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










Intervalo de ano de publicação
1.
Invest Ophthalmol Vis Sci ; 62(10): 25, 2021 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-34424263

RESUMO

Purpose: Paxillin (PXN) is a key component of focal adhesions and plays an important role in angiogenesis. The aim of the present study was to investigate the effect of PXN in vascular endothelial growth factor A (VEGF-A)-induced angiogenesis in human umbilical vein endothelial cells (HUVECs). Methods: HUVECs were transfected with PXN overexpression and PXN interference vectors. Biochemical detection was used to detect adenosine triphosphate and lactic acid production. The morphology of mitochondria was observed under an electron microscope, and flow cytometry was conducted to measure mitochondrial membrane potential. Transwell experiments were used to detect the migration and tube formation ability of each group of cells. The expression of hexokinase (HK)1, HK2, glucose transporter 1 (GLUT1), phosphorylated phosphatidylinositol 3-kinase (PI3K), phosphorylated AKT, and phosphorylated mechanistic target of rapamycin (mTOR) was evaluated by western blot. Results: PXN silencing reduced the levels of lactic acid and adenosine triphosphate, downregulated HK1, HK2, and GLUT1, suppressed PI3K/AKT/mTOR signaling activation, and inhibited VEGF-A-induced mitochondria injury in VEGF-A-induced HUVECs. We also determined that miR-145-5p decreased the VEGF-A-induced expression of PXN and inhibited the invasion and angiogenesis of HUVECs. Also, miR-145-5p inhibition blocked the protective effect of PXN interference on VEGF-A-induced HUVEC injury. Furthermore, PXN interference significantly decreased lactic acid and adenosine triphosphate levels, inhibited PI3K/AKT/mTOR activation, and decreased the levels of HK1, HK2, and GLUT1 in VEGF-A-treated mouse corneal. Conclusions: The results indicate that PXN silencing inhibited the VEGF-A-induced invasion and angiogenesis of HUVECs via regulation of cell metabolism and mitochondrial damage, suggesting that PXN may be a potential target for antiangiogenic therapies.


Assuntos
Neovascularização da Córnea/genética , Regulação da Expressão Gênica , MicroRNAs/genética , Fator A de Crescimento do Endotélio Vascular/efeitos adversos , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Neovascularização da Córnea/induzido quimicamente , Neovascularização da Córnea/metabolismo , Humanos , MicroRNAs/biossíntese , Paxilina/biossíntese , Paxilina/genética , RNA/genética , Transdução de Sinais/efeitos dos fármacos
2.
Pediatr Hematol Oncol ; 34(4): 221-230, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-29040002

RESUMO

BACKGROUND: Neuroblastoma (NB) often presents with metastatic disease and poor survival. The need for new prognostic markers remains invaluable. The FAK-Src-Paxillin protein system is associated with aggressive phenotype in adult malignancies but is largely unexplored in pediatric NB. OBJECTIVE: To assess FAK-Src-Paxillin protein expression in human NB cell lines and clinical cytology material and to delineate its association with survival. DESIGN/METHODS: Western blot and immunohistochemistry were applied for FAK-Src-Paxillin expression in NB cell lines and 23 human cytology specimens, respectively. Protein expression in human clinical samples was correlated with clinicopathological parameters, MYCN amplification and survival. RESULTS: FAK, Src and Paxillin proteins are expressed in human NB cells lines, and can be detected in clinical cytology specimens from NB patients, (59%, 32% and 33% respectively). Simultaneous FAK-Src-Paxillin expression was noted in 30% of NB patients. Children with concomitant positivity FAK, Src, and Paxillin tumors, as well as MYCN amplification, had increased mortality compared to those without. CONCLUSIONS: FAK-Src-Paxillin system is a marker of unfavorable prognosis for human NB patients but also a promising therapeutic target.


Assuntos
Biomarcadores Tumorais/biossíntese , Quinase 1 de Adesão Focal/biossíntese , Regulação Neoplásica da Expressão Gênica , Neuroblastoma , Paxilina/biossíntese , Proteínas Proto-Oncogênicas pp60(c-src)/biossíntese , Animais , Criança , Pré-Escolar , Intervalo Livre de Doença , Feminino , Humanos , Lactente , Células K562 , Masculino , Camundongos , Proteína Proto-Oncogênica N-Myc/biossíntese , Células NIH 3T3 , Neuroblastoma/metabolismo , Neuroblastoma/mortalidade , Neuroblastoma/patologia , Taxa de Sobrevida
3.
PLoS One ; 11(3): e0152566, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27031101

RESUMO

INTRODUCTION: Recently, we introduced a new deposition method, based on Ion Plating Plasma Assisted technology, to coat titanium implants with a thin but hard nanostructured layer composed of titanium carbide and titanium oxides, clustered around graphitic carbon. The nanostructured layer has a double effect: protects the bulk titanium against the harsh conditions of biological tissues and in the same time has a stimulating action on osteoblasts. RESULTS: The aim of this work is to describe the biological effects of this layer on osteoblasts cultured in vitro. We demonstrate that the nanostructured layer causes an overexpression of many early genes correlated to proteins involved in bone turnover and an increase in the number of surface receptors for α3ß1 integrin, talin, paxillin. Analyses at single-cell level, by scanning electron microscopy, atomic force microscopy, and single cell force spectroscopy, show how the proliferation, adhesion and spreading of cells cultured on coated titanium samples are higher than on uncoated titanium ones. Finally, the chemistry of the layer induces a better formation of blood clots and a higher number of adhered platelets, compared to the uncoated cases, and these are useful features to improve the speed of implant osseointegration. CONCLUSION: In summary, the nanostructured TiC film, due to its physical and chemical properties, can be used to protect the implants and to improve their acceptance by the bone.


Assuntos
Materiais Revestidos Biocompatíveis/química , Grafite/química , Membranas Artificiais , Nanoestruturas/química , Osteoblastos/metabolismo , Titânio/química , Adesão Celular , Linhagem Celular Tumoral , Proliferação de Células , Humanos , Integrina alfa3beta1/biossíntese , Osseointegração , Osteoblastos/citologia , Paxilina/biossíntese , Talina/biossíntese
4.
Biochim Biophys Acta ; 1860(8): 1753-63, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27068854

RESUMO

We previously demonstrated that focal adhesion kinase (FAK), p130Cas and paxillin are crucially involved in the enhanced malignant properties under expression of ganglioside GD3 in melanoma cells. Therefore, molecules existing in the GD3-mediated signaling pathway could be considered as suitable targets for therapeutic intervention in malignant melanoma. The aim of this study was to determine whether blockade of p130Cas and/or paxillin by RNAi suppresses melanoma growth. We found a suitable dose (40 µM siRNA, 25 µl/tumor) of the siRNA to suppress p130Cas in the xenografts generated in nu/nu mice. Based on these results, we performed intratumoral (i.t.) treatment with anti-p130Cas and/or anti-paxillin siRNAs mixed with atelocollagen as a drug delivery system in a xenograft tumor of a human melanoma cell line, SK-MEL-28. Mixture of atelocollagen (1.75%) and an siRNA (500 or 1000 pmol/tumor) was injected into the tumors every 3 days after the first injection. An siRNA against human p130Cas markedly suppressed tumor growth of the xenograft in a dose-dependent manner, whereas siRNA against human paxillin slightly inhibited the tumor growth. A control siRNA against firefly luciferase showed no effect. To our surprise, siRNA against human p130Cas (500 or 1000 pmol/tumor) combined with siRNA against human paxillin dramatically suppressed tumor growth. In agreement with the tumor suppression effects of the anti-p130Cas siRNA, reduction in Ki-67 positive cell number as well as in p130Cas expression was demonstrated by immunohistostaining. These results suggested that blockade of GD3-mediated growth signaling pathways by siRNAs might be a novel and promising therapeutic strategy against malignant melanomas, provided signaling molecules such as p130Cas and paxillin are significantly expressed in individual cases. This article is part of a Special Issue entitled "Glycans in personalised medicine" Guest Editor: Professor Gordan Lauc.


Assuntos
Proteína Substrato Associada a Crk , Gangliosídeos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Melanoma , Proteínas de Neoplasias , Paxilina , RNA Interferente Pequeno , Animais , Linhagem Celular Tumoral , Proteína Substrato Associada a Crk/antagonistas & inibidores , Proteína Substrato Associada a Crk/biossíntese , Proteína Substrato Associada a Crk/genética , Gangliosídeos/biossíntese , Gangliosídeos/genética , Humanos , Melanoma/tratamento farmacológico , Melanoma/genética , Melanoma/metabolismo , Melanoma/patologia , Camundongos , Camundongos Nus , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Paxilina/antagonistas & inibidores , Paxilina/biossíntese , Paxilina/genética , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/farmacologia , Transdução de Sinais , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Exp Eye Res ; 145: 10-16, 2016 04.
Artigo em Inglês | MEDLINE | ID: mdl-26546726

RESUMO

Topical fluoroquinolones are widely used to prevent ocular infections after ophthalmic surgery. However, they have been shown to affect the corneal cell motility, whose mechanism remains indefinite. The purpose of this study was to investigate how fluoroquinolones affect corneal stromal cell motility. Human corneal fibroblasts (HCFs) were incubated in ciprofloxacin (CIP), levofloxacin (LEV), or moxifloxacin (MOX) at 0, 10, 50, and 100 µg/ml for up to 3 days. Effect of CIP, LEV, or MOX on HCF migration was monitored using migration assay. HCF viability was determined by WST-1 assay. Expression of focal adhesion kinase (FAK), paxillin (PXN), and their phosphorylated forms were analyzed by immunoblotting. Binding affinity between FAK and PXN was determined by co-immunoprecipitation. Our results revealed that CIP and MOX, but not LEV, noticeably retarded HCF migration. HCF proliferation was significantly reduced by CIP (38.2%), LEV (29.5%), and MOX (21.3%), respectively (p = 0.002). CIP and MOX suppressed the phosphorylation of PXN at tyrosines (10.2 ± 4.3%, p < 0.001; 11.7 ± 2.4%, p < 0.001, respectively), including tyrosine 118 (33.3 ± 5.2%, p < 0.001; 34.0 ± 4.4%, p < 0.001, respectively). CIP and MOX diminished the binding affinity between FAK and PXN (8.2 ± 1.8%, p < 0.001; 9.0 ± 4.5%, p < 0.001, respectively). Nevertheless, tyrosine dephosphorylation and FAK dissociation of PXN were not found in LEV-treated HCFs. None of these fluoroquinolones affect phosphorylation of FAK-Y397. We conclude that CIP and MOX, but not LEV, might delay corneal fibroblast migration via interfering with recruitment of PXN to focal adhesions and dephosphorylation of PXN at the tyrosines.


Assuntos
Úlcera da Córnea/tratamento farmacológico , Epitélio Corneano/metabolismo , Fluoroquinolonas/administração & dosagem , Proteína-Tirosina Quinases de Adesão Focal/biossíntese , Paxilina/biossíntese , Adulto , Idoso , Antibacterianos/administração & dosagem , Contagem de Células , Movimento Celular/efeitos dos fármacos , Úlcera da Córnea/metabolismo , Úlcera da Córnea/patologia , Epitélio Corneano/efeitos dos fármacos , Epitélio Corneano/patologia , Feminino , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibroblastos/patologia , Proteína-Tirosina Quinases de Adesão Focal/efeitos dos fármacos , Humanos , Immunoblotting , Masculino , Pessoa de Meia-Idade , Soluções Oftálmicas , Paxilina/efeitos dos fármacos , Adulto Jovem
6.
Cell Adh Migr ; 9(6): 441-51, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26491999

RESUMO

Directed cell migration is a crucial orchestrated process in embryonic development, wound healing, and immune response. The underlying substrate can provide physical and/or chemical cues that promote directed cell migration. Here, using electrospinning we developed substrates of aligned poly(lactic-co-glycolic acid) nanofibres to study the influence of glial cells on endothelial cells (ECs) in a 3-dimensional (3D) co-culture model. ECs build blood vessels and regulate their plasticity in coordination with neurons. Likewise, neurons construct nerves and regulate their circuits in coordination with ECs. In our model, the neuro-vascular cross-talk was assessed using a direct co-culture model of human umbilical vein endothelial cells (HUVECs) and rat Schwann cells (rSCs). The effect of rSCs on ECs behavior was demonstrated by earlier and higher velocity values and genetic expression profiles different of those of HUVECs when seeded alone. We observed 2 different gene expression trends in the co-culture models: (i) a later gene expression of angiogenic factors, such as interleukin-8 (IL-8) and vascular endothelial growth factor (VEGF), and (ii) an higher gene expression of genes involved in actin filaments rearrangement, such as focal adhesion kinase (FAK), Mitogen-activated protein kinase-activated protein kinase 13 (MAPKAPK13), Vinculin (VCL), and Profilin (PROF). These results suggested that the higher ECs migration is mainly due to proteins involved in the actin filaments rearrangement and in the directed cell migration rather than the effect of angiogenic factors. This co-culture model provides an approach to enlighten the neurovascular interactions, with particular focus on endothelial cell migration.


Assuntos
Movimento Celular/genética , Células Endoteliais/metabolismo , Nanofibras/química , Células de Schwann/metabolismo , Animais , Técnicas de Cocultura , Células Endoteliais/citologia , Quinase 1 de Adesão Focal/biossíntese , Proteína-Tirosina Quinases de Adesão Focal/biossíntese , Regulação da Expressão Gênica , Células Endoteliais da Veia Umbilical Humana , Humanos , Interleucina-8/biossíntese , Ácido Láctico/química , Ácido Láctico/farmacologia , Paxilina/biossíntese , Ácido Poliglicólico/química , Ácido Poliglicólico/farmacologia , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Ratos , Células de Schwann/citologia , Fator A de Crescimento do Endotélio Vascular/biossíntese , Vinculina/biossíntese
7.
J Biol Chem ; 290(10): 6653-69, 2015 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-25586177

RESUMO

Type 2 diabetes involves defective insulin secretion with islet inflammation governed in part by IL-1ß. Prolonged exposure of islets to high concentrations of IL-1ß (>24 h, 20 ng/ml) impairs beta cell function and survival. Conversely, exposure to lower concentrations of IL-1ß for >24 h improves these same parameters. The impact on insulin secretion of shorter exposure times to IL-1ß and the underlying molecular mechanisms are poorly understood and were the focus of this study. Treatment of rat primary beta cells, as well as rat or human whole islets, with 0.1 ng/ml IL-1ß for 2 h increased glucose-stimulated (but not basal) insulin secretion, whereas 20 ng/ml was without effect. Similar differential effects of IL-1ß depending on concentration were observed after 15 min of KCl stimulation but were prevented by diazoxide. Studies on sorted rat beta cells indicated that the enhancement of stimulated secretion by 0.1 ng/ml IL-1ß was mediated by the NF-κB pathway and c-JUN/JNK pathway acting in parallel to elicit focal adhesion remodeling and the phosphorylation of paxillin independently of upstream regulation by focal adhesion kinase. Because the beneficial effect of IL-1ß was dependent in part upon transcription, gene expression was analyzed by RNAseq. There were 18 genes regulated uniquely by 0.1 but not 20 ng/ml IL-1ß, which are mostly involved in transcription and apoptosis. These results indicate that 2 h of exposure of beta cells to a low but not a high concentration of IL-1ß enhances glucose-stimulated insulin secretion through focal adhesion and actin remodeling, as well as modulation of gene expression.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Adesões Focais/efeitos dos fármacos , Insulina/metabolismo , Interleucina-1beta/administração & dosagem , Actinas/efeitos dos fármacos , Actinas/metabolismo , Animais , Diabetes Mellitus Tipo 2/patologia , Proteína-Tirosina Quinases de Adesão Focal/biossíntese , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Adesões Focais/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Glucose/administração & dosagem , Glucose/metabolismo , Humanos , Secreção de Insulina , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Interleucina-1beta/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/biossíntese , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Paxilina/biossíntese , Cultura Primária de Células , Ratos
8.
J Mol Biol ; 427(4): 775-791, 2015 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-25268803

RESUMO

Mucopolysaccharidoses type III (MPSIII, Sanfilippo syndrome) are genetic diseases due to deficient heparan sulfate (HS) saccharide digestion by lysosomal exoglycanases. Progressive accumulation of undigested saccharides causes early-onset behavioural and cognitive symptoms. The precise role of these saccharides in the pathophysiological cascade is still unclear. We showed that exposure of wild-type neural cells to exogenous soluble HS fragments of at least eight saccharides activated integrin-based focal adhesions (FAs), which attach cells to the extracellular matrix. FAs were constitutively activated in MPSIII type B astrocytes or neural stem cells unless undigested saccharides were cleared by exogenous supply of the missing exoglycanase. Defective cell polarisation and oriented migration in response to focal extracellular stimuli in affected cells suggest improper sensing of the environment. We consistently observed abnormal organisation of the rostral migratory stream in the brain of adult mice with MPSIII type B. These results suggest that cell polarisation and oriented migration defects participate to the neurological disorders associated with Sanfilippo syndrome.


Assuntos
Astrócitos/metabolismo , Adesões Focais/metabolismo , Heparitina Sulfato/farmacologia , Mucopolissacaridose III/patologia , Células-Tronco Neurais/metabolismo , Animais , Astrócitos/citologia , Encéfalo/patologia , Movimento Celular/genética , Polaridade Celular/genética , Células Cultivadas , Ativação Enzimática , Quinase 1 de Adesão Focal/metabolismo , Heparitina Sulfato/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células-Tronco Neurais/citologia , Paxilina/biossíntese , Paxilina/genética , Fosforilação , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Suínos
9.
Methods Cell Biol ; 124: 363-89, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25287850

RESUMO

Integration of light and electron microscopes provides imaging tools in which fluorescent proteins can be localized to cellular structures with a high level of precision. However, until recently, there were few methods that could deliver specimens with sufficient fluorescent signal and electron contrast for dual imaging without intermediate staining steps. Here, we report protocols that preserve green fluorescent protein (GFP) in whole cells and in ultrathin sections of resin-embedded cells, with membrane contrast for integrated imaging. Critically, GFP is maintained in a stable and active state within the vacuum of an integrated light and scanning electron microscope. For light microscopists, additional structural information gives context to fluorescent protein expression in whole cells, illustrated here by analysis of filopodia and focal adhesions in Madin Darby canine kidney cells expressing GFP-Paxillin. For electron microscopists, GFP highlights the proteins of interest within the architectural space of the cell, illustrated here by localization of the conical lipid diacylglycerol to cellular membranes.


Assuntos
Adesões Focais/ultraestrutura , Pseudópodes/ultraestrutura , Animais , Membrana Celular/metabolismo , Membrana Celular/ultraestrutura , Criopreservação , Diglicerídeos/metabolismo , Cães , Proteínas de Fluorescência Verde/biossíntese , Células HeLa , Humanos , Células Madin Darby de Rim Canino , Microscopia Eletrônica de Varredura/métodos , Microscopia Eletrônica de Transmissão/métodos , Microscopia de Fluorescência/métodos , Microtomia , Paxilina/biossíntese , Inclusão em Plástico , Proteínas Recombinantes de Fusão/biossíntese
10.
Methods ; 66(2): 208-21, 2014 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-24184188

RESUMO

Mechanical force modulates myriad cellular functions including migration, alignment, proliferation, and gene transcription. Mechanotransduction, the transmission of mechanical forces and its translation into biochemical signals, may be mediated by force induced protein conformation changes, subsequently modulating protein signaling. For the paxillin and focal adhesion kinase interaction, we demonstrate that force-induced changes in protein complex conformation, dissociation constant, and binding Gibbs free energy can be quantified by lifetime-resolved fluorescence energy transfer microscopy combined with intensity imaging calibrated by fluorescence correlation spectroscopy. Comparison with in vitro data shows that this interaction is allosteric in vivo. Further, spatially resolved imaging and inhibitor assays show that this protein interaction and its mechano-sensitivity are equal in the cytosol and in the focal adhesions complexes indicating that the mechano-sensitivity of this interaction must be mediated by soluble factors but not based on protein tyrosine phosphorylation.


Assuntos
Mecanotransdução Celular , Algoritmos , Regulação Alostérica , Animais , Calibragem , Bovinos , Células Cultivadas , Citosol/metabolismo , Células Endoteliais/metabolismo , Transferência Ressonante de Energia de Fluorescência , Quinase 1 de Adesão Focal/biossíntese , Quinase 1 de Adesão Focal/química , Paxilina/biossíntese , Paxilina/química , Ligação Proteica , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/química , Análise de Célula Única , Espectrometria de Fluorescência , Termodinâmica
11.
Salvador; s.n; 2014. 145 p. ilus.
Tese em Português | LILACS | ID: biblio-1000981

RESUMO

As ORFs I e IV do genoma do HTLV-1 codificam, respectivamente, as proteínas p12/p8 (acessória) e Tax (regulatória). p12/p8, de 99 aminoácidos, pode ser clivada em sua extremidade amino terminal gerando a proteína p8. A primeira clivagem proteolítica de p12 remove o sinal de retenção ao RE, enquanto a segunda clivagem, gera o produto de 8kDa, referido como p8. p12 localiza-se no sistema de endomembranes, residindo em RE e aparato de Golgi, enquanto p8 dirige-se para a membrana plasmática, onde é recrutada para a sinapse imunológica, através da ligação com o receptor de células T (TCR), além de participar da sinapse virológica e da formação de conduítes. A proteína Tax, por outro lado, atua como transativador transcricional do HTLV-1, sendo referida também na indução da expressão de diversos genes celulares, aumentando a proliferação e a migração das células infectadas. Na via de transporte de vesículas secretórias, vesículas produzidas como pós-Golgi são transportadas ao longo do citoesqueleto por motores celulares. A Miosina-Va, um motor não convencional, transporta diversos cargos, incluindo vesículas secretórias, vesículas sinápticas e de retículo endoplasmático. Outra proteína relacionada ao citoesqueleto é a Paxilina, que atua como molécula adaptadora nas adesões focais e cuja expressão está aumentada em indivíduos TSP-HAM...


HTLV-1 ORFs I and IV encode respectively p12/p8 (accessory protein) and Tax (regulatory protein). The 99 amino acid p12 protein can be proteolytically cleaved at the amino terminus to generate the p8 protein. The first proteolytic cleavage removes the ER retention/retrieval signal at the amino terminus of p12, while the second cleavage generates the p8 protein. The p12 protein localizes to cellular endomembranes, within the ER and Golgi apparatus, while p8 traffics to lipid rafts at the cell surface and is recruited to the immunological synapse upon T-cell receptor (TCR) ligation, virological synapse and conduits. Tax on the other hand acts as viral transactivator and induces expression of many cellular genes, increasing proliferation and migration of infected cells. In secretory vesicle transport, vesicles produced as post-Golgi are moved along the cytoskeleton by motor proteins. The unconventional myosin motor, Myosin-Va, moves several cargoes including secretory vesicles, synaptic vesicles, and the endoplasmic reticulum. Another cytoskeleton associated protein is Paxillin, an adapter on focal adhesions which expression is increased in TSP-HAM patients...


Assuntos
Humanos , Paxilina/biossíntese , Paxilina/toxicidade , Paxilina/ultraestrutura , Produtos do Gene tax/análise , Produtos do Gene tax/imunologia , Produtos do Gene tax/isolamento & purificação , Produtos do Gene tax/sangue , Produtos do Gene tax/síntese química , Vírus Linfotrópico T Tipo 1 Humano/imunologia , Vírus Linfotrópico T Tipo 1 Humano/patogenicidade
12.
Pancreatology ; 13(4): 384-92, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23890137

RESUMO

BACKGROUND: Expression of WNT5A associated with aggressive tumor biology and poor clinical outcome of various types of cancer. However its function in the metastasis property of pancreatic cells still needs to be elucidated. METHODS: We detected the expressions of WNT5A, JNK1/p-JNK1 and Paxillin/p-Paxillin in cancer and the para-carcinoma tissues of pancreatic cancer. To understand how WNT5A/JNK signaling affects pancreatic cancer cell migration through the phosphorylation of cellular substrates of Paxillin, In vitro, we knocked down the WNT5A in PANC1, Capan-2 and HT1080 cell lines, and then tested the expression of JNK1. We detected the proteins of phosphorylation of Paxillin after JNK1 was inhibited and then the cells migration assay was evaluated. Moreover, JNK1 functionally phosphorylates serine178 on paxillin in vitro was detected .At last we subsequently observed whether WNT5A/JNK signaling modulates some molecule expressions relevant to focal adhesion (FA) formation and mesenchymal transition (EMT) and cell cycle. RESULTS: WNT5A, p-JNK1 and p-Paxillin were highly expressed in early stage of tumor tissues. In vitro, WNT5A/JNK signaling promotes cell migration in pancreatic cancer by phosphorylating serine178 on Paxillin, an FA adaptor, which means WNT5A may regulate FA's function.WNT5A up-regulates the molecule's expressions relevant to cell adhesion through the phosphorylation of JNK1, including MMP1, MMP2, ICAM and CD44. In addition, WNT5A/JNK signaling promoted the mRNA expressions of vimentin, but decreased in E-Cadherin expression, which suggested its regulatory effects on the EMT processes. WNT5A/JNK signaling didn't modulate cell proliferation. CONCLUSION: WNT5A/JNK signaling initiate cell migration of pancreatic cancer through activation of Paxillin, which suggested WNT5A has the potency of being an effective therapeutic target for the metastasis of pancreatic cancer.


Assuntos
Movimento Celular/efeitos dos fármacos , Proteína Quinase 8 Ativada por Mitógeno/fisiologia , Neoplasias Pancreáticas/fisiopatologia , Paxilina/metabolismo , Proteínas Proto-Oncogênicas/fisiologia , Transdução de Sinais/fisiologia , Proteínas Wnt/fisiologia , Adenocarcinoma/fisiopatologia , Linhagem Celular Tumoral , Humanos , Proteína Quinase 8 Ativada por Mitógeno/biossíntese , Paxilina/biossíntese , Fosforilação , Proteínas Proto-Oncogênicas/biossíntese , Proteínas Wnt/biossíntese , Proteína Wnt-5a
13.
J Cell Biochem ; 113(7): 2523-31, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22415859

RESUMO

Repulsive guidance molecules (RGMs) coordinate axon formation and iron homestasis. These molecules are also known as co-receptors of bone morphogenetic proteins (BMPs). However, the role played by RGMs in breast cancer remains unclear. The present study investigated the impact of RGMB on functions of breast cancer cells and corresponding mechanisms. RGMB was knocked down in breast cancer cells by way of an anti-RGMB ribozyme transgene. Knockdown of RGMB resulted in enhanced capacities of proliferation, adhesion, and migration in breast cancer cells. Further investigations demonstrated RGMB knockdown resulted in a reduced expression and activity of Caspase-3, accompanied with better survival in RGMB knockdown cells under serum starvation, which might be induced by its repression on MAPK JNK pathway. Up-regulations of Snai1, Twist, FAK, and Paxillin via enhanced Smad dependent sigaling led to increased capacities of adhesion and migration. Our current data firstly revealed that RGMB may act as a negative regulator in breast cancer through BMP signaling.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Neoplasias da Mama/metabolismo , Moléculas de Adesão Celular Neuronais/metabolismo , Proteínas Smad/metabolismo , Apoptose/genética , Caspase 3/metabolismo , Adesão Celular , Moléculas de Adesão Celular Neuronais/genética , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Sobrevivência Celular , Feminino , Quinase 1 de Adesão Focal/biossíntese , Adesões Focais/metabolismo , Técnicas de Silenciamento de Genes , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Nucleares/biossíntese , Paxilina/biossíntese , Transdução de Sinais , Fatores de Transcrição da Família Snail , Fatores de Transcrição/biossíntese , Proteína 1 Relacionada a Twist/biossíntese
14.
Mol Vis ; 17: 1877-90, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21850162

RESUMO

PURPOSE: The outflow facility for aqueous humor across the trabecular meshwork (TM) is enhanced by agents that oppose the actomyosin contraction of its resident cells. Phosphorylation of MYPT1 (myosin light chain [MLC] phosphatase complex of Type 1) at Thr853 and Thr696 inhibits dephosphorylation of MLC, leading to an increase in actomyosin contraction. In this study, we examined the effects of Rho kinase (ROCK) inhibitors on the relative dephosphorylation of the two sites of MYPT1 using human TM cells (GTM3). METHODS: Dephosphorylation of MYPT1 at Thr853 and Thr696 was determined by western blot analysis following exposure to selective inhibitors of ROCK, namely Y-27632 and Y-39983. Consequent dephosphorylation of MLC and decreases in actomyosin contraction were assessed by western blot analysis and collagen gel contraction assay, respectively. Changes in the cell-matrix adhesion were measured in real time by electric cell-substrate impedance sensing and also assessed by staining for paxillin, vinculin, and focal adhesion kinase (FAK). RESULTS: Both ROCK inhibitors produced a concentration-dependent dephosphorylation at Thr853 and Thr696 of MYPT1 in adherent GTM3 cells. IC50 values for Y-39983 were 15 nM and 177 nM for dephosphorylation at Thr853 and Thr696, respectively. Corresponding values for Y-27632 were 658 nM and 2270 nM. Analysis of the same samples showed a decrease in MLC phosphorylation with IC50 values of 14 nM and 1065 nM for Y-39983 and Y-27632, respectively. Consistent with these changes, both inhibitors opposed contraction of collagen gels induced by TM cells. Exposure of cells to the inhibitors led to a decrease in the electrical cell-substrate resistance, with the effect of Y-39983 being more pronounced than Y-27632. Treatment with these ROCK inhibitors also showed a loss of stress fibers and a concomitant decrease in tyrosine phosphorylation of paxillin and FAK. CONCLUSIONS: Y-39983 and Y-27632 oppose ROCK-dependent phosphorylation of MYPT1 predominantly at Thr853 with a corresponding decrease in MLC phosphorylation. A relatively low effect of both ROCK inhibitors at Thr696 suggests a role for other Ser/Thr kinases at this site. Y-39983 was several-fold more potent when compared with Y-27632 at inhibiting the phosphorylation of MYPT1 at either Thr853 or Thr696 commensurate with its greater potency at inhibiting the activity of human ROCK-I and ROCK-II enzymes.


Assuntos
Actomiosina/metabolismo , Junções Célula-Matriz/metabolismo , Fosfatase de Miosina-de-Cadeia-Leve/metabolismo , Fosfoproteínas/metabolismo , Fibras de Estresse/efeitos dos fármacos , Malha Trabecular , Amidas/farmacologia , Humor Aquoso/fisiologia , Western Blotting , Linhagem Celular Transformada , Colágeno/análise , Proteína-Tirosina Quinases de Adesão Focal/análise , Proteína-Tirosina Quinases de Adesão Focal/biossíntese , Géis/análise , Humanos , Concentração Inibidora 50 , Fosfatase de Miosina-de-Cadeia-Leve/antagonistas & inibidores , Paxilina/análise , Paxilina/biossíntese , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Piridinas/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Malha Trabecular/citologia , Malha Trabecular/efeitos dos fármacos , Malha Trabecular/metabolismo , Vinculina/análise , Vinculina/biossíntese
15.
J Histochem Cytochem ; 59(9): 864-74, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21705647

RESUMO

Focal adhesions play a critical role as centers that transduce signals by cell-matrix interactions and regulate fundamental processes such as proliferation, migration, and differentiation. Focal adhesion kinase (FAK), paxillin, integrin-linked kinase (ILK), and hydrogen peroxide-inducible clone-5 (Hic-5) are major proteins that contribute to these events. In this study, we investigated the expression of focal adhesion proteins in the developing rat kidney. Western blotting analysis revealed that the protein levels of FAK, p-FAK(397), paxillin, p-paxillin(118), and Hic-5 were high in embryonic kidneys, while ILK expression persisted from the embryonic to the mature stage. Immunohistochemistry revealed that FAK, p-FAK(397), paxillin, and p-paxillin(118) were strongly expressed in condensed mesenchymal cells and the ureteric bud. They were detected in elongating tubules and immature glomerular cells in the nephrogenic zone. Hic-5 was predominantly expressed in mesenchymal cells as well as immature glomerular endothelial and mesangial cells, suggesting that Hic-5 might be involved in mesenchymal cell development. ILK expression was similar to that of FAK in the developmental stages. Interestingly, ILK was strongly expressed in podocytes in mature glomeruli. ILK might play a role in epithelial cell differentiation as well as kidney growth and morphogenesis. In conclusion, the temporospatially regulated expression of focal adhesion proteins during kidney development might play a role in morphogenesis and cell differentiation.


Assuntos
Adesões Focais/enzimologia , Adesões Focais/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Rim/crescimento & desenvolvimento , Rim/metabolismo , Animais , Western Blotting , Proteínas do Citoesqueleto/análise , Proteínas do Citoesqueleto/biossíntese , Proteínas do Citoesqueleto/imunologia , Proteínas de Ligação a DNA/análise , Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/imunologia , Proteína-Tirosina Quinases de Adesão Focal/análise , Proteína-Tirosina Quinases de Adesão Focal/biossíntese , Proteína-Tirosina Quinases de Adesão Focal/imunologia , Perfilação da Expressão Gênica , Imuno-Histoquímica , Rim/embriologia , Rim/imunologia , Proteínas com Domínio LIM , Paxilina/análise , Paxilina/biossíntese , Paxilina/imunologia , Proteínas Serina-Treonina Quinases/análise , Proteínas Serina-Treonina Quinases/biossíntese , Proteínas Serina-Treonina Quinases/imunologia , Ratos , Ratos Sprague-Dawley
16.
J Virol ; 85(3): 1360-9, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21084488

RESUMO

We have established that human cytomegalovirus (HCMV) infection modulates the biology of target primary peripheral blood monocytes, allowing HCMV to use monocytes as "vehicles" for its systemic spread. HCMV infection of monocytes results in rapid induction of phosphatidylinositol-3-kinase [PI(3)K] and NF-κB activities. Integrins, which are upstream of the PI(3)K and NF-κB pathways, were shown to be involved in HCMV binding to and entry into fibroblasts, suggesting that receptor ligand-mediated signaling following viral binding to integrins on monocytes could trigger the functional changes seen in infected monocytes. We now show that integrin engagement and the activation of the integrin/Src signaling pathway are essential for the induction of HCMV-infected monocyte motility. To investigate how integrin engagement by HCMV triggers monocyte motility, we examined the infected-monocyte transcriptome and found that the integrin/Src signaling pathway regulates the expression of paxillin, which is an important signal transducer in the regulation of actin rearrangement during cell adhesion and movement. Functionally, we observed that paxillin is activated via the integrin/Src signaling pathway and is required for monocyte motility. Because motility is intimately connected to cellular cytoskeletal organization, a process that is also important in viral entry, we investigated the role paxillin regulation plays in the process of viral entry into monocytes. New results confirmed that HCMV entry into target monocytes was significantly reduced in cells deficient in paxillin expression or the integrin/Src/paxillin signaling pathway. From our data, HCMV-cell interactions emerge as an essential trigger for the cellular changes that allow for HCMV entry and hematogenous dissemination.


Assuntos
Movimento Celular , Citomegalovirus/imunologia , Citomegalovirus/patogenicidade , Interações Hospedeiro-Patógeno , Monócitos/virologia , Paxilina/biossíntese , Ligação Viral , Células Cultivadas , Perfilação da Expressão Gênica , Humanos , Integrinas/metabolismo , Transdução de Sinais , Quinases da Família src/metabolismo
17.
Eur J Cell Biol ; 90(2-3): 249-60, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-20663583

RESUMO

Anchorage to matrix is mediated for many cells not only by integrin-based focal adhesions but also by a parallel assembly of integral and peripheral membrane proteins known as the Dystroglycan Complex. Deficiencies in either dystrophin (mdx mice) or γ-sarcoglycan (γSG(-/-) mice) components of the Dystroglycan Complex lead to upregulation of numerous focal adhesion proteins, and the phosphoprotein paxillin proves to be among the most prominent. In mdx muscle, paxillin-Y31 and Y118 are both hyper-phosphorylated as are key sites in focal adhesion kinase (FAK) and the stretch-stimulatable pro-survival MAPK pathway, whereas γSG(-/-) muscle exhibits more erratic hyper-phosphorylation. In cultured myotubes, cell tension generated by myosin-II appears required for localization of paxillin to adhesions while vinculin appears more stably integrated. Overexpression of wild-type (WT) paxillin has no obvious effect on focal adhesion density or the physical strength of adhesion, but WT and a Y118F mutant promote contractile sarcomere formation whereas a Y31F mutant shows no effect, implicating Y31 in striation. Self-peeling of cells as well as Atomic Force Microscopy (AFM) probing of cells with or without myosin-II inhibition indicate an increase in cell tension within paxillin-overexpressing cells. However, prednisolone, a first-line glucocorticoid for muscular dystrophies, decreases cell tension without affecting paxillin at adhesions, suggesting a non-linear relationship between paxillin and cell tension. Hypertension that results from upregulation of integrin adhesions is thus a natural and treatable outcome of Dystroglycan Complex down-regulation.


Assuntos
Distroglicanas/metabolismo , Adesões Focais/metabolismo , Paxilina/metabolismo , Animais , Distroglicanas/genética , Humanos , Camundongos , Células Musculares/citologia , Células Musculares/metabolismo , Distrofias Musculares/metabolismo , Distrofias Musculares/patologia , Paxilina/biossíntese , Paxilina/genética , Transdução de Sinais , Regulação para Cima
18.
Anticancer Res ; 30(12): 4861-6, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21187463

RESUMO

Motility of endothelial cells is a requirement for the vascularization of solid malignancies. While tumors have been shown to produce a host of angiogenic factors, including TGF-ß, the mechanisms by which such factors regulate endothelial cell motility have not yet been defined. Thus, the role of the serine/threonine phosphatase PP-1 in regulating endothelial cell motility and cytoskeletal architecture was studied. The present study demonstrated that TGF-ß stimulation of motility is dependent on PP-1. Likewise, TGF-ß was shown to up-regulate paxillin expression through a process that was PP-1 dependent. The interplay between PP-1 and TGF-ß was further observed by the induction of cell rounding and the loss of paxillin-actin co precipitations upon PP-1 inhibition and the compensation for these effects by TGF-ß. Studies initiated to determine how PP-1 might regulate motility showed its role in maintaining cytoskeletal organization and its capacity to directly dephosphorylate the focal adhesion scaffolding protein paxillin. These studies suggest that the interplay between TGF-ß and PP-1 regulates the motility of endothelial cells that is critical to the process of angiogenesis.


Assuntos
Carcinoma Pulmonar de Lewis/patologia , Movimento Celular/fisiologia , Citoesqueleto/patologia , Células Endoteliais/patologia , Proteína Fosfatase 1/fisiologia , Fator de Crescimento Transformador beta/fisiologia , Actinas/metabolismo , Animais , Carcinoma Pulmonar de Lewis/irrigação sanguínea , Carcinoma Pulmonar de Lewis/metabolismo , Movimento Celular/efeitos dos fármacos , Meios de Cultivo Condicionados , Citoesqueleto/efeitos dos fármacos , Citoesqueleto/metabolismo , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Adesões Focais/efeitos dos fármacos , Furanos/farmacologia , Humanos , Lipídeos/farmacologia , Camundongos , Paxilina/biossíntese , Paxilina/metabolismo , Fosforilação , Proteína Fosfatase 1/antagonistas & inibidores , Proteína Fosfatase 1/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta/farmacologia , Regulação para Cima/efeitos dos fármacos
19.
Cancer Res ; 70(24): 10392-401, 2010 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-21159652

RESUMO

Paxillin (PXN) gene mutations are associated with lung adenocarcinoma progression and PXN is known to be a target gene of microRNA-218 (miR-218). On this basis, we hypothesized that PXN overexpression via miR-218 suppression may promote tumor progression and metastasis and that PXN may predict survival and relapse in non-small cell lung cancer (NSCLC). Expression of miR-218 and PXN in 124 surgically resected lung tumors were evaluated by real-time PCR and immunohistochemical analysis. The prognostic value of miR-218 and PXN expression on overall survival (OS) and relapse-free survival (RFS) was analyzed by the Kaplan-Meier test and Cox regression analysis. miR-218 expression in lung tumors was negatively associated with PXN expression. Multivariate analyses showed that PXN and miR-218 might independently predict OS and RFS, respectively, in NSCLC. Moreover, patients with low miR-218 combined with PXN-positive had the worst OS and RFS among the 4 combinations. In a cell model, PXN was negatively regulated by miR-218 and cell proliferation, invasion, and soft agar colony formation were enhanced by PXN overexpression induced by miR-218 suppression. Taken together, our findings suggest that PXN overexpression induced by miR-218 suppression is an independent predictor of survival and relapse in NSCLC, highlighting PXN as a potential therapeutic target to improve clinical outcomes in this disease.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , MicroRNAs/antagonistas & inibidores , MicroRNAs/biossíntese , Paxilina/biossíntese , Carcinoma Pulmonar de Células não Pequenas/patologia , Carcinoma Pulmonar de Células não Pequenas/terapia , Linhagem Celular Tumoral , Feminino , Técnicas de Silenciamento de Genes , Humanos , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/terapia , Masculino , MicroRNAs/genética , Terapia de Alvo Molecular/métodos , Invasividade Neoplásica , Proteínas Oncogênicas Virais/genética , Proteínas Oncogênicas Virais/metabolismo , Paxilina/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Transcrição Gênica , Regulação para Cima
20.
Am J Physiol Lung Cell Mol Physiol ; 298(6): L735-43, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20190033

RESUMO

We have isolated mesenchymal stem cells (MSCs) from tracheal aspirates of premature infants with respiratory distress. We examined the capacity of MSCs to differentiate into myofibroblasts, cells that participate in lung development, injury, and repair. Gene expression was measured by array, qPCR, immunoblot, and immunocytochemistry. Unstimulated MSCs expressed mRNAs encoding contractile (e.g., ACTA2, TAGLN), extracellular matrix (COL1A1 and ELN), and actin-binding (DBN1, PXN) proteins, consistent with a myofibroblast phenotype, although there was little translation into immunoreactive protein. Incubation in serum-free medium increased contractile protein (ACTA2, MYH11) gene expression. MSC-conditioned medium showed substantial levels of TGF-beta1, and treatment of serum-deprived cells with a type I activin receptor-like kinase inhibitor, SB-431542, attenuated the expression of genes encoding contractile and extracellular matrix proteins. Treatment of MSCs with TGF-beta1 further induced the expression of mRNAs encoding contractile (ACTA2, MYH11, TAGLN, DES) and extracellular matrix proteins (FN1, ELN, COL1A1, COL1A2), and increased the protein expression of alpha-smooth muscle actin, myosin heavy chain, and SM22. In contrast, human bone marrow-derived MSCs failed to undergo TGF-beta1-induced myofibroblastic differentiation. Finally, primary cells from tracheal aspirates behaved in an identical manner as later passage cells. We conclude that human neonatal lung MSCs demonstrate an mRNA expression pattern characteristic of myofibroblast progenitor cells. Autocrine production of TGF-beta1 further drives myofibroblastic differentiation, suggesting that, in the absence of other signals, fibrosis represents the "default program" for neonatal lung MSC gene expression. These data are consistent with the notion that MSCs play a key role in neonatal lung injury and repair.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Pulmão/citologia , Células-Tronco Mesenquimais/metabolismo , Fator de Crescimento Transformador beta1/biossíntese , Actinas/biossíntese , Células Cultivadas , Colágeno Tipo I/biossíntese , Cadeia alfa 1 do Colágeno Tipo I , Elastina/biossíntese , Feminino , Perfilação da Expressão Gênica , Humanos , Ácidos Hidroxâmicos/farmacologia , Recém-Nascido , Recém-Nascido Prematuro , Pulmão/metabolismo , Masculino , Proteínas dos Microfilamentos/biossíntese , Proteínas Musculares/biossíntese , Neuropeptídeos/biossíntese , Paxilina/biossíntese , RNA Mensageiro/metabolismo , Síndrome do Desconforto Respiratório do Recém-Nascido/fisiopatologia , Fator de Crescimento Transformador beta1/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...