Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Biochim Biophys Acta ; 1843(12): 3029-37, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25261706

RESUMO

Schlafen 3 (Slfn3) mediates rodent enterocyte differentiation in vitro and in vivo, required for intestinal function. Little is known about Schlafen protein structure-function relationships. To define the Slfn3 domain that promotes differentiation, we studied villin and sucrase isomaltase (SI) promoter activity in Slfn3-null human Caco-2BBE cells transfected with full-length rat Slfn3 DNA or truncated constructs. Confocal microscopy and Western blots showed that Slfn3 is predominantly cytosolic. Villin promoter activity, increased by wild type Slfn3, was further enhanced by adding a nuclear exclusion sequence, suggesting that Slfn3 does not affect transcription by direct nuclear action. We therefore sought to dissect the region in Slfn3 stimulating promoter activity. Since examination of the Slfn3 N-terminal region revealed sequences similar to both an aminopeptidase (App) and a divergent P-loop resembling those in NTPases, we initially divided Slfn3 into an N-terminal domain containing the App and P-loop regions, and a C-terminal region. Only the N-terminal construct stimulated promoter activity. Further truncation indicated that both the App and the smaller P-loop constructs enhanced promoter activity similarly to the N-terminal sequence. Point mutations within the N-terminal region (R128L, altering a critical active site residue in the App domain, and L212D, conserved in Schlafens but variable in P-loop proteins) did not affect activity. These results show that Slfn3 acts in the cytosol to trigger a secondary signal cascade that elicits differentiation marker expression and narrows the active domain to the third of the Slfn3 sequence homologous to P-loop NTPases, a first step in understanding its mechanism of action.

2.
J Nanosci Nanotechnol ; 15(9): 6413-22, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26690867

RESUMO

Triple negative breast cancer exhibit increased IL-6 expression compared with matched healthy breast tissue and a strong link between inflammation and cancer progression and metastasis has been reported. We investigated whether doxorubicin-hyaluronan-super-paramagnetic iron oxide nanoparticles (DOX-HA-SPION) would show greater therapeutic efficacy in human triple negative breast cancer cells (TNBC) MDA-MB-231, as was recently shown in drug-sensitive and multi-drug-resistant ovarian cancer cells. Therefore, we measured cellular DOX uptake via confocal microscopy; observed morphologic changes: mitochondrial and nuclear changes with electron microscopy, and quantitated apoptosis using FACS analysis after Annexin V and PI staining in MDA-MB-231 cells treated with either DOX alone or DOX-HA-SPION. We also measured both proinflammatory and anti-inflammatory cytokines; IL-6, IL-10 respectively and also measured nitrate levels in the conditioned medium by ELISA. Inaddition, NF-κB activity was measured by luciferase assay. Confocal microscopy demonstrated greater cytoplasmic uptake of DOX-HA-SPION than free DOX. We also demonstrated reduction of Vimentin with DOX-HA-SPION which is significantly less than both control and DOX. DOX-HA-SPION enhanced apoptosis and significantly down regulated both pro-inflammatory mediators IL-6 and NF-κB in comparison to DOX alone. The secretion levels of anti-inflammatory mediators IL-10 and nitrate was not decreased in the DOX or DOX-HA-SPION treatment groups. Our data suggest that DOX-HA-SPION nanomedicine-based drug delivery could have promising potential in treating metastasized and chemoresistant breast cancer by enhancing the drug efficacy and minimizing off-target effects.


Assuntos
Antineoplásicos/farmacocinética , Apoptose/efeitos dos fármacos , Doxorrubicina/farmacocinética , Ácido Hialurônico/química , Interleucina-6/metabolismo , Nanopartículas de Magnetita/química , NF-kappa B/metabolismo , Antineoplásicos/química , Antineoplásicos/farmacologia , Neoplasias da Mama , Linhagem Celular Tumoral , Forma Celular/efeitos dos fármacos , Doxorrubicina/química , Doxorrubicina/farmacologia , Humanos , Ácido Hialurônico/farmacocinética , Nanopartículas de Magnetita/toxicidade
3.
J Clin Invest ; 118(9): 3170-80, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18704196

RESUMO

Iatrogenic tumor cell implantation within surgical wounds can compromise curative cancer surgery. Adhesion of cancer cells, in particular colon cancer cells, is stimulated by exposure to increased extracellular pressure through a cytoskeleton-dependent signaling mechanism requiring FAK, Src, Akt, and paxillin. Mechanical stimuli during tumor resection may therefore negatively impact patient outcome. We hypothesized that perioperative administration of colchicine, which prevents microtubule polymerization, could disrupt pressure-stimulated tumor cell adhesion to surgical wounds and enhance tumor-free survival. Ex vivo treatment of Co26 and Co51 colon cancer cells with colchicine inhibited pressure-stimulated cell adhesion to murine surgical wounds and blocked pressure-induced FAK and Akt phosphorylation. Surgical wound contamination with pressure-activated Co26 and Co51 cells significantly reduced tumor-free survival compared with contamination with tumor cells under ambient pressure. Mice treated with pressure-activated Co26 and Co51 cells from tumors preoperatively treated with colchicine in vivo displayed reduced surgical site implantation and significantly increased tumor-free survival compared with mice exposed to pressure-activated cells from tumors not pretreated with colchicine. Our data suggest that pressure activation of malignant cells promotes tumor development and impairs tumor-free survival and that perioperative colchicine administration or similar interventions may inhibit this effect.


Assuntos
Colchicina/farmacologia , Neoplasias Experimentais/tratamento farmacológico , Cicatrização/efeitos dos fármacos , Animais , Linhagem Celular Tumoral , Intervalo Livre de Doença , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Transplante de Neoplasias , Neoplasias Experimentais/mortalidade , Paxilina/metabolismo , Fosforilação , Pressão , Proteínas Proto-Oncogênicas c-akt/metabolismo , Moduladores de Tubulina/farmacologia , Quinases da Família src/metabolismo
4.
Biochim Biophys Acta ; 1789(2): 88-98, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19059368

RESUMO

TGF-beta and FAK modulate cell migration, differentiation, proliferation and apoptosis, and TGF-beta promotes FAK transcription in intestinal epithelial cells via Smad-dependent and independent pathways. We utilized a 1320 bp FAK promoter-luciferase construct to characterize basal and TGF-beta-mediated FAK gene transcription in IEC-6 cells. Inhibiting JNK or Akt negated TGF-beta-stimulated promoter activity; ERK inhibition did not block the TGF-beta effect but increased basal activity. Co-transfection with Co-Smad4 enhanced the TGF-beta response while the inhibitory Smad7 abolished it. Serial deletions sequentially removing the four Smad binding elements (SBE) in the 5' untranslated region of the promoter revealed that the two most distal SBE's are positive regulators while SBE3 exerts a negative influence. Mutational deletion of two upstream p53 sites enhanced basal but did not affect TGF-beta-stimulated increases in promoter activity. TGF-beta increased DNA binding of Smad4, phospho-Smad2/3 and Runx1/AML1a to the most distal 435 bp containing 3 SBE and 2 AML1a sites by ChIP assay. However, although point mutation of SBE1 ablated the TGF-beta-mediated rise in SV40-promoter activity, mutation of AML1a sites did not. TGF-beta regulation of FAK transcription reflects a complex interplay between positive and negative non-Smad signals and SBE's, the last independent of p53 or AML1a.


Assuntos
Células Epiteliais/metabolismo , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Intestinos/citologia , Proteínas Smad/metabolismo , Fator de Crescimento Transformador beta1/farmacologia , Regiões 5' não Traduzidas/genética , Animais , Western Blotting , Linhagem Celular , Imunoprecipitação da Cromatina , Proteína-Tirosina Quinases de Adesão Focal/genética , Mutagênese Sítio-Dirigida , Regiões Promotoras Genéticas , Ratos , Proteínas Smad/genética , Proteína Smad4/genética , Proteína Smad4/metabolismo , Proteína Smad7/genética , Proteína Smad7/metabolismo
5.
Am J Pathol ; 173(2): 385-99, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18583311

RESUMO

Focal adhesion kinase (FAK) regulates cell migration, proliferation, and apoptosis. FAK protein is reduced at the edge of migrating gut epithelial sheets in vitro, but it has not been characterized in restitutive gut mucosa in vivo. Here we show that FAK and activated phospho-FAK (FAK(397)) immunoreactivity was lower in epithelial cells immediately adjacent to human gastric and colonic ulcers in vivo, but dramatically increased in epithelia near the ulcers, possibly reflecting stimulation by growth factors absent in vitro. Transforming growth factor (TGF)-beta, but not fibroblast growth factor, platelet-derived growth factor, or vascular endothelial growth factor, increased FAK levels in Caco-2 and IEC-6 cells. Epithelial immunoreactivity to TGF-beta and phospho-Smad3 was also higher near the ulcers, varying in parallel with FAK. The TGF-beta receptor antagonist SB431542 completely blocked TGF-beta-induced Smad2/3 and p38 activation in IEC-6 cells. SB431542, the p38 antagonist SB203580, and siRNA-mediated reduction of Smad2 and p38alpha prevented TGF-beta stimulation of both FAK transcription and translation (as measured via a FAK promoter-luciferase construct). FAK(397) levels were directly related to total FAK protein expression. Although gut epithelial motility is associated with direct inhibition of FAK protein adjacent to mucosal wounds, TGF-beta may increase FAK protein near but not bordering mucosal ulcers via Smad2/3 and p38 signals. Our results show that regulation of FAK expression may be as important as FAK phosphorylation in critically influencing gut epithelial cell migration after mucosal injury.


Assuntos
Proteína-Tirosina Quinases de Adesão Focal/biossíntese , Mucosa Intestinal/metabolismo , Proteína Quinase 14 Ativada por Mitógeno/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Proteína Smad2/metabolismo , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta/fisiologia , Animais , Benzamidas/farmacologia , Linhagem Celular , Movimento Celular , Dioxóis/farmacologia , Humanos , Imidazóis/farmacologia , Mucosa Intestinal/citologia , Proteína Quinase 14 Ativada por Mitógeno/antagonistas & inibidores , Fosforilação , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Piridinas/farmacologia , Ratos , Receptor do Fator de Crescimento Transformador beta Tipo I , Receptores de Fatores de Crescimento Transformadores beta/antagonistas & inibidores , Transdução de Sinais , Fator de Crescimento Transformador beta/farmacologia
6.
Cell Tissue Res ; 336(2): 213-23, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19340459

RESUMO

Increasing evidence is available showing the importance of the FAK (focal adhesion kinase) protein level in the migration and homeostasis of intestinal cells. TGFbeta (transforming growth factor beta) modulates FAK protein expression in a complex fashion not only by inducing the activation of p38 and Smad signaling resulting in increased fak promoter activity and increased FAK protein levels, but also by activating ERK (extracellular signal regulated kinases), p38, and the Smad pathway. We show that the blockade of ERK signaling by a specific MEK (MAPK kinase) inhibitor attenuates TGFbeta-induced FAK mRNA stability and reduces FAK protein levels in rat IEC-6 intestinal epithelial cells. The mTOR (mammalian target of rapamycin)-specific inhibitor rapamycin and small interfering RNAs for mTOR and p70(S6) kinase also block TGFbeta-induced FAK protein synthesis. Furthermore, we have found that a TGFbeta-induced increase in wound closures in monolayers of these cells is abolished in the presence ERK or mTOR inhibition. Thus, TGFbeta also modulates FAK protein levels in cultured rat IEC-6 intestinal epithelial cells via ERK activation, acting at the transcriptional level to complement Smad signaling and at on the translational level via the mTOR pathway downstream of ERK, which in turn promotes intestinal epithelial cell migration.


Assuntos
Enterócitos/enzimologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Quinase 1 de Adesão Focal/metabolismo , Biossíntese de Proteínas/efeitos dos fármacos , Proteínas Quinases/metabolismo , Estabilidade de RNA/efeitos dos fármacos , Fator de Crescimento Transformador beta/farmacologia , Animais , Proteínas de Transporte/metabolismo , Células Cultivadas , Enterócitos/efeitos dos fármacos , Flavonoides/farmacologia , Quinase 1 de Adesão Focal/genética , Peptídeos e Proteínas de Sinalização Intracelular , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Fosfoproteínas/metabolismo , Fosforilação/efeitos dos fármacos , Regiões Promotoras Genéticas/genética , Estabilidade Proteica/efeitos dos fármacos , Ratos , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Serina-Treonina Quinases TOR , Transcrição Gênica/efeitos dos fármacos , Cicatrização/efeitos dos fármacos
7.
J Surg Res ; 156(1): 57-63, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19577252

RESUMO

BACKGROUND: Surgical wounds are frequently contaminated by microbes, but rarely become infected if the bacterial burden is low, and irrigation is used to reduce contamination. Wound fluids are low in calcium and high in magnesium. We hypothesized that manipulating irrigant divalent cation concentrations might influence bacterial adhesion. METHODS: Staphylococcus aureus, E. coli, and Pseudomonas aeruginosa were stained with fluorescent calcein AM before plating onto fibroblast monolayers, collagen I, or uncoated bacteriologic plastic. After 1 h, wells were washed with HEPES-buffered pH-balanced sterile water without or with 5 mM CaCl(2), 5 mM MgCl(2), or 1 mM EDTA+EGTA, and the remaining adherent bacteria were assayed fluorometrically. RESULTS: Supplementing the irrigation with magnesium or chelators increased but calcium-supplemented irrigation reduced bacterial adhesion to collagen or fibroblasts. Nonspecific electrostatic bacterial adhesion to uncoated plastic was unaffected by calcium. CONCLUSION: Bacterial adhesion to mammalian cells and matrix proteins is influenced by divalent cations, and pathogenic bacteria may be adapted to adhere under the low calcium high magnesium conditions in wounds. Although these results await confirmation for other bacteria, and in vivo validation and safety-testing, they suggest that supplementing wound irrigation with 5 mM CaCl(2) may reduce bacterial adhesion and subsequent wound infection.


Assuntos
Aderência Bacteriana/efeitos dos fármacos , Cálcio/farmacologia , Cátions Bivalentes/farmacologia , Magnésio/farmacologia , Infecção da Ferida Cirúrgica/prevenção & controle , Animais , Linhagem Celular , Quelantes/farmacologia , Ácido Edético/farmacologia , Ácido Egtázico/farmacologia , Escherichia coli , Camundongos , Plásticos , Pseudomonas aeruginosa , Staphylococcus aureus , Irrigação Terapêutica
8.
J Invest Surg ; 28(4): 202-7, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26268420

RESUMO

AIM: Mature, differentiated enterocytes are essential for normal gut function and critical to recovery from pathological conditions. Little is known about the factors that regulate intestinal epithelial cell differentiation in the adult intestine. The transcription factor, Cdx2, involved in enterocytic differentiation, remains expressed in the adult. Since we have implicated Slfn3 in differentiation in vivo and in vitro, we examined whether it also mediated differentiation in the IEC-Cdx2-L1 cell model of differentiation. MATERIALS AND METHODS: IEC-Cdx2-L1 cells, permanently transfected with Cdx2 under the control of isopropyl-ß-D-thiogalactoside (IPTG), were stimulated to differentiate by 16-day exposure to IPTG. Transcript levels of Cdx2, Slfn 3, and villin were determined by quantitative reverse transcriptase-polymerase chain reaction of mRNA isolated from IPTG-treated and control cells. Slfn3 expression was lowered with specific siRNA to investigate the role of Slfn3 in Cdx2-driven villin expression in IPTG-differentiated cells. RESULTS: Slfn3 and villin expression were significantly greater in IPTG-treated cells. Slfn3 siRNA lowered Slfn3 expression and abolished the IPTG-induced rise in villin expression (p < .05 by ANOVA); Cdx2 expression was unaffected by Slfn3 siRNA. DISCUSSION: The data indicate that the presence of Slfn3 is required for Cdx2 to induce villin expression, and thus differentiation. However, Slfn3 must also promote differentiation of Cdx2 independently since IEC-6 cells that do not normally express Cdx2 can be differentiated by a variety of Slfn3-dependent mechanisms.


Assuntos
Enterócitos/metabolismo , Proteínas de Homeodomínio/fisiologia , Proteínas/fisiologia , Fatores de Transcrição/fisiologia , Animais , Fator de Transcrição CDX2 , Diferenciação Celular , Linhagem Celular , Regulação da Expressão Gênica , Íleo/citologia , Isopropiltiogalactosídeo/farmacologia , Proteínas dos Microfilamentos/biossíntese , Proteínas dos Microfilamentos/genética , Proteínas/genética , Interferência de RNA , RNA Interferente Pequeno/genética , Ratos , Proteínas Recombinantes de Fusão/biossíntese , Transfecção
9.
Metabolism ; 51(3): 380-6, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11887178

RESUMO

Phosphatidylinositol 3-kinase (PI3-K) has been shown to mediate insulin and insulin-like growth factor-1 (IGF-1)-induced nitric oxide (NO) generation and, thus, vascular tone. A role for PI3-K in G-protein-coupled receptor signal transduction has also been reported. As beta2 -adrenergic vascular actions are partly dependent on NO, this study the role of PI3-K on in vitro isoproterenol (Iso)-induced endothelial cell (EC) nitric oxide synthase (NOS) activation and rat aortic vascular relaxation. Cell lysates of rat aortic EC (RAEC), exposed to Iso (10 micromol/L) for 5 minutes, were immunoprecipitated with an antiphosphotyrosine antibody prior to assay for Western blot for the p85-kd regulatory subunit of PI3-K. Endothelial NOS activity was determined by measuring nitrite production. Endothelium-intact aortic rings from male Wistar rats were preincubated with the PI3-K inhibitors, wortmannin (WT), or LY294002 (LY), precontracted with phenylepinephrine (PE), and relaxation to graded doses of Iso was measured. NO contribution to vascular relaxation was assessed by L-N(G)-nitroarginine methyl ester (L-NAME), a NOS inhibitor. Both Iso and IGF-1 induced an increase in p85 subunit phosphorylation as demonstrated by Western analysis, effects inhibited by preincubation with WT. Iso also enhanced association of p85 with the Triton X-100-insoluble fraction of RAEC, reflecting translocation of this enzyme to a cytoskeletal fraction. In addition, Iso as well as IGF-1 significantly increased eNOS activity measured by nitrite production. Both WT and LY markedly inhibited relaxation to Iso, while L-NAME nearly abolished this beta-adrenergic-mediated vasorelaxation. These data indicate that both Iso and IGF-1 activate the EC PI3-K pathway which mediates, in part, the release of NO and subsequent vasorelaxation in response to this beta-agonist Iso as well as to IGF-1.


Assuntos
Agonistas Adrenérgicos beta/farmacologia , Isoproterenol/farmacologia , Óxido Nítrico/biossíntese , Fosfatidilinositol 3-Quinases/fisiologia , Vasodilatação/fisiologia , Androstadienos/farmacologia , Animais , Aorta/citologia , Aorta/fisiologia , Células Cultivadas , Cromonas/farmacologia , Citoesqueleto/metabolismo , Endotélio Vascular/citologia , Endotélio Vascular/fisiologia , Inibidores Enzimáticos/farmacologia , Técnicas In Vitro , Masculino , Morfolinas/farmacologia , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico Sintase Tipo III , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação/efeitos dos fármacos , Ratos , Ratos Wistar , Vasoconstrição/fisiologia , Wortmanina
10.
Am J Surg ; 204(5): 598-601, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22906252

RESUMO

BACKGROUND: Understanding gut development may illuminate the adaptive response to massive small-bowel resection and facilitate enteral nutrition. We reported that Schlafen-3 (Slfn3) mediates differentiation in vitro in rat intestinal epithelial. We hypothesized that Slfn3 is involved in intestinal development in vivo. METHODS: We removed fetal intestines, liver, and lungs on day 20 of gestation, at birth, and on postnatal days 1 and 5. Expression of Slfn3, markers of intestinal differentiation, and Slfn5, to address specificity, were determined by quantitative reverse-transcription polymerase chain reaction. RESULTS: Villin expression increased on days 1 and 5 (8.7 ± .6 and 5.4 ± .4, respectively; P < .01). Intestinal Slfn3 expression was increased substantially after birth (2.1- ± .5-fold) and on days 1 and 5 (P < .02). Slfn3 was higher after birth in liver and lung but decreased sharply thereafter. Slfn5 expression was mostly unchanged. CONCLUSIONS: The data suggest that the developmental/maturation effects we observed correlate with Slfn3 but not Slfn5 and are more relevant to the intestines. A better understanding of how Slfn3 promotes intestinal differentiation could help promote intestinal maturation, improving outcomes in children or adults with short-gut syndrome.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Mucosa Intestinal/metabolismo , Jejuno/metabolismo , Proteínas/metabolismo , Animais , Biomarcadores/metabolismo , Dipeptidil Peptidase 4/metabolismo , Mucosa Intestinal/embriologia , Mucosa Intestinal/crescimento & desenvolvimento , Jejuno/embriologia , Jejuno/crescimento & desenvolvimento , Fígado/embriologia , Fígado/crescimento & desenvolvimento , Fígado/metabolismo , Pulmão/embriologia , Pulmão/crescimento & desenvolvimento , Pulmão/metabolismo , Proteínas dos Microfilamentos/metabolismo , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
11.
Am J Physiol Gastrointest Liver Physiol ; 291(3): G491-9, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16899713

RESUMO

Mucosal healing requires migration and proliferation. Most studies of focal adhesion kinase (FAK), a protein that regulates motility, proliferation, and apoptosis, have focused on rapid phosphorylation. We reported lower FAK protein levels in motile Caco-2 colon cancer cells and postulated that this reduction in FAK available for activation might impact cell migration and mucosal healing. Therefore, total and active FAK (FAK(397)) immunoreactivity was assessed at the migrating fronts of human Caco-2 and rat IEC-6 intestinal epithelial cells. Caco-2 and IEC-6 motility, quantitated as migration into linear or circular wounds, was examined following FAK protein inhibition by small interfering RNA (siRNA). FAK protein stability and mRNA expression were ascertained by cycloheximide decay, RT-PCR, and in situ hybridization in static and migrating Caco-2 cells. Cells at the migrating front of Caco-2 and IEC-6 monolayers exhibited lower immunostaining for both total and activated FAK than cells immediately behind the front. Western blot analysis also demonstrated diminished FAK protein levels in motile cells by >/=30% in both the differential density seeding and multiple scrape models. siRNA FAK protein inhibition enhanced motility in both the linear scrape (20% in Caco-2) and circular wound (16% in Caco-2 and 19% in IEC-6 cells) models. FAK protein degradation did not differ in motile and static Caco-2 cells and was unaffected by FAK(397) phosphorylation, but FAK mRNA was lower in migrating Caco-2 cells. Thus FAK protein abundance appears regulated at the mRNA level during gut epithelial cell motility and may influence epithelial cell migration coordinately with signals that modify FAK phosphorylation.


Assuntos
Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Motilidade Gastrointestinal/fisiologia , Mucosa Intestinal/citologia , Mucosa Intestinal/fisiologia , Animais , Células CACO-2 , Adesão Celular/fisiologia , Movimento Celular/fisiologia , Células HT29 , Humanos , Ratos
12.
J Cell Biochem ; 98(6): 1507-16, 2006 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-16552730

RESUMO

Paxillin is an adapter protein regulating signaling and focal adhesion assembly that has been linked to malignant potential in many malignancies. Overexpression of paxillin has been noted in aggressive tumors. Integrin-mediated binding through the focal adhesion complex is important in metastatic adhesion and is upregulated by extracellular pressure in malignant colonocytes through FAK and Src activation. Neither head and neck cancers nor paxillin have been studied in this regard. We hypothesized that paxillin would play a role in modulating squamous cancer adhesion both at baseline and under conditions of increased extracellular pressure. Using SCC25 tongue squamous cancer cells stably transfected with either an empty selection vector or paxillin expression and selection vectors, we studied adhesion to collagen, paxillin, FAK, and Src expression and phosphorylation in cells maintained for 30 min under ambient or 15 mmHg increased pressure conditions. Paxillin-overexpressing cells exhibited adhesion 121 +/- 2.9% of that observed in vector-only cells (n = 6, P < 0.001) under ambient pressure. Paxillin-overexpression reduced FAK phosphorylation. Pressure stimulated adhesion to 118 +/- 2.3% (n = 6, P < 0.001) of baseline in vector-only cells, similar to its effect in the parental line, and induced paxillin, FAK, and Src phosphorylation. However, increased pressure did not stimulate adhesion or phosphorylate paxillin, FAK, or Src further in paxillin-overexpressing cells. Metastasizing squamous cancer cell adhesiveness may be increased by paxillin-overexpression or by paxillin activation by extracellular pressure during surgical manipulation or growth within a constraining compartment. Targeting paxillin in patients with malignancy and minimal tumor manipulation during surgical resection may be important therapeutic adjuncts.


Assuntos
Quinase 1 de Adesão Focal/metabolismo , Neoplasias de Células Escamosas/metabolismo , Paxilina/metabolismo , Proteínas Proto-Oncogênicas pp60(c-src)/metabolismo , Adesão Celular , Linhagem Celular Tumoral , Humanos , Paxilina/fisiologia , Fosforilação , Pressão , Transdução de Sinais , Transfecção
13.
Cell Physiol Biochem ; 13(3): 135-46, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12876384

RESUMO

BACKGROUND: Upon adhesion, anchorage-dependent cells transmit survival signals from the matrix into the cell. Loss of anchorage leads to anoikis. Resistance to anoikis may influence tumor progression and metastasis. To better understand the pathways that regulate the choice between adhesion and cell death, we examined FAK, c-Src andMAPKinase activities in SW620 human colon cancer cells. METHODS: SW620 cell suspensions were first allowed to adhere to collagen I for 30 minutes and adherent cells were subsequently counted. FAK, p38, c-Src and ERK1/2 phosphorylation were assessed by Western blot in adherent cells and in cells prevented from adhesion by plating unto BSA-pacificated dishes. p38 and FAK were inhibited by SB203580 (20 microM) or by specific FAK antisense nucleotides or FAK siRNA, respectively, and adhesion quantitated. Apoptosis (anoikis) after lack of adhesion was measured colorimetrically in control cells and in cells treated with SB203580. RESULTS: Adhesion to collagen I nearly doubled FAK phosphorylation at Y397, the autophosphorylation site, and decreased p38 activation by 60% (p<0.001) but did not affect FAK phosphorylation at Y576, the c-Src dependent site. Lowering FAK expression with FAK antisense decreased adhesion to collagen I; the larger decrease in FAK expression obtained with the siRNA (43 +/- 2%) resulted in significantly greater inhibition of adhesion not only to collagen I but also to collagen IV and fibronectin. The p38 inhibitor diminished anoikis and enhanced adhesion. Interestingly, the SB compound also significantly inhibited FAK phosphorylation at Y397 (23 +/- 3% in adherent, 30 +/- 4% in non-adherent cells at 30 minutes and 35 +/- 4% in adherent, 46 +/- 14% in non-adherent cells after 6 hours, p<0.05 for each) and greatly enhanced phosphorylation of ERK1/2, a putative anti-apoptotic component of the MAPK cascade. CONCLUSIONS: In the absence of adhesion, SW620 cells exhibit increased p38 but decreased FAK activation, signals that may promote cell death. Our observations with the p38 inhibitor SB203580 indicate that inside-out signals, from p38 to FAK, may regulate both adhesion and anoikis in SW620 cells. In addition, the data suggest the presence of cross-talk between the pro-apoptotic p38 and anti-apoptotic ERK1/2 pathways.


Assuntos
Apoptose/fisiologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas Tirosina Quinases/metabolismo , Transdução de Sinais/fisiologia , Apoptose/efeitos dos fármacos , Proteína Tirosina Quinase CSK , Adesão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Neoplasias do Colo/enzimologia , Neoplasias do Colo/patologia , Neoplasias do Colo/fisiopatologia , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Quinase 1 de Adesão Focal , Proteína-Tirosina Quinases de Adesão Focal , Humanos , Imidazóis/farmacologia , Proteína Quinase 1 Ativada por Mitógeno/efeitos dos fármacos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases Ativadas por Mitógeno/efeitos dos fármacos , Oligonucleotídeos Antissenso/genética , Oligonucleotídeos Antissenso/metabolismo , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/efeitos dos fármacos , Proteínas Tirosina Quinases/genética , Piridinas/farmacologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno , Quinases da Família src
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA