70-S6 kinase (p70S6K) pathway. Both neutrophil chemotaxis and chemokinesis elicited by granulocyte-macrophage colony-stimulating factor (GM-CSF) were strongly inhibited by rapamycin with an IC(50) of 0.3 nM. Inhibition, although at a higher dose, was also observed when the chemoattractant was interleukin-8. As for the mechanism, rapamycin targeted the increase of phosphorylation of p70S6K due to GM-CSF treatment, as demonstrated with specific anti-p70S6K immunoprecipitation and subsequent immunoblotting with anti-T(421)/S(424) antibodies. Rapamycin also inhibited GM-CSF-induced actin polymerization, a hallmark of leukocyte migration. The specificity of the effect of rapamycin was confirmed by the use of the structural analog FK506, which did not have a significant effect on chemotaxis but effectively rescued rapamycin-induced p70S6K inhibition. This was expected from a competitive effect of both molecules on FK506-binding proteins (FKBP). Additionally, GM-CSF-induced chemotaxis was completely (>90%) blocked by a combination of rapamycin and the MAPK kinase (MEK) inhibitor PD-98059. In summary, the results presented here indicate for the first time that rapamycin, at sub-nanomolar concentrations, inhibits GM-CSF-induced chemotaxis and chemokinesis. This serves to underscore the relevance of the mTOR/S6K pathway in neutrophil migration.


Assuntos
Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , MAP Quinase Quinase Quinase 1 , Neutrófilos/efeitos dos fármacos , Sirolimo/farmacologia , Actinas/efeitos dos fármacos , Actinas/metabolismo , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Quimiotaxia/efeitos dos fármacos , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Flavonoides/farmacologia , Humanos , Concentração Inibidora 50 , Neutrófilos/citologia , Fosforilação/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/efeitos dos fármacos , Proteínas Quinases S6 Ribossômicas/efeitos dos fármacos , Proteínas Quinases S6 Ribossômicas/metabolismo , Tacrolimo/farmacologia
12.
Biochem Pharmacol ; 59(9): 1163-71, 2000 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-10704947

RESUMO

Arginine vasopressin (AVP) and lysophosphatidic acid (LPA) have been shown to stimulate protein kinase C (PKC) and mitogen-activated protein (MAP) kinases and the proliferation of vascular smooth muscle cells. However, the actions of these two agents in cardiomyocytes are less well understood. To investigate the signal transduction pathways of AVP and LPA, freshly isolated adult rat cardiomyocytes were examined. Both AVP and LPA induced concentration- and time-dependent stimulation of the phosphotransferase activities of p90 ribosomal S6 kinases (RSK) and their upstream activators, extracellularly regulated kinases (ERK) 1 and 2. The activation of ERK1 and ERK2 by LPA was PKC- and phosphatidylinositol 3-kinase (PI 3-kinase)-dependent. However, AVP-induced activation of RSK2, a downstream substrate of ERK1 and ERK2, was PKC-dependent and PI 3-kinase-independent. AVP and LPA were also observed to increase the phosphotransferase activity of p70 ribosomal protein S6 kinase (p70 S6K) in a time- and concentration-dependent manner. The activation of p70 S6K by LPA and AVP was PI 3-kinase-dependent. PKC was necessary in AVP- but not in LPA-induced activation of p70 S6K. Since RSK and p70 S6K have been implicated in the regulation of translational control of protein synthesis, we concluded that AVP and LPA may stimulate the growth of cardiomyocytes through these two protein kinase cascades.


Assuntos
Arginina Vasopressina/farmacologia , Coração/efeitos dos fármacos , Lisofosfolipídeos/farmacologia , Miocárdio/enzimologia , Proteínas Quinases S6 Ribossômicas/metabolismo , Animais , Ativação Enzimática/efeitos dos fármacos , Técnicas In Vitro , Masculino , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Peso Molecular , Miocárdio/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Ratos , Ratos Sprague-Dawley , Proteínas Quinases S6 Ribossômicas/efeitos dos fármacos
13.
Eur J Pharmacol ; 427(3): 175-85, 2001 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-11567647

RESUMO

Immunosuppressants are now known to modulate bone metabolism, including bone formation and resorption. Because cartilage, formed by differentiated chondrocytes, serves as a template for endochondral bone formation, we examined the effects of the immunosuppressant rapamycin on the chondrogenesis of mesenchymal cells and on the cell signaling that is required for chondrogenesis, such as protein kinase C, extracellular signal-regulated kinase-1 (ERK-1), and p38 mitogen-activated protein (MAP) kinase pathways. Rapamycin inhibited the expression of type II collagen and the accumulation of sulfate glycosaminoglycan, indicating inhibition of the chondrogenesis of mesenchymal cells. Rapamycin treatment did not affect precartilage condensation, but it prevented cartilage nodule formation. Exposure of chondrifying mesenchymal cells to rapamycin blocked activation of the protein kinase C alpha and p38 MAP kinase, but had no discernible effect on ERK-1 signaling. Selective inhibition of PKCalpha or p38 MAP kinase activity, which is dramatically increased during chondrogenesis, with specific inhibitors in the absence of rapamycin blocked the chondrogenic differentiation of mesenchymal cells. Taken together, our data indicate that the immunosuppressant rapamycin inhibits the chondrogenesis of mesenchymal cells at the post-precartilage condensation stage by modulating signaling pathways including those of PKCalpha and p38 MAP kinase.


Assuntos
Condrogênese/efeitos dos fármacos , Imunossupressores/farmacologia , Isoenzimas/antagonistas & inibidores , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Proteína Quinase C/antagonistas & inibidores , Sirolimo/farmacologia , Animais , Cartilagem/efeitos dos fármacos , Cartilagem/embriologia , Técnicas de Cultura de Células/métodos , Divisão Celular/efeitos dos fármacos , Células Cultivadas , Embrião de Galinha , Ativação Enzimática/efeitos dos fármacos , Isoenzimas/metabolismo , Mesoderma/citologia , Mesoderma/efeitos dos fármacos , Mesoderma/enzimologia , Proteína Quinase 3 Ativada por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/efeitos dos fármacos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosforilação/efeitos dos fármacos , Proteína Quinase C/metabolismo , Proteína Quinase C-alfa , Proteínas Quinases S6 Ribossômicas/efeitos dos fármacos , Proteínas Quinases S6 Ribossômicas/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno
14.
Neurosci Lett ; 259(3): 137-40, 1999 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-10025577

RESUMO

Normal adult rat chromaffin cells show a robust proliferative response in vitro to nerve growth factor (NGF) and other mitogens. In contrast, PC12 rat pheochromocytoma cells proliferate in the absence of exogenous mitogens and undergo neuronal differentiation in response to NGF. We demonstrate in this work that the antiproliferative drug rapamycin suppresses normal chromaffin cell proliferation. This effect is blocked by FK 506, indicating that it occurs via interaction of rapamycin with its intracellular binding protein, FKBP. Rapamycin must be added within 2 days of mitogen stimulation in order to be fully effective. PC12 cells are refractory to the antiproliferative effect of rapamycin, although rapamycin does exert its expected inhibitory effect in PC12 cells on both basal and NGF-stimulated activation of one of its biochemical targets, the 70-kDa S6 protein kinase (p70S6K). The discordant findings suggest that a proliferative signal normally requiring activation of p70S6K either is unnecessary in PC12 cells or is provided by a downstream or cross-communicating pathway. They also suggest that p70S6K does not participate in the morphological responses of PC12 cells to NGF. Determining the basis for rapamycin resistance in PC12 cells might help to identify signaling abnormalities involved in the pathogenesis of pheochromocytoma.


Assuntos
Células Cromafins/efeitos dos fármacos , Imunossupressores/farmacologia , Células PC12/efeitos dos fármacos , Proteínas Quinases S6 Ribossômicas/efeitos dos fármacos , Sirolimo/farmacologia , Animais , Divisão Celular/efeitos dos fármacos , Células Cromafins/citologia , Células Cromafins/enzimologia , Feminino , Células PC12/citologia , Fosforilação/efeitos dos fármacos , Ratos , Ratos Endogâmicos F344 , Proteínas Quinases S6 Ribossômicas/metabolismo , Tacrolimo/farmacologia
15.
Chem Phys Lipids ; 127(2): 199-206, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14726002

RESUMO

In this study, we investigated the effects of lysophosphatidic acid (LPA) on melanogenesis in Mel-Ab cells. We found that LPA significantly attenuates melanin synthesis, and reduces the activity of tyrosinase, the rate-limiting melanogenic enzyme. Interestingly, LPA was also found to induce the activation of a 90 kDa ribosomal S6 kinase (RSK-1), which is known to phosphorylate microphthalmia-associated transcription factor (MITF) at serine 409. Though it has been previously reported that the phosphorylation of MITF is followed by the degradation of MITF, we found that LPA significantly inhibited MITF promoter activity, and that this reduced MITF and tyrosinase protein production. Our results indicate that LPA contributes to reduced melanin synthesis via the down-regulation of MITF.


Assuntos
Lisofosfolipídeos/farmacologia , Melaninas/biossíntese , Animais , Linhagem Celular , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Regulação para Baixo/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Melanócitos/citologia , Melanócitos/efeitos dos fármacos , Melanócitos/metabolismo , Camundongos , Fator de Transcrição Associado à Microftalmia , Monofenol Mono-Oxigenase/antagonistas & inibidores , Monofenol Mono-Oxigenase/metabolismo , Fosforilação , Regiões Promotoras Genéticas/efeitos dos fármacos , Proteínas Quinases S6 Ribossômicas/efeitos dos fármacos , Proteínas Quinases S6 Ribossômicas/metabolismo , Serina/metabolismo , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
16.
J Nutr Sci Vitaminol (Tokyo) ; 48(1): 59-64, 2002 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12026190

RESUMO

Leucine performs a signaling role to enhance protein synthesis by phosphorylating eukaryotic initiation factor (eIF) 4E-binding protein 1 (4E-BP1) and 70-kDa ribosomal protein S6 kinase (S6K1), two key regulatory proteins involved in the initiation of mRNA translation. The purpose of the current study was to assess whether the phosphorylation of 4E-BP1 and S6K1 was increased in skeletal muscle and liver by an oral administration of leucine to diabetic rats and to determine the in vivo contribution of insulin to a leucine-dependent induction of 4E-BP1 and S6K1 phosphorylation. Food-deprived (18 h) normal and diabetic rats were orally administered 135 mg/ 100 g body weight L-leucine and sacrificed at 1 h after administration. Leucine administration resulted in enhanced phosphorylation of 4E-BP1 and S6K1 in skeletal muscle and in liver of nondiabetic rats. The stimulatory action of leucine on the phosphorylation of 4E-BP1 and S6K1 in skeletal muscle was not abolished in rats with streptozotocin-induced diabetes. In contrast, leucine administration did not stimulate the phosphorylation of 4E-BP1 and S6K1 in the liver of diabetic rats. These findings suggest that in skeletal muscle, leucine functions as a nutritional signaling molecule that independently regulates the phosphorylation states of 4E-BP1 and S6K1. In contrast to skeletal muscle, insulin is essential in mediating the leucine-dependent induction of 4E-BP1 and S6K1 phosphorylation in liver. leucine, 4E-BP1, S6K1, translation initiation, diabetes


Assuntos
Proteínas de Transporte/efeitos dos fármacos , Proteínas de Transporte/metabolismo , Diabetes Mellitus/induzido quimicamente , Insulina/sangue , Leucina/administração & dosagem , Fígado/efeitos dos fármacos , Fígado/metabolismo , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/metabolismo , Fosfoproteínas/efeitos dos fármacos , Fosfoproteínas/metabolismo , Proteínas Quinases S6 Ribossômicas/efeitos dos fármacos , Proteínas Quinases S6 Ribossômicas/metabolismo , Administração Oral , Animais , Glicemia/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Peso Corporal/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular , Masculino , Fosforilação/efeitos dos fármacos , Ratos , Ratos Wistar
17.
Age (Dordr) ; 34(2): 371-87, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21472380

RESUMO

Leucine acts as a signal nutrient in promoting protein synthesis in skeletal muscle and adipose tissue via mTOR pathway activation, and may be of interest in age-related sarcopenia. However, hyper-activation of mTOR/S6K1 has been suggested to inhibit the first steps of insulin signaling and finally promote insulin resistance. The impact of long-term dietary leucine supplementation on insulin signaling and sensitivity was investigated in old rats (18 months old) fed a 15% protein diet supplemented (LEU group) or not (C group) with 4.5% leucine for 6 months. The resulting effects on muscle and fat were examined. mTOR/S6K1 signaling pathway was not significantly altered in muscle from old rats subjected to long-term dietary leucine excess, whereas it was increased in adipose tissue. Overall glucose tolerance was not changed but insulin-stimulated glucose transport was improved in muscles from leucine-supplemented rats related to improvement in Akt expression and phosphorylation in response to food intake. No change in skeletal muscle mass was observed, whereas perirenal adipose tissue mass accumulated (+45%) in leucine-supplemented rats. A prolonged leucine supplementation in old rats differently modulates mTOR/S6K pathways in muscle and adipose tissue. It does not increase muscle mass but seems to promote hypertrophy and hyperplasia of adipose tissue that did not result in insulin resistance.


Assuntos
Tecido Adiposo/metabolismo , Envelhecimento/fisiologia , Suplementos Nutricionais , Resistência à Insulina/fisiologia , Insulina/metabolismo , Leucina/administração & dosagem , Músculo Esquelético/metabolismo , Tecido Adiposo/efeitos dos fármacos , Envelhecimento/efeitos dos fármacos , Animais , Transporte Biológico/genética , Western Blotting , Modelos Animais de Doenças , Seguimentos , Glucose/metabolismo , Teste de Tolerância a Glucose , Masculino , Músculo Esquelético/efeitos dos fármacos , RNA/genética , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase em Tempo Real , Proteínas Quinases S6 Ribossômicas/biossíntese , Proteínas Quinases S6 Ribossômicas/efeitos dos fármacos , Proteínas Quinases S6 Ribossômicas/genética , Sarcopenia/dietoterapia , Sarcopenia/genética , Sarcopenia/metabolismo , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR/biossíntese , Serina-Treonina Quinases TOR/efeitos dos fármacos , Serina-Treonina Quinases TOR/genética , Fatores de Tempo
18.
Oncol Rep ; 23(4): 1167-72, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20204306

RESUMO

Esophageal cancer is one of the most frequently occurring cancers in the world. Targeting therapy strategy of cancer with specific inhibitors is developing and has showed promising antitumor efficacy. It is known that mTOR is an important controller of cell growth. RAD001 (everolimus) is a specific inhibitor of mTOR that can block the mTOR signaling pathway. The purposes of this study was to explore the inhibitory effects of RAD001 on mTOR signaling and the mechanism of cell growth suppression by RAD001. We examined both the expression of mTOR, p70S6K and S6 in SEG-1 esophageal cancer cells and KOB-13 normal esophageal epithelial cells and the efficacy of RAD001 against SEG-1 esophageal cancer cells. mTOR, p70S6K and S6 were overexpressed in SEG-1 esophageal cancer cells compared with KOB-13 normal esophageal epithelial cells. SEG-1 esophageal cancer cells were sensitive to RAD001. The survival rate of the cells treated with RAD001 over 0.33 microM was significantly different compared with that of control (P<0.01). RAD001 inhibited the phosphorylation of mTOR (Ser2448) and S6 (Ser240/244) in different grades and the expressions of mTOR, p70S6K and S6. As a result, RAD001 induced a dose-dependent decrease in cell proliferation, G1/S arrest and damage of cell shape. Taken together, these data showed that RAD001 can inhibit mTOR signaling and proliferation in SEG-1 esophageal cancer cells in vitro. It offers a therapeutic intervention through inhibition of mTOR as a potential strategy for esophageal cancer.


Assuntos
Antineoplásicos/farmacologia , Neoplasias Esofágicas/tratamento farmacológico , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Sirolimo/análogos & derivados , Western Blotting , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Separação Celular , Neoplasias Esofágicas/metabolismo , Everolimo , Citometria de Fluxo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/efeitos dos fármacos , Proteínas Quinases S6 Ribossômicas/efeitos dos fármacos , Proteínas Quinases S6 Ribossômicas/metabolismo , Proteínas Quinases S6 Ribossômicas 70-kDa/efeitos dos fármacos , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Transdução de Sinais/efeitos dos fármacos , Sirolimo/farmacologia , Serina-Treonina Quinases TOR
19.
J Dent Res ; 88(12): 1113-8, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19892916

RESUMO

Oral mucosa progenitor/stem cells reside as a small-sized cell population that eventually differentiates concurrently with an increase in cell size. Activation of the mammalian target of rapamycin (mTOR) leads to an increase in cell size. We hypothesized that rapamycin, a specific inhibitor of mTOR, will maintain primary human oral keratinocytes as a small-sized, undifferentiated cell population capable of retaining their proliferative capacity. Primary, rapamycin-treated (2 nM, 20 nM) oral keratinocytes showed a diminished cell size that correlated with a higher clonogenicity, a longer-term proliferative potential, and a slower cycling cell population concurrent with decreased expression of a differentiation marker when compared with untreated cells. Only the 2-nM rapamycin-treated oral keratinocytes maintained their ability to regenerate oral mucosa in vitro after 15 weeks of culture. Rapamycin, a Food and Drug Administration-approved drug, may have applicability for use in creating a highly proliferative cell population for use in regenerative medicine.


Assuntos
Queratinócitos/efeitos dos fármacos , Mucosa Bucal/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos , Antibacterianos/farmacologia , Adesão Celular/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Tamanho Celular/efeitos dos fármacos , Células Cultivadas , Células Clonais/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/fisiologia , Citometria de Fluxo , Humanos , Queratinócitos/fisiologia , Mucosa Bucal/citologia , Inibidores de Fosfoinositídeo-3 Quinase , Proteínas Quinases/efeitos dos fármacos , Regeneração/efeitos dos fármacos , Regeneração/fisiologia , Proteínas Quinases S6 Ribossômicas/efeitos dos fármacos , Sirolimo/farmacologia , Células-Tronco/fisiologia , Serina-Treonina Quinases TOR
20.
Eur J Biochem ; 251(1-2): 269-74, 1998 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-9492293

RESUMO

Dormant Artemia salina cysts contain desiccated gastrulae that are metabolically inactive, and physiologically arrested. Following rehydration, embryos resume development via alterations in protein expression, in the complete absence of cell division. In mammals, activation of p70 ribosomal S6 kinase (p70S6k) has been implicated in translational control, in particular the selective up-regulation of translation of mRNAs with polypyrimidine tracts at their 5' start sites. We therefore investigated ribosomal S6 kinase activity in preemergence development. We demonstrate that an S6 kinase activity is rapidly stimulated (within < 15 min) following rehydration and coincides with the onset of ribosomal S6 subunit phosphorylation. This S6 kinase activity displays chromatographic and biochemical characteristics that are similar to those of mammalian p70S6k. Partially purified Artemia S6 kinase was inactivated by treatment with protein phosphatase 2A. Activation of S6 kinase activity was shown to be due to an enzymatic step(s), and not simply rehydration of stored, active enzyme. The temporal profile of activation of S6 kinase activity is compatible with a regulatory function for p70S6k in early preemergence development of encysted Artemia. These studies identify activated Artemia cysts as a system for biochemical studies of p70S6k regulation.


Assuntos
Artemia/embriologia , Proteínas Quinases S6 Ribossômicas/metabolismo , Sequência de Aminoácidos , Animais , Western Blotting , Cromatografia Líquida/métodos , Citosol/enzimologia , Desidratação , Embrião não Mamífero/enzimologia , Embrião não Mamífero/fisiologia , Ativação Enzimática/efeitos dos fármacos , Dados de Sequência Molecular , Oligopeptídeos/metabolismo , Fosfoproteínas Fosfatases/farmacologia , Proteína Fosfatase 2 , Proteínas Quinases S6 Ribossômicas/efeitos dos fármacos , Especificidade por Substrato
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA