Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sensors (Basel) ; 19(9)2019 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-31058820

RESUMO

In this paper, one solution for an end-to-end deep neural network for autonomous driving is presented. The main objective of our work was to achieve autonomous driving with a light deep neural network suitable for deployment on embedded automotive platforms. There are several end-to-end deep neural networks used for autonomous driving, where the input to the machine learning algorithm are camera images and the output is the steering angle prediction, but those convolutional neural networks are significantly more complex than the network architecture we are proposing. The network architecture, computational complexity, and performance evaluation during autonomous driving using our network are compared with two other convolutional neural networks that we re-implemented with the aim to have an objective evaluation of the proposed network. The trained model of the proposed network is four times smaller than the PilotNet model and about 250 times smaller than AlexNet model. While complexity and size of the novel network are reduced in comparison to other models, which leads to lower latency and higher frame rate during inference, our network maintained the performance, achieving successful autonomous driving with similar efficiency compared to autonomous driving using two other models. Moreover, the proposed deep neural network downsized the needs for real-time inference hardware in terms of computational power, cost, and size.

2.
Prostate Cancer ; 2013: 398253, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23766912

RESUMO

Purpose. To determine whether Ski-interacting protein (SKIP) regulates TGF- ß 1-stimulated expression of urokinase-type plasminogen activator (uPA), matrix metalloproteinase-9 (MMP-9), and uPA Inhibitor (PAI-1) in the androgen-independent human prostate cancer cell model. Materials and Methods. PC-3 prostate cancer cell line was used. The role of SKIP was evaluated using synthetic small interference RNA (siRNA) compounds. The expression of uPA, MMP-9, and PAI-1 was evaluated by zymography assays, RT-PCR, and promoter transactivation analysis. Results. In PC-3 cells TGF- ß 1 treatment stimulated uPA, PAI-1, and MMP-9 expressions. The knockdown of SKIP in PC-3 cells enhanced the basal level of uPA, and TGF- ß 1 treatment inhibited uPA production. Both PAI-1 and MMP-9 production levels were increased in response to TGF- ß 1. The ectopic expression of SKIP inhibited both TGF- ß 1-induced uPA and MMP-9 promoter transactivation, while PAI-1 promoter response to the factor was unaffected. Conclusions. SKIP regulates the expression of uPA, PAI-1, and MMP-9 stimulated by TGF- ß 1 in PC-3 cells. Thus, SKIP is implicated in the regulation of extracellular matrix degradation and can therefore be suggested as a novel therapeutic target in prostate cancer treatment.

3.
Srp Arh Celok Lek ; 141(3-4): 178-86, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23745340

RESUMO

INTRODUCTION: Mesenchymal stem cells (MSCs) are a promising tool for regenerative medicine, but due to the heterogeneity of their populations, different sources and isolation techniques, the characteristics defining MSCs are inconsistent. OBJECTIVE: The aim of this study was to compare the characteristics of MSCs derived from two different human tissues: peripheral blood (PB-MSCs) and umbilical cord Wharton's Jelly (UC-MSCs). METHODS: The PB-MSC and UC-MSC were isolated by adherence to plastic after gradient-density separation or an explant culture method, respectively, and compared regarding their morphology, clonogenic efficiency, proliferating rates, immunophenotype and differentiation potential. RESULTS: MSCs derived from both sources exhibit similar morphology, proliferation capacity and multilineage (osteogenic, chondrogenic, adipogenic and myogenic) differentiation potential. Differences were observed in the clonogenic capacity and the immunophenotype, since UC-MSCs showed higher CFU-F (colony-forming units-fibroblastic) cloning efficiency, as well as higher embryonic markers (Na-nog, Sox2, SSEA4) expression. When additional surface antigens were analyzed by flow cytometry (CD44, CD90, CD105, CD33, CD34, CD45, CD11b, CD235a) or immunofluorescent labeling (vimentin, STRO-1 and alpha-smooth muscle actin), most appeared to have similar epitope profiles irrespective of MSC source. CONCLUSION: The results obtained demonstrated that both MSCs represent good alternative sources of adult MSCs that could be used in cell therapy applications.


Assuntos
Células Sanguíneas/citologia , Células-Tronco Mesenquimais/citologia , Cordão Umbilical/citologia , Geleia de Wharton/citologia , Adulto , Diferenciação Celular , Separação Celular/métodos , Células Cultivadas , Ensaio de Unidades Formadoras de Colônias , Feminino , Humanos , Imunofenotipagem , Gravidez
4.
Bone ; 52(1): 524-31, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23111315

RESUMO

We investigated the role of AMP-activated protein kinase (AMPK), Akt, mammalian target of rapamycin (mTOR), autophagy and their interplay in osteogenic differentiation of human dental pulp mesenchymal stem cells. The activation of various members of AMPK, Akt and mTOR signaling pathways and autophagy was analyzed by immunoblotting, while osteogenic differentiation was assessed by alkaline phosphatase staining and real-time RT-PCR/immunoblot quantification of osteocalcin, Runt-related transcription factor 2 and bone morphogenetic protein 2 mRNA and/or protein levels. Osteogenic differentiation of mesenchymal stem cells was associated with early (day 1) activation of AMPK and its target Raptor, coinciding with the inhibition of mTOR and its substrate p70S6 kinase. The early induction of autophagy was demonstrated by accumulation of autophagosome-bound LC3-II, upregulation of proautophagic beclin-1 and a decrease in the selective autophagic target p62. This was followed by the late activation of Akt/mTOR at days 3-7 of differentiation. The RNA interference-mediated silencing of AMPK, mTOR or autophagy-essential LC3ß, as well as the pharmacological inhibitors of AMPK (compound C), Akt (10-DEBC hydrochloride), mTOR (rapamycin) and autophagy (bafilomycin A1, chloroquine and ammonium chloride), each suppressed mesenchymal stem cell differentiation to osteoblasts. AMPK knockdown prevented early mTOR inhibition and autophagy induction, as well as late activation of Akt/mTOR signaling, while Akt inhibition suppressed mTOR activation without affecting AMPK phosphorylation. Our data indicate that AMPK controls osteogenic differentiation of human mesenchymal stem cells through both early mTOR inhibition-mediated autophagy and late activation of Akt/mTOR signaling axis.


Assuntos
Adenilato Quinase/metabolismo , Autofagia , Diferenciação Celular/fisiologia , Osteogênese , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo , Feminino , Humanos , Células-Tronco/metabolismo
5.
Int J Biochem Cell Biol ; 45(2): 464-75, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23183001

RESUMO

Interleukin-17 belongs to a family of pro-inflammatory cytokines with pleiotropic effects, which can be associated with several inflammatory diseases of the muscle tissue. Although elevated levels of interleukin-17 have been described in inflammatory myopathies, its role in muscle homeostasis remains to be elucidated. The requirement of the urokinase type plasminogen activator in skeletal myogenesis was recently demonstrated in vivo and in vitro, suggesting its involvement in the regulation of extracellular matrix remodeling, cell migration and myoblast fusion. Our previous results have demonstrated that interleukin-17 inhibits myogenic differentiation of C2C12 myoblasts in vitro concomitantly with the inhibition of cell migration. However, the involvement of urokinase type plasminogen activator in interleukin-17-inhibited myogenesis and migration remained to be analyzed. Therefore, the effect of interleukin-17 on the production of urokinase type plasminogen activator by C2C12 myoblasts was determined in the present study. Our results demonstrated that interleukin-17 strongly inhibits urokinase type plasminogen activator expression during myogenic differentiation. This reduction of urokinase type plasminogen activator production corresponded with the inhibition of cell migration by interleukin-17. Activation of p38 signaling pathway elicited by interleukin-17 mediated the inhibition of both urokinase type plasminogen activator expression and cell migration. Additionally, IL-17 inhibited C2C12 cells migration by causing the cells to reorganize their cytoskeleton and lose polarity. Therefore, our results suggest a novel mechanism by which interleukin-17 regulates myogenic differentiation through the inhibition of urokinase type plasminogen activator expression and cell migration. Accordingly, interleukin-17 may represent a potential clinical target worth investigating for the treatment of inflammatory muscle diseases.


Assuntos
Movimento Celular , Interleucina-17/fisiologia , Mioblastos/fisiologia , Ativador de Plasminogênio Tipo Uroquinase/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Citoesqueleto de Actina/metabolismo , Animais , Diferenciação Celular , Linhagem Celular , Proliferação de Células , Regulação para Baixo , Ativação Enzimática , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Imidazóis/farmacologia , Sistema de Sinalização das MAP Quinases , Camundongos , Piridinas/farmacologia , Ativação Transcricional , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores
6.
Biochim Biophys Acta ; 1823(4): 838-49, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22285818

RESUMO

The present study evaluated the role of interleukin (IL) 17 in multilineage commitment of C2C12 myoblastic cells and investigated associated signaling pathways. The results concerning the effects on cell function showed that IL-17 inhibits the migration of C2C12 cells, while not affecting their proliferation. The data regarding the influence on differentiation demonstrated that IL-17 inhibits myogenic differentiation of C2C12 cells by down-regulating the myogenin mRNA level, myosin heavy chain expression and myotube formation, but promotes their osteogenic differentiation by up-regulating the Runt-related transcription factor 2 mRNA level, cyclooxygenase-2 expression and alkaline phosphatase activity. IL-17 exerted these effects by activating ERK1,2 mitogen activated protein kinase signaling pathway, which in turn regulated the expression of relevant genes and proteins to inhibit myogenic differentiation and induce osteogenic differentiation. Additional analysis showed that the induction of osteogenic differentiation by IL-17 is independent of BMP signaling. The results obtained demonstrate the potential of IL-17 not only to inhibit the myogenic differentiation of C2C12 myoblasts but also to convert their differentiation pathway into that of osteoblast lineage providing new insight into the capacities of IL-17 to modulate the differentiation commitment.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Interleucina-17/farmacologia , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Desenvolvimento Muscular/efeitos dos fármacos , Mioblastos/citologia , Osteogênese/efeitos dos fármacos , Animais , Proteínas Morfogenéticas Ósseas/metabolismo , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Modelos Biológicos , Mioblastos/efeitos dos fármacos , Mioblastos/enzimologia , Receptores de Interleucina-17/metabolismo
7.
Biomol Concepts ; 3(5): 429-45, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25436548

RESUMO

Abstract Transforming growth factor-ß (TGF-ß) family members, including TGF-ßs and bone morphogenetic proteins (BMPs), play important roles in directing the fate of stem cells. In embryonic stem cells, the TGF-ß superfamily participates in almost all stages of cell development, such as cell maintenance, lineage selection, and progression of differentiation. In adult mesenchymal stem cells (MSCs), TGF-ßs can provide competence for early stages of chondroblastic and osteoblastic differentiation, but they inhibit myogenesis, adipogenesis, and late-stage osteoblast differentiation. BMPs also inhibit adipogenesis and myogenesis, but they strongly promote osteoblast differentiation. The TGF-ß superfamily members signal via specific serine/threonine kinase receptors and their nuclear effectors termed Smad proteins as well as through non-Smad pathways, which explain their pleiotropic effects in self-renewal and differentiation of stem cells. This review summarizes the current knowledge on the pleiotropic effects of the TGF-ß superfamily of growth factors on the fate of stem cells and also discusses the mechanisms by which the TGF-ß superfamily members control embryonic and MSCs differentiation.

8.
Scientifica (Cairo) ; 2012: 861647, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-24278749

RESUMO

Transforming growth factor-beta (TGF-ß1) is a potent inductor of matrix metalloproteinase-9 (MMP-9) in transformed cells. Recently, Ski-interacting protein (SKIP) has been described as a regulator of TGF-ß1 signal transduction, but its role in the induction of cell malignance by TGF-ß1 has not been fully elucidated so far. In the present study, we analyzed the role of SKIP on TGF-ß1-induced MMP-9 production. Mouse transformed keratinocytes (PDV) were stably transfected with SKIP antisense construct. We observed that SKIP depletion provoked an enhancement in the expression of MMP-9 in response to TGF-ß1 treatment. The downregulation of SKIP produced an enhancement in TGF-ß1-activated ERK1,2 MAP kinase as well as increased transactivation of downstream Elk1 transcription factor. The increased MMP-9 production in response to TGF-ß1 was dependent of MAPK activation as PD98059, an MEK inhibitor, reduced MMP-9 expression in SKIP antisense transfected cells. Thus, we propose SKIP as a regulatory protein in TGF-ß1-induced MMP-9 expression acting by controlling ERK1,2 signaling in transformed cells.

9.
Eur J Cancer ; 48(10): 1550-7, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21798735

RESUMO

Transforming growth factor-ß1 (TGF-ß1) stimulates the extracellular matrix degrading proteases expression and cell migration in order to enhance cancer cells malignancy. In the present study, we analysed the role of TGF-ß1-induced Smad3 activation in the urokinase type plasminogen activator (uPA) production, as well as in cell migration and E-cadherin downregulation in transformed PDV keratinocyte cell line. TGF-ß1 signalling was interfered by the chemical inhibitor of the TGF-ß1-receptor 1 (ALK5), SB505124, and the specific Smad3 inhibitor, SiS3. Our results showed that TGF-ß1 stimulates uPA expression directly through ALK5 activation. The inhibition of Smad3 strongly reduced the capacity of TGF-ß1 to stimulate uPA expression, in parallel decreasing the uPA inhibitor plasminogen activator inhibitor type 1 (PAI-1) expression. In addition, the transient expression of dominant negative Smad3 mutant inhibited the TGF-ß1-induced uPA promoter transactivation. Moreover, Smad3-/- mouse embryonic fibroblasts were refractory to the induction of uPA by TGF-ß1. The inhibition of both ALK5 and Smad3 dramatically blocked the TGF-ß1-stimulated E-cadherin downregulation, F-actin reorganisation and migration of PDV cells. Taken together, our results suggest that the TGF-ß1-induced activation of Smad3 is the critical step for the uPA upregulation and E-cadherin downregulation, which are the key events preceding the induction of cell migration by TGF-ß1 in transformed cells.


Assuntos
Regulação da Expressão Gênica , Queratinócitos/citologia , Proteína Smad3/genética , Proteína Smad3/fisiologia , Fator de Crescimento Transformador beta1/metabolismo , Ativador de Plasminogênio Tipo Uroquinase/biossíntese , Animais , Caderinas/metabolismo , Movimento Celular , Células Cultivadas , Fibroblastos/metabolismo , Humanos , Camundongos , Camundongos Transgênicos , Modelos Biológicos , Proteínas Serina-Treonina Quinases/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo I , Receptores de Fatores de Crescimento Transformadores beta/metabolismo
10.
Cell Tissue Res ; 346(3): 305-16, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22160457

RESUMO

The mouse is a suitable experimental model to study the biology of mesenchymal stem cells (MSCs), as well as to be used in biocompatibility studies and tissue engineering models. However, the isolation and purification of murine MSCs is far more challenging than their counterparts from other species. In this study, we isolated, expanded and characterized mouse MSCs from bone marrow (BM-MSCs). Additionally, we analyzed the effects of two regulatory molecules, interleukin 17 (IL-17) and basic fibroblast growth factor (bFGF), on BM-MSCs growth and elucidated the signaling pathways involved. The results revealed that IL-17 increased the frequency of colony-forming units fibroblast (CFU-F) as well as the BM-MSCs proliferation in a dose-dependent manner, while bFGF supplementation had no significant effect on CFU-F frequency but induced an increase in cell proliferation. Their combined usage did not produce additive effects on BM-MSCs proliferation and even induced reduction in the number of CFU-F. Also, the involvement of both p38 and extracellular signal-regulated kinase (ERK) mitogen-activated protein kinases (MAPKs) signaling in proliferative activity of IL-17 and bFGF on murine BM-MSCs and, moreover, the increased co-activation of a common signaling molecule, p38 MAPK, were demonstrated. Together, the data presented highlighted the role of IL-17 and bFGF in murine BM-MSCs proliferation and pointed to the complexity and specificity of the signaling networks leading to MSCs proliferation in response to different regulatory molecules.


Assuntos
Fatores de Crescimento de Fibroblastos/farmacologia , Interleucina-17/farmacologia , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Animais , Processos de Crescimento Celular/efeitos dos fármacos , Processos de Crescimento Celular/fisiologia , Células Cultivadas , Fatores de Crescimento de Fibroblastos/metabolismo , Interleucina-17/metabolismo , Masculino , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos A , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Proteínas Recombinantes/farmacologia , Transdução de Sinais
11.
FEBS Lett ; 584(22): 4586-92, 2010 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-20965173

RESUMO

Transforming growth factor-ß1 (TGF-ß1) potently induces the epithelial-mesenchymal transition (EMT) during tumoral progression. Although Sky-interacting protein (SKIP) regulates TGF-ß1-induced Smad activation, its role in the induction of cell malignance remains uncertain. We found that TGF-ß1 increases SKIP expression in PDV cells. In cells stably transfected with SKIP antisense, AS-S, Smad3 activation decreased, along with an inhibition of TGF-ß1-induced EMT, and the cells were sensitized to the TGF-ß1-dependent inhibition of proliferation. Also, AS-S cells showed a weaker migration and invasion response. Moreover, TGF-ß1-induced urokinase-type plasminogen activator expression was inhibited, concomitantly with a TGF-ß1-independent increment of the plasminogen-activator inhibitor-1 expression. Thus, these results suggest that SKIP is required for EMT and invasiveness induced by TGF-ß1 in transformed cells.


Assuntos
Movimento Celular/efeitos dos fármacos , Transformação Celular Neoplásica , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Queratinócitos/citologia , Queratinócitos/efeitos dos fármacos , Proteínas Nucleares/metabolismo , Fator de Crescimento Transformador beta1/farmacologia , Animais , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Queratinócitos/metabolismo , Queratinócitos/patologia , Camundongos , Invasividade Neoplásica , Proteínas Nucleares/deficiência , Proteínas Nucleares/genética , Inibidor 1 de Ativador de Plasminogênio/genética , Fatores de Transcrição , Ativador de Plasminogênio Tipo Uroquinase/genética
12.
Eur J Med Chem ; 45(9): 3669-76, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20684856

RESUMO

Three new complexes of the general formula L[RuCl(3)(DMSO)(3)] (1-3), where L = chlorpromazine hydrochloride, trifluoroperazine dihydrochloride or thioridazine hydrochloride, were prepared and characterized by elemental analysis and spectroscopic methods (FT-IR, UV-Vis, (1)H NMR and (13)C NMR). In addition, the crystal structure of the complex 2 containing trifluoroperazine dihydrochloride was solved by single crystal X-ray diffraction. The complex crystallizes in the monoclinic system, space group P2(1)/n, with a = 10.4935(7) A, b = 18.6836(12) A, c = 19.9250(13) A, beta = 98.448(2) degrees, V = 3864.0(4) A(3). The structure was refined to the agreement factors of R = 4.79%, R(w) = 11.23%. The effect of three different doses (0.4, 4.5 and 90.4 microM/kg bw) of complex 2 on superoxide dismutase (SOD) and catalase (CAT) activity was investigated under physiological conditions. Influence on nitrite production (NO(2)(-)) and the level of erythrocytes malondialdehyde (MDA) in rats blood was also evaluated. Complex 2 did not affect the CAT enzyme activity in vivo and did not cause the hydroxyl radicals production. In the 0.4 and 4.5 microM/kg bw doses it showed almost the same or lower SOD activity and nitrite levels, while the dose of 90.4 microM/kg bw significantly increased these parameters. Finally, the cytotoxicity of complexes were assayed in four human carcinoma cell lines MCF-7, MDA-MB-453 (breast carcinoma), SW-480 (colon adenocarcinoma) and IM9 (myeloma multiple cells). Antiproliferative activity in vitro with low IC(50) during 48 h of treatment was observed.


Assuntos
Compostos Organometálicos/química , Compostos Organometálicos/farmacologia , Fenotiazinas/química , Rutênio/química , Animais , Antineoplásicos/síntese química , Antineoplásicos/química , Antineoplásicos/farmacologia , Catalase/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Cristalografia por Raios X , Sequestradores de Radicais Livres/síntese química , Sequestradores de Radicais Livres/química , Sequestradores de Radicais Livres/farmacologia , Radicais Livres/química , Humanos , Masculino , Modelos Moleculares , Conformação Molecular , Compostos Organometálicos/síntese química , Ratos , Superóxido Dismutase/metabolismo
13.
FEBS Lett ; 584(11): 2305-10, 2010 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-20353788

RESUMO

Transforming growth factor-beta1 (TGF-beta1) activates Rac1 GTPase in mouse transformed keratinocytes. Expression of a constitutively active Q61LRac1 mutant induced an epithelial to mesenchymal transition (EMT) linked to stimulation of cell migration and invasion. On the contrary, expression of a dominant-negative N17TRac1 abolished TGF-beta1-induced cell scattering, migration and invasion. Moreover, Q61LRac1 enhanced metalloproteinase-9 (MMP9) production to levels comparable to those induced by TGF-beta1, while N17TRac1 was inhibitory. TGF-beta1-mediated EMT involves the expression of the E-cadherin repressor Snail1, regulated by the Rac1 and mitogen-activated protein kinase (MAPK) pathways. Furthermore, MMP9 production was MAPK-dependent, as the MEK inhibitor PD98059 decreased TGF-beta1-induced MMP9 expression and secretion in Q61LRac1 expressing cells. We propose that regulation of TGF-beta1-mediated plasticity of transformed keratinocytes requires the cooperation between the Rac1 and MAPK signalling pathways.


Assuntos
Células Epiteliais/metabolismo , Queratinócitos/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Animais , Caderinas/metabolismo , Desdiferenciação Celular , Movimento Celular , Inibidores Enzimáticos/farmacologia , Flavonoides/farmacologia , Camundongos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Transdução de Sinais
14.
Int J Cancer ; 127(1): 77-85, 2010 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-19908229

RESUMO

TGF-beta1 is a potent inductor of malignance in cancer cells. TGF-beta1 stimulates the expression of extracellular matrix degrading proteases, cell migration and it is also involved in the epithelial-mesenchymal transition (EMT). In the present work, we analyzed the role of Spred2 in the urokinase-type plasminogen activator (uPA) stimulation, EMT and cell migration by TGF-beta1. We found that both the expression of mRNA and the protein level of Spred2 were lower in transformed keratinocytes PDV compared with immortalized keratinocytes MCA-3D. The transient ectopic expression of Spred2 in PDV cells inhibited the TGF-beta1-transactivated SRE-Luc reporter which is related with the ERK1,2 signal. The stable ectopic expression of Spred2 in PDV cells (SP cells) led to the loss of ERK 1,2 activation by TGF-beta1, although Smad2 activation was not affected, and the knockdown of Spred2 enhanced the activation of ERK1,2 signal by TGF-beta1. The increment of uPA expression induced by TGF-beta1 was suppressed in SP cells. In contrast, the stimulus on PAI-1 expression was not affected and comparable to parental PDV cells. SP cells under TGF-beta1 treatment were unable to display the EMT, since the overexpression of Spred2 abolished the TGF-beta1-induced disruption of the E-cadherin cell to cell interactions, reorganization of the actin cytoskeleton and upregulation of the mesenchymal marker vimentin. Finally, SP cells could not respond to the TGF-beta1 stimulus on cell migration. Taken together, the data in the present study suggests that Spred2 is a regulator of TGF-beta1-induced malignance in transformed keratinocytes.


Assuntos
Movimento Celular/fisiologia , Células Epiteliais/citologia , Mesoderma/citologia , Proteínas Repressoras/fisiologia , Fator de Crescimento Transformador beta1/fisiologia , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Sequência de Bases , Primers do DNA , Ativação Enzimática , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Imunofluorescência , Técnicas de Silenciamento de Genes , Humanos , Proteínas Repressoras/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
15.
Vojnosanit Pregl ; 66(3): 223-7, 2009 Mar.
Artigo em Sérvio | MEDLINE | ID: mdl-19341229

RESUMO

BACKGROUND/AIM: Polymethyl methacrylate is used for producing a denture basis. It is a material made by the polymerization process of methyl methacrylate. Despite of the polymerization type, there is a certain amount of free methyl methacrylate (residual monomer) incorporated in the denture, which can cause irritation of the oral mucosa. The aim of this study was to determine the amount of residual monomer in four different denture base acrylic resins by liquid chromatography and the possibility of its reduction. METHODS: After the polymerization, a postpolymerization treatment was performed in three different ways: in boiling water for thirty minutes, with 500 W microwaves for three minutes and in steam bath at 22 degrees C for one to thirty days. RESULTS: The obtained results showed that the amount of residual monomer is significantly higher in cold polymerizing acrylates (9.1-11%). The amount of residual monomer after hot polymerization was in the tolerance range (0.59-0.86%). CONCLUSION: The obtained results denote a low content of residual monomer in the samples which have undergone postpolymerization treatment. A lower percent of residual monomer is established in samples undergone a hot polymerization.


Assuntos
Materiais Dentários/química , Bases de Dentadura , Metilmetacrilato/análise , Cromatografia Líquida
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...