Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Tipo de estudo
Intervalo de ano de publicação
1.
Cell Mol Life Sci ; 70(17): 3057-66, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23096778

RESUMO

Cell motility is defined as cell movement in the three-dimensional space leading to repositioning of the cell. Atypical protein kinase C (aPKC, including ζ and λ/ι) are a subfamily of PKC. Different from classic PKC and novel PKC, the activation of atypical PKC is not dependent on diacylglycerol or calcium. PKCζ can be activated by lipid components, such as phosphatidylinositols, phosphatidic acid, arachidonic acid, and ceramide. Both phosphatidylinositol (3,4,5)-trisphosphate and PDK1 are necessary for the complete and stable activation of PKCζ. Atypical PKC is involved in the regulation of cell polarization, directional sensing, formation of filopodia, and cell motility. It is essential for migration and invasion of multiple cancer cell types. Particularly, atypical PKC has been found in the regulation of the motility of hematopoietic cells. It also participates in the regulation of proteolytic activity of podosomes and invadopodia. It has been found that atypical PKC can work coordinately with other PKC subfamily members and other signaling pathways. Research on the roles of atypical PKC in cell motility may lead to new therapeutic strategies for cancer and other diseases.


Assuntos
Movimento Celular/genética , Proteína Quinase C/metabolismo , Animais , Humanos , Neoplasias/enzimologia
2.
J Cell Physiol ; 228(2): 416-27, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22740332

RESUMO

Podosomes are adhesive structures on the ventral surface of cells that invade and degrade the extracellular matrix. Recently, we reported that phorbol 12,13-dibutyrate (PDBu), a protein kinase C (PKC) activator, induced podosome formation in normal human bronchial epithelial (NHBE) cells, and atypical PKCζ regulated MMP-9 recruitment to podosomes for its release and activation. The objective of this study was to explore signaling pathways that are involved in PKC activation-induced podosome formation and matrix degradation. Herein, we found that PDBu increased phosphorylation of PI3K p85, Akt, Src, ERK1/2, and JNK. Inhibitors for PI3K, Akt, and Src suppressed PDBu-induced podosome formation and matrix degradation. In contrast, blockers for MEK/ERK or JNK did not inhibit podosome formation but reduced proteolytic activity of podosomes. Inhibition of PKCζ activity with its pseudosubstrate peptide (PS)-inhibited PDBu-induced phosphorylation of MEK/ERK and JNK. On the other hand, inhibition of MEK/ERK or JNK pathway did not affect PKCζ phosphorylation, but reduced the recruitment of PKCζ and MMP-9 to podosomes. We conclude that PKCζ may regulate MEK/ERK and JNK phosphorylation and in turn activated MEK/ERK and JNK may regulate the proteolytic activity of PDBu-induced podosomes by influencing the recruitment of PKCζ and MMP-9 to podosomes.


Assuntos
Brônquios/enzimologia , Sistema de Sinalização das MAP Quinases , Metaloproteinase 9 da Matriz/metabolismo , Proteína Quinase C/metabolismo , Mucosa Respiratória/enzimologia , Brônquios/efeitos dos fármacos , Carcinógenos/farmacologia , Células Cultivadas , Inibidores Enzimáticos/farmacologia , Matriz Extracelular/metabolismo , Humanos , Dibutirato de 12,13-Forbol/farmacologia , Fosforilação , Mucosa Respiratória/efeitos dos fármacos
3.
Cell Mol Life Sci ; 69(24): 4149-62, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22718093

RESUMO

Airway epithelial cell migration is essential for lung development and growth, as well as the maintenance of respiratory tissue integrity. This vital cellular process is also important for the repair and regeneration of damaged airway epithelium. More importantly, several lung diseases characterized by aberrant tissue remodeling result from the improper repair of damaged respiratory tissue. Epithelial cell migration relies upon extracellular matrix molecules and is further regulated by numerous local, neuronal, and hormonal factors. Under inflammatory conditions, cell migration can also be stimulated by certain cytokines and chemokines. Many well-known environmental factors involved in the pathogenesis of chronic lung diseases (e.g., cigarette smoking, air pollution, alcohol intake, inflammation, viral and bacterial infections) can inhibit airway epithelial cell migration. Further investigation of cellular and molecular mechanisms of cell migration with advanced techniques may provide knowledge that is relevant to physiological and pathological conditions. These studies may eventually lead to the development of therapeutic interventions to improve lung repair and regeneration and to prevent aberrant remodeling in the lung.


Assuntos
Movimento Celular , Mucosa Respiratória/fisiopatologia , Doenças Respiratórias/fisiopatologia , Remodelação das Vias Aéreas , Consumo de Bebidas Alcoólicas/efeitos adversos , Fibrose Cística/patologia , Células Epiteliais/patologia , Células Epiteliais/fisiologia , Humanos , Pulmão/citologia , Pulmão/fisiopatologia , Regeneração , Mucosa Respiratória/citologia , Doenças Respiratórias/patologia , Fumar/efeitos adversos
4.
Cell Mol Life Sci ; 69(7): 1137-51, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21984596

RESUMO

Actin filament-associated protein (AFAP) plays a critical role in the regulation of actin filament integrity, formation and maintenance of the actin network, function of focal contacts, and cell migration. Here, we show that endogenous AFAP was present not only in the cytoskeletal but also in the cytosolic fraction. Depolymerization of actin filaments with cytochalasin D or latrunculin A increased AFAP in the cytosolic fraction. AFAP harbors an actin-binding domain (ABD) in its C-terminus. AFAPΔABD, an AFAP mutant with selective ABD deletion, was mainly in the cytosolic fraction when overexpressed in the cells, which was associated with a disorganized cytoskeleton with reduced stress fibers, accumulation of F-actin on cellular membrane, and formation of actin-rich small dots. Cortactin, a well-known podosome marker, was colocalized with AFAPΔABD in these small dots at the ventral surface of the cell, indicating that these small dots fulfill certain criteria of podosomes. However, these podosome-like small dots did not digest gelatin matrix. This may be due to the reduced interaction between AFAPΔABD and c-Src. When AFAPΔABD-transfected cells were stimulated with phorbol ester, they formed podosome-like structures with larger sizes, less numerous and longer life span, in comparison with wild-type AFAP-transfected cells. These results indicate that the association of AFAP with F-actin through ABD is crucial for AFAP to regulate cytoskeletal structures. The AFAPΔABD, as cytosolic proteins, may be more accessible to the cellular membrane, podosome-like structures, and thus be more interactive for the regulation of cellular functions.


Assuntos
Galinhas/metabolismo , Citoesqueleto/metabolismo , Proteínas dos Microfilamentos/metabolismo , Animais , Linhagem Celular , Citoplasma/química , Citoesqueleto/química , Ativação Enzimática , Humanos , Proteínas dos Microfilamentos/química
5.
FEBS Lett ; 585(3): 471-7, 2011 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-21236256

RESUMO

AFAP is an adaptor protein involved in cytoskeletal organization and intracellular signaling. AFAP binds and activates c-Src; however, the downstream signals of this interaction remain unknown. Here we show that co-expression of AFAP and c-Src induce transcriptional activation of SRE and AP-1 in a c-Src activity dependent fashion. Structural-functional studies suggest that the proline-rich motif in the N-terminus of AFAP is critical for c-Src activation, and subsequent SRE/AP-1 transactivation and the actin-binding domain in the AFAP C-terminus is negatively involved in the regulation of AFAP/c-Src mediated SRE/AP-1 transactivation. Selective deletion of this domain enhances transactivation of SRE. We conclude that in addition to its role in the regulation of cytoskeletal structures, AFAP may also be involved in the c-Src related transcriptional activities.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteínas dos Microfilamentos/metabolismo , Transdução de Sinais , Fator de Transcrição AP-1/metabolismo , Fatores de Transcrição/metabolismo , Ativação Transcricional , Quinases da Família src/metabolismo , Animais , Células COS , Galinhas , Chlorocebus aethiops , Proteínas de Ligação a DNA/genética , Genes Reporter , Células HEK293 , Humanos , Proteínas dos Microfilamentos/química , Proteínas dos Microfilamentos/genética , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Domínios e Motivos de Interação entre Proteínas , Proteínas Recombinantes de Fusão/metabolismo , Fator de Transcrição AP-1/genética , Fatores de Transcrição/genética , Transfecção , Domínios de Homologia de src , Quinases da Família src/genética
6.
Mol Cell Biol ; 30(23): 5545-61, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20937775

RESUMO

Podosomes are transient cell surface structures essential for degradation of extracellular matrix during cell invasion. Protein kinase C (PKC) is involved in the regulation of podosome formation; however, the roles of individual PKC isoforms in podosome formation and proteolytic function are largely unknown. Recently, we reported that PDBu, a PKC activator, induced podosome formation in normal human bronchial epithelial cells. Here, we demonstrate that phorbol-12,13-dibutyrate (PDBu)-induced podosome formation is mainly mediated through redistribution of conventional PKCs, especially PKCα, from the cytosol to the podosomes. Interestingly, although blocking atypical PKCζ did not affect PDBu-induced podosome formation, it significantly reduced matrix degradation at podosomes. Inhibition of PKCζ reduced recruitment of matrix metalloprotease 9 (MMP-9) to podosomes and its release and activation. Downregulation of MMP-9 by small interfering RNA (siRNA) or neutralization antibody also significantly reduced matrix degradation. The regulatory effects of PKCζ on matrix degradation and recruitment of MMP-9 to podosomes were PKCζ kinase activity dependent. PDBu-induced recruitment of PKCζ and MMP-9 to podosomes was blocked by inhibition of novel PKC with rottlerin or PKCδ siRNA. Our data suggest that multiple PKC isozymes form a signaling cascade that controls podosome formation and dynamics and MMP-9 recruitment, release, and activation in a coordinated fashion.


Assuntos
Extensões da Superfície Celular/enzimologia , Metaloproteinase 9 da Matriz/metabolismo , Proteína Quinase C/metabolismo , Sequência de Bases , Transporte Biológico Ativo/efeitos dos fármacos , Extensões da Superfície Celular/efeitos dos fármacos , Extensões da Superfície Celular/ultraestrutura , Células Cultivadas , Primers do DNA/genética , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/enzimologia , Células Epiteliais/ultraestrutura , Matriz Extracelular/efeitos dos fármacos , Matriz Extracelular/metabolismo , Técnicas de Silenciamento de Genes , Humanos , Isoenzimas/antagonistas & inibidores , Isoenzimas/genética , Isoenzimas/metabolismo , Metaloproteinase 14 da Matriz/genética , Metaloproteinase 14 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/genética , Inibidores de Metaloproteinases de Matriz , Dibutirato de 12,13-Forbol/farmacologia , Fosforilação , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/genética , RNA Interferente Pequeno/genética , Transdução de Sinais
7.
J Cell Physiol ; 218(2): 366-75, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18932175

RESUMO

Spreading and migration of the basal cells neighboring a wound is essential for airway epithelial repair. To gain insight into the molecular mechanisms that govern these cellular processes, we asked whether normal human airway epithelial cells can form podosomes, a cellular structure discovered from cancer and mesenchymal cells that controls migration and invasion. Herein, we report that phorbol-12, 13-dibutyrate (PDBu), a protein kinase C activator, induced reorganization of cytoskeletal structure in primary normal human bronchial epithelial cells, and in normal human airway epithelial BEAS2B cells. Z-stack scanning confocal microscopy showed that PDBu-induced podosome-like structures contain actin-rich columns that arise from the ventral surface of the cell, and also revealed the presence of circular ruffles/waves at the dorsal cell surface. The molecular components of these cytoskeletal structures were determined with immunofluorescent staining. Using in situ zymography, we demonstrated that PDBu-induced podosomes were capable of degrading fibronectin-gelatin-sucrose matrix. PDBu also increased epithelial cell invasion across Transwell chamber. Podosomes and circular dorsal ruffles may be important for epithelial cell migration and invasion, thus contributing to respiratory epithelial repair and regeneration.


Assuntos
Brônquios/citologia , Estruturas da Membrana Celular/efeitos dos fármacos , Estruturas da Membrana Celular/metabolismo , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Dibutirato de 12,13-Forbol/farmacologia , Linhagem Celular , Estruturas da Membrana Celular/enzimologia , Movimento Celular/efeitos dos fármacos , Citoesqueleto/efeitos dos fármacos , Citoesqueleto/metabolismo , Células Epiteliais/enzimologia , Matriz Extracelular/efeitos dos fármacos , Matriz Extracelular/metabolismo , Humanos , Metaloproteinases da Matriz/metabolismo , Proteínas Tirosina Quinases/metabolismo , Pseudópodes/efeitos dos fármacos , Pseudópodes/metabolismo , Vinculina/metabolismo
8.
Arch Biochem Biophys ; 468(2): 183-92, 2007 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-17964528

RESUMO

Advanced glycation end-products (AGEs) are one of the major factors of hyperglycemia related complications for diabetic patients. We studied the formation of AGEs in type I collagen after Fe2+-catalyzed non-enzymatic glycosylation in vitro. Type I collagen isolated from rat tail tendon was incubated with glucose and increasing concentrations of iron ions Fe2+. After 4 weeks incubation, cytotoxity of AGEs was indicated by the cytotoxity assay of primary human umbilical vein endothelial cells and primary human monocytes cultured with glycosylated collagen AGEs. Fourier transform infrared spectroscopy analysis revealed that structural changes of functional groups in glycosylated collagen are accelerated by the catalyst Fe2+. Using two-dimensional Fourier-transform infrared correlation spectroscopy analyses, for the first time, we demonstrated that the order of structural changes of these functional groups is -CH->Amide I>Amide II>Amide III>nu(C=O) the carboxylic group of Asn, Gln or polyproline amino acid residue in the course of AGE-collagen formation. Knowing the positions of these functional groups in collagen, this order of changes indicates that during glycation of collagen, the structure of the main chain residues in collagen changed first, and then the side chain changed gradually, which may lead to more carboxylic groups exposed to glucose for further formation of AGE-collagen irreversibly. The findings presented may support the design of new therapeutic strategies to prevent or slow down the Fe2+-catalyzed glycosylation of collagen and other matrix proteins.


Assuntos
Colágeno/metabolismo , Produtos Finais de Glicação Avançada/metabolismo , Ferro/administração & dosagem , Monócitos/química , Monócitos/metabolismo , Animais , Catálise , Células Cultivadas , Colágeno/química , Colágeno/ultraestrutura , Produtos Finais de Glicação Avançada/química , Glicosilação , Humanos , Ferro/química , Conformação Proteica/efeitos dos fármacos , Ratos
9.
J Biol Chem ; 282(22): 16401-12, 2007 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-17412687

RESUMO

Adaptor proteins are important mediators in signal transduction. In the present study, we report the cloning and characterization of a novel adaptor protein, XB130. This gene is located on human chromosome 10q25.3 and encodes a protein of 818 amino acids. It contains several Src homology (SH)2- and SH3-binding motifs, two pleckstrin homology domains, a coiled-coil region, and a number of potential tyrosine or serine/threonine phosphorylation sites. Endogenous XB130 interacts with c-Src tyrosine kinase. Their co-expression in COS-7 cells resulted in activation of c-Src and elevated tyrosine phosphorylation of multiple proteins, including XB130 itself. XB130 expression in HEK293 cells enhanced serum response element- and AP-1-dependent transcriptional activation mediated by c-Src. XB130DeltaN, an N-terminal deletion mutant lacking a putative SH3-binding motif and several putative SH2-binding sites, reduced its ability to mediate Src signal transduction. Down-regulation of endogenous XB130 with siRNA reduced c-Src activity, IL-8 production, EGF-induced phosphorylation of Akt and GSK3beta, and altered cell cycles in human lung epithelial cells. These data suggest that XB130 as an adaptor may play an important role in the regulation of signal transduction and cellular functions.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Cromossomos Humanos Par 10 , Células Epiteliais/metabolismo , Pulmão/metabolismo , Transdução de Sinais/fisiologia , Ativação Transcricional/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/antagonistas & inibidores , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Células COS , Proteína Tirosina Quinase CSK , Chlorocebus aethiops , Cromossomos Humanos Par 10/genética , Fator de Crescimento Epidérmico/farmacologia , Células Epiteliais/citologia , Humanos , Interleucina-8/biossíntese , Pulmão/citologia , Fosforilação/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/fisiologia , Processamento de Proteína Pós-Traducional/fisiologia , Proteínas Tirosina Quinases/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/farmacologia , Proteínas Repressoras/metabolismo , Elemento de Resposta Sérica/fisiologia , Transdução de Sinais/efeitos dos fármacos , Fator de Transcrição AP-1/genética , Fator de Transcrição AP-1/metabolismo , Ativação Transcricional/efeitos dos fármacos , Domínios de Homologia de src/genética , Quinases da Família src
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA