Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Cells ; 12(21)2023 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-37947602

RESUMO

Epithelial and stromal/mesenchymal limbal stem cells contribute to corneal homeostasis and cell renewal. Extracellular vesicles (EVs), including exosomes (Exos), can be paracrine mediators of intercellular communication. Previously, we described cargos and regulatory roles of limbal stromal cell (LSC)-derived Exos in non-diabetic (N) and diabetic (DM) limbal epithelial cells (LECs). Presently, we quantify the miRNA and proteome profiles of human LEC-derived Exos and their regulatory roles in N- and DM-LSC. We revealed some miRNA and protein differences in DM vs. N-LEC-derived Exos' cargos, including proteins involved in Exo biogenesis and packaging that may affect Exo production and ultimately cellular crosstalk and corneal function. Treatment by N-Exos, but not by DM-Exos, enhanced wound healing in cultured N-LSCs and increased proliferation rates in N and DM LSCs vs. corresponding untreated (control) cells. N-Exos-treated LSCs reduced the keratocyte markers ALDH3A1 and lumican and increased the MSC markers CD73, CD90, and CD105 vs. control LSCs. These being opposite to the changes quantified in wounded LSCs. Overall, N-LEC Exos have a more pronounced effect on LSC wound healing, proliferation, and stem cell marker expression than DM-LEC Exos. This suggests that regulatory miRNA and protein cargo differences in DM- vs. N-LEC-derived Exos could contribute to the disease state.


Assuntos
Diabetes Mellitus , Exossomos , Limbo da Córnea , MicroRNAs , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , Limbo da Córnea/metabolismo , Córnea , Diabetes Mellitus/metabolismo , Células Epiteliais/metabolismo , Células Estromais , Comunicação Celular
3.
Cell ; 179(1): 251-267.e24, 2019 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-31539496

RESUMO

In situ transgenesis methods such as viruses and electroporation can rapidly create somatic transgenic mice but lack control over copy number, zygosity, and locus specificity. Here we establish mosaic analysis by dual recombinase-mediated cassette exchange (MADR), which permits stable labeling of mutant cells expressing transgenic elements from precisely defined chromosomal loci. We provide a toolkit of MADR elements for combination labeling, inducible and reversible transgene manipulation, VCre recombinase expression, and transgenesis of human cells. Further, we demonstrate the versatility of MADR by creating glioma models with mixed reporter-identified zygosity or with "personalized" driver mutations from pediatric glioma. MADR is extensible to thousands of existing mouse lines, providing a flexible platform to democratize the generation of somatic mosaic mice. VIDEO ABSTRACT.


Assuntos
Neoplasias Encefálicas/genética , Modelos Animais de Doenças , Marcação de Genes/métodos , Loci Gênicos/genética , Glioma/genética , Mutagênese Insercional/métodos , Transgenes/genética , Animais , Linhagem Celular Tumoral , Feminino , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células-Tronco Neurais/metabolismo , Recombinases/metabolismo , Transfecção
4.
Oncotarget ; 8(5): 7977-7988, 2017 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-28002788

RESUMO

Telomere and Telomerase have recently been explored as anti-aging and anti-cancer drug targets with only limited success. Previously we showed that the Chinese herbal medicine Tianshengyuan-1 (TSY-1), an agent used to treat bone marrow deficiency, has a profound effect on stimulating Telomerase activity in hematopoietic cells. Here, the mechanism of TSY-1 on cellular Telomerase activity was further investigated using HL60, a promyelocytic leukemia cell line, normal peripheral blood mononuclear cells, and CD34+ hematopoietic stem cells derived from umbilical cord blood. TSY-1 increases Telomerase activity in normal peripheral blood mononuclear cells and CD34+ hematopoietic stem cells with innately low Telomerase activity but decreases Telomerase activity in HL60 cells with high intrinsic Telomerase activity, both in a dose-response manner. Gene profiling analysis identified Telomerase reverse transcriptase (TERT) as the potential target gene associated with the TSY-1 effect, which was verified by both RT-PCR and western blot analysis. The ß-galactosidase reporter staining assay showed that the effect of TSY-1 on Telomerase activity correlates with cell senescence. TSY-1 induced hypomethylation within TERT core promoter in HL60 cells but induced hypermethylation within TERT core promoter in normal peripheral blood mononuclear cells and CD34+ hematopoietic stem cells. TSY-1 appears to affect the Telomerase activity in different cell lines differently and the effect is associated with TERT expression, possibly via the methylation of TERT promoter.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Metilação de DNA/efeitos dos fármacos , Medicamentos de Ervas Chinesas/farmacologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Leucemia Promielocítica Aguda/tratamento farmacológico , Leucócitos Mononucleares/efeitos dos fármacos , Telomerase/metabolismo , Telômero/efeitos dos fármacos , Antígenos CD34/metabolismo , Proliferação de Células/efeitos dos fármacos , Senescência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Células HL-60 , Células-Tronco Hematopoéticas/enzimologia , Humanos , Leucemia Promielocítica Aguda/enzimologia , Leucemia Promielocítica Aguda/genética , Leucemia Promielocítica Aguda/patologia , Leucócitos Mononucleares/enzimologia , Regiões Promotoras Genéticas , Telomerase/genética , Telômero/genética , Telômero/metabolismo
5.
Sci Rep ; 5: 17595, 2015 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-26626154

RESUMO

Cell mechanical phenotype or 'mechanotype' is emerging as a valuable label-free biomarker. For example, marked changes in the viscoelastic characteristics of cells occur during malignant transformation and cancer progression. Here we describe a simple and scalable technique to measure cell mechanotype: this parallel microfiltration assay enables multiple samples to be simultaneously measured by driving cell suspensions through porous membranes. To validate the method, we compare the filtration of untransformed and HRas(V12)-transformed murine ovary cells and find significantly increased deformability of the transformed cells. Inducing epithelial-to-mesenchymal transition (EMT) in human ovarian cancer cells by overexpression of key transcription factors (Snail, Slug, Zeb1) or by acquiring drug resistance produces a similar increase in deformability. Mechanistically, we show that EMT-mediated changes in epithelial (loss of E-Cadherin) and mesenchymal markers (vimentin induction) correlate with altered mechanotype. Our results demonstrate a method to screen cell mechanotype that has potential for broader clinical application.


Assuntos
Transição Epitelial-Mesenquimal , Filtração/métodos , Proteínas de Neoplasias/biossíntese , Neoplasias Ovarianas , Ovário , Fatores de Transcrição/biossíntese , Animais , Feminino , Células HL-60 , Humanos , Camundongos , Proteínas de Neoplasias/genética , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Ovário/metabolismo , Ovário/patologia , Fatores de Transcrição/genética
6.
Integr Biol (Camb) ; 6(6): 611-7, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24718685

RESUMO

Changes in cell stiffness (Young's modulus, E), as measured via Atomic Force Microscopy (AFM), is a newly recognized characteristic of cancer cells and may play a role in platinum drug resistance of ovarian cancers. We previously showed that, compared to their syngeneic cisplatin-sensitive counterpart, cisplatin-resistant ovarian cancer cells are stiffer, and this cell stiffness was dependent on actin polymerization and presence of stress fibers. Here, we measured the correlation between Young's modulus (via AFM measurements on live, non-apoptotic cells in physiological buffer) and cisplatin-sensitivity (IC50 as determined via the XTT cell viability assay) in a panel of nine ovarian cancer cell lines representing a range of cisplatin sensitivities. We found that cisplatin-sensitive cells had a lower Young's modulus, compared to cisplatin-resistant cells and resistant cells had a cytoskeleton composed of long actin stress fibers. As Rho GTPase mediates stress fiber formation, we examined the role of Rho GTPase in cell stiffness and platinum resistance. Rho inhibition decreased cell stiffness in cisplatin-resistant CP70 cells and increased their cisplatin sensitivity while Rho activation increased cell stiffness in cisplatin-sensitive A2780 cells and decreased their cisplatin sensitivity. Based on changes in cell stiffness, IC50 and cellular actin stress fiber organization in CP70 and A2780 cells, our findings reveal a direct role of Rho mediated actin remodeling mechanism in cisplatin resistance of ovarian cancer cells. These findings suggest the potential applicability of cell mechanical phenotyping as a model for determining sensitivity of ovarian cancer cells that could have major implications in ovarian cancer diagnosis and personalized medicine.


Assuntos
Antineoplásicos/farmacologia , Cisplatino/farmacologia , Resistencia a Medicamentos Antineoplásicos/fisiologia , Neoplasias Ovarianas/tratamento farmacológico , Proteínas rho de Ligação ao GTP/metabolismo , Actinas/fisiologia , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Cisplatino/uso terapêutico , Módulo de Elasticidade/fisiologia , Feminino , Humanos , Concentração Inibidora 50 , Microscopia de Força Atômica , Neoplasias Ovarianas/enzimologia , Estatísticas não Paramétricas
7.
Anticancer Res ; 32(2): 445-52, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22287731

RESUMO

BACKGROUND: Pharmacological inhibition of the phosphoinositide 3-kinase (PI3K)/Akt pathway prevents G(1) cell cycle progression into S, resulting in G(1) accumulation. The hypothesis that this arrest might negatively impact on chemotherapeutic agents primarily effective in S, G(2) or M-phase was investigated. MATERIALS AND METHODS: Inhibition of PI3K/Akt pathway signaling via LY294002 and Akti-1/2 was demonstrated by immunoblotting. Cell cycle progression was determined by flow cytometric analysis. Cell proliferation was assayed using the XTT cell viability assay. The Chou and Talalay median effect principal was used to evaluate drug interaction. RESULTS: In SKOV3 and IGROV1 human ovarian cancer cells, LY294002 and Akti-1/2 increased the percentage of cells in G(1) and reversed the cell cycle effects of cisplatin, paclitaxel, gemcitabine and topotecan. Pathway blockade synergistically enhanced the cytotoxicity of cisplatin and paclitaxel, but antagonized gemcitabine and topotecan effects. CONCLUSION: Pharmacological PI3K/Akt inhibition antagonizes the efficacy of chemotherapeutic agents primarily effective in the S or G(2)-phase of the cell cycle.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Pontos de Checagem da Fase G1 do Ciclo Celular/efeitos dos fármacos , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/enzimologia , Inibidores de Fosfoinositídeo-3 Quinase , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Benzilaminas/administração & dosagem , Linhagem Celular Tumoral , Cromonas/administração & dosagem , Cisplatino/administração & dosagem , Desoxicitidina/administração & dosagem , Desoxicitidina/análogos & derivados , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Feminino , Humanos , Morfolinas/administração & dosagem , Neoplasias Ovarianas/patologia , Paclitaxel/administração & dosagem , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/administração & dosagem , Proteínas Proto-Oncogênicas c-akt/metabolismo , Quinoxalinas/administração & dosagem , Transdução de Sinais , Topotecan/administração & dosagem , Gencitabina
8.
Nanomedicine ; 8(5): 757-66, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22024198

RESUMO

The exact molecular mechanisms of ovarian cancer platinum resistance are not well understood, and biomarkers to reliably predict ovarian cancer resistance to platinum and other chemotherapeutic agents are lacking. Biomechanics of cisplatin-treated ovarian cancer cells were measured quantitatively at nanoscale level using atomic force microscopy. We demonstrate that cisplatin modulates the cellular nanomechanics of ovarian cancer cells; sensitive cells show dose-dependent increase in cell stiffness, which is effected by disrupting the F-actin polymerization. In contrast, resistant cells show no significant changes in cell stiffness upon cisplatin treatment. Further, stimulated emission depletion, an emerging super-resolution microscopy, shows that at the molecular level, F-actin is indeed remodeled considerably in cisplatin-sensitive and cisplatin-resistant cells. These findings reveal a direct role of the actin remodeling mechanism in cisplatin resistance of ovarian cancer cells, suggesting potential future applications of nanomechanical profiling as a marker for cancer drug sensitivity. FROM THE CLINICAL EDITOR: In this paper, nanomechanical profiling and an emerging super-resolution microscopy method was utilized to decipher the mechanisms of cisplatin resistance in ovarian cancer cells, paving the way to future studies of this and similar other problems with drug resistance in cancer biology.


Assuntos
Citoesqueleto de Actina , Actinas , Cisplatino/farmacologia , Resistencia a Medicamentos Antineoplásicos , Citoesqueleto de Actina/química , Citoesqueleto de Actina/efeitos dos fármacos , Citoesqueleto de Actina/ultraestrutura , Actinas/química , Actinas/ultraestrutura , Linhagem Celular Tumoral , Feminino , Humanos , Microscopia de Força Atômica , Neoplasias Ovarianas/tratamento farmacológico , Polimerização
9.
Clin Cancer Res ; 17(8): 2373-84, 2011 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-21372221

RESUMO

PURPOSE: This study evaluates the effect of dual PI3K and mTOR inhibition using NVP-BEZ235 in preclinical models of ovarian cancer as a potential novel therapeutic strategy. EXPERIMENTAL DESIGN: Inhibition of PI3K/Akt/mTOR signaling by NVP-BEZ235 was demonstrated by immunoblotting. The effect on cell proliferation was assessed in 18 ovarian cancer cell lines, including four pairs of syngeneic cisplatin-sensitive and cisplatin-resistant cell lines. The in vivo effects of NVP-BEZ235 on established tumor growth were evaluated using an immunocompetent, transgenic murine ovarian cancer model (LSL-K-ras(G12D/+)Pten(loxP/loxP)). RESULTS: NVP-BEZ235 decreased cell proliferation in all ovarian cancer cell lines assayed and sensitized cisplatin-resistant cells to the cytotoxic effects of cisplatin. Cell lines with PI3K-activating mutations or Pten deletions were significantly more sensitive to the effect of NVP-BEZ235 than cell lines without these mutations (P < 0.05). A statistically significant correlation was found between relative levels of p4E-BP1 and the IC(50) for NVP-BEZ235. In LSL-K-ras(G12D/+)Pten(loxP/loxP) mice with established intraperitoneal tumor disease, oral administration of NVP-BEZ235 decreased pAkt, p4E-BP1 and Ki67 in tumor tissue, and resulted in significantly longer survival compared to control animals (P < 0.05). NVP-BEZ235 also induced cell cycle arrest, caspase 3 activity, and reduced cell migration. CONCLUSIONS: Targeting PI3K and mTOR simultaneously using NVP-BEZ235 effectively inhibits ovarian cancer cell growth even in the presence of platinum resistance and prolongs survival of mice with intra-abdominal ovarian tumor disease. We propose that dual PI3K and mTOR inhibition using NVP-BEZ235 may be an effective novel therapeutic approach in patients with ovarian cancer.


Assuntos
Imidazóis/uso terapêutico , Neoplasias Ovarianas/tratamento farmacológico , Inibidores de Fosfoinositídeo-3 Quinase , Quinolinas/uso terapêutico , Serina-Treonina Quinases TOR/antagonistas & inibidores , Animais , Antineoplásicos/farmacologia , Caspase 3/metabolismo , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Cisplatino/farmacologia , Modelos Animais de Doenças , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Feminino , Humanos , Immunoblotting , Camundongos , Camundongos Transgênicos , Mutação , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , PTEN Fosfo-Hidrolase/genética , PTEN Fosfo-Hidrolase/metabolismo , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo
10.
Cytoskeleton (Hoboken) ; 67(8): 535-44, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20607860

RESUMO

Targeting of the PI3K (phosphoinositide3-kinase)/Akt/mTOR pathway in human ovarian cancer cells is a promising novel therapeutic strategy. We investigated the effects of cisplatin and the PI3K inhibitor LY294002 on invasion, migration and the expression of essential matrix metalloproteinases (MMPs) in ovarian cancer cells. SKOV3, OVCAR5 and IGROV1 human ovarian cancer cell lines were treated with cisplatin, LY294002 and a combination of both drugs. Invasion and migration of treated cells was assessed using Matrigel and uncoated PET membrane assays. Expression levels of pro-MMP2, MMP2, TIMP1, TIMP2 and MT1-MMP were determined using Western Blotting. Gel zymography was used to quantitate the functional levels of active MMP2. All three cell lines showed significantly reduced invasion and migration after treatment with cisplatin, LY294002, and the combination of both drugs compared to untreated controls. In SKOV3 cells, cisplatin alone and in combination with LY294002 resulted in a 6.3 and 7.1-fold reduction in the total amount of activated MMP2. TIMP1 expression decreased by 5.0, 6.6 and 28.4-fold with cisplatin, LY294002 and the combination respectively (P < 0.05). In contrast, only cisplatin and the combination of both drugs resulted in a significant, 3.7 and 5.1-fold reduction in the level of TIMP2. Expression levels of MT1-MMP remained unchanged. These observations were corroborated in IGROV1 cell lines that showed similar changes of activated MMP2 and TIMP2 expression, but no significant decrease in TIMP1 levels. Our data suggests that inhibition of ovarian cancer cell motility is mediated via down-regulation of activated MMP2, TIMP1 and TIMP2 expression under these treatment conditions.


Assuntos
Cromonas/farmacologia , Cisplatino/farmacologia , Metaloproteinase 2 da Matriz/biossíntese , Morfolinas/farmacologia , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/enzimologia , Inibidores de Fosfoinositídeo-3 Quinase , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Feminino , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Neoplasias Ovarianas/patologia , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR , Inibidor Tecidual de Metaloproteinase-1/biossíntese , Inibidor Tecidual de Metaloproteinase-2/biossíntese
11.
Cell Signal ; 19(6): 1348-57, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17307332

RESUMO

While a great deal of attention has been focused on G-protein-coupled receptor (GPCR)-induced epidermal growth factor receptor (EGFR) transactivation, it has been known for many years that the tyrosine kinase activity of the EGFR is inhibited in cells treated with tumor-promoting phorbol esters, a process termed EGFR transmodulation. Because many GPCR agonists that elicit EGFR transactivation also stimulate the Gq/phospholipase C (PLC)/protein kinase C (PKC) pathway, we hypothesized that PKC-mediated inhibition of EGFR transactivation operates physiologically as a feedback loop that regulates the intensity and/or duration of GPCR-elicited EGFR transactivation. In support of this hypothesis, we found that treatment of intestinal epithelial IEC-18 cells with the PKC inhibitors GF 109203X or Ro 31-8220 or chronic exposure of these cells to phorbol-12,13-dibutyrate (PDB) to downregulate PKCs, markedly enhanced the increase in EGFR tyrosine phosphorylation induced by angiotensin II or vasopressin in these cells. Similarly, PKC inhibition enhanced EGFR transactivation in human colonic epithelial T84 cells stimulated with carbachol, as well as in bombesin-stimulated Rat-1 fibroblasts stably transfected with the bombesin receptor. Furthermore, cell treatment with inhibitors with greater specificity towards PKCalpha, including Gö6976, Ro 31-7549 or Ro 32-0432, also increased GPCR-induced EGFR transactivation in IEC-18, T84 and Rat-1 cells. Transfection of siRNAs targeting PKCalpha also enhanced bombesin-induced EGFR tyrosine phosphorylation in Rat-1 cells. Thus, multiple lines of evidence support the hypothesis that conventional PKC isoforms, especially PKCalpha, mediate feedback inhibition of GPCR-induced EGFR transactivation.


Assuntos
Receptores ErbB/genética , Retroalimentação Fisiológica , Proteína Quinase C-alfa/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Ativação Transcricional/genética , Angiotensina II/farmacologia , Animais , Linhagem Celular , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Retroalimentação Fisiológica/efeitos dos fármacos , Humanos , Indóis/farmacologia , Maleimidas/farmacologia , Fosfotirosina/metabolismo , Proteína Quinase C-alfa/antagonistas & inibidores , Ratos , Ativação Transcricional/efeitos dos fármacos
12.
Am J Physiol Gastrointest Liver Physiol ; 288(2): G182-94, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15358595

RESUMO

The role of epidermal growth factor receptor (EGFR) tyrosine kinase and its downstream targets in the regulation of the transition from the G0/G1 phase into DNA synthesis in response to ANG II has not been previously investigated in intestinal epithelial IEC-18 cells. ANG II induced a rapid and striking EGFR tyrosine phosphorylation, which was prevented by selective inhibitors of EGFR tyrosine kinase activity (e.g., AG-1478) or by broad-spectrum matrix metalloproteinase (MMP) inhibitor GM-6001. Pretreatment of these cells with either AG-1478 or GM-6001 reduced ANG II-stimulated DNA synthesis by approximately 50%. To elucidate the downstream targets of EGFR, we demonstrated that ANG II stimulated phosphorylation of Akt at Ser473, mTOR at Ser2448, p70S6K1 at Thr389, and S6 ribosomal protein at Ser(235/236). Pretreatment with AG-1478 inhibited Akt, p70S6K1, and S6 ribosomal protein phosphorylation. Inhibition of phosphatidylinositol (PI)3-kinase with LY-294002 or mTOR/p70S6K1 with rapamycin reduced [3H]thymidine incorporation by 50%, i.e., to levels comparable to those achieved by addition of either AG-1478 or GM-6001. Utilizing Akt small-interfering RNA targeted to Akt1 and Akt2, Akt protein knockdown dramatically inhibited p70S6K1 and S6 ribosomal protein phosphorylation. In contrast, AG-1478 or Akt gene silencing exerted no detectable inhibitory effect on ANG II-induced extracellular signal-regulated kinase 1/2 phosphorylation in IEC-18 cells. Taken together, our results demonstrate that EGFR transactivation mediates ANG II-stimulated mitogenesis through the PI3-kinase/Akt/mTOR/p70S6K1 signaling pathway in IEC-18 cells.


Assuntos
Angiotensina II/fisiologia , Receptores ErbB/fisiologia , Mucosa Intestinal/citologia , Fosfotransferases (Aceptor do Grupo Álcool)/fisiologia , Ativação Transcricional/fisiologia , Animais , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , DNA/biossíntese , Inativação Gênica , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/fisiologia , Fosfatidilinositol 3-Quinases/fisiologia , Proteínas Quinases/fisiologia , Proteínas Serina-Treonina Quinases/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Proteínas Proto-Oncogênicas c-akt , Proteínas Quinases S6 Ribossômicas 70-kDa/fisiologia , Transdução de Sinais , Sirolimo/farmacologia , Serina-Treonina Quinases TOR
13.
Biochem Biophys Res Commun ; 318(4): 826-32, 2004 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-15147945

RESUMO

The binding of bombesin to its cognate G-protein coupled receptor stimulates quiescent Swiss 3T3 cells to re-initiate DNA synthesis and cell division. Addition of a non-mitogenic concentration of insulin dramatically potentiates bombesin-induced cell proliferation. We examined whether bombesin-induced EGFR transactivation mediates synergistic cell proliferation induced by bombesin and insulin. Treatment with selective EGFR tyrosine kinase inhibitors blocked EGFR transactivation, DNA synthesis, the transition of cells from quiescence into the cell cycle, and the expression of cyclins D1 and E induced by bombesin alone. In contrast, the inhibitors prevented cell cycle progression to a much lesser degree in cells stimulated with the combination of bombesin and insulin. Our results indicate that EGFR transactivation does not mediate synergistic cell proliferation induced by bombesin and insulin, and imply that insulin compensates for the requirement for EGFR transactivation in bombesin-induced DNA synthesis.


Assuntos
Bombesina/farmacologia , DNA/biossíntese , Receptores ErbB/metabolismo , Insulina/farmacologia , Animais , Sinergismo Farmacológico , Inibidores Enzimáticos/farmacologia , Fator de Crescimento Epidérmico/farmacologia , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/genética , Camundongos , Proteínas Tirosina Fosfatases/antagonistas & inibidores , Quinazolinas , Células Swiss 3T3 , Timidina/metabolismo , Ativação Transcricional/efeitos dos fármacos , Trítio , Tirfostinas/antagonistas & inibidores , Tirfostinas/farmacologia
14.
Exp Cell Res ; 290(2): 437-46, 2003 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-14568001

RESUMO

Matrix metalloproteinases (MMPs) have been implicated in the transactivation of the epidermal growth factor receptor (EGFR) induced by G-protein coupled receptor (GPCR) agonists. Although EGFR phosphorylation and downstream signaling have been shown to be dependent on MMP activity in many systems, a role for MMPs in GPCR-induced DNA synthesis has not been studied in any detail. In this study we utilized the broad-spectrum matrix metalloproteinase inhibitor, galardin (Ilomastat, GM 6001), to study the mechanism of bombesin- or LPA-induced EGFR transactivation and the role of MMPs in early and late response mitogenic signaling in Rat-1 cells stably transfected with the bombesin/GRP receptor (BoR-15 cells). Addition of galardin to cells stimulated with bombesin or LPA specifically inhibited total EGFR phosphorylation, as well as site-specific phosphorylation of tyrosine 845, a putative Src phosphorylation site, and tyrosine 1068, a typical autophosphorylation site. Galardin treatment also inhibited extracellular signal-regulated kinase (ERK) activation induced by bombesin or LPA, but not by EGF. In addition, galardin inhibited bombesin- or LPA-induced DNA synthesis in a dose dependent manner, when stimulated by increasing concentrations of bombesin, and when added after bombesin stimulation. Furthermore, addition of galardin post-bombesin stimulation indicated that by 3 h sufficient accumulation of EGFR ligands had occurred to continue to induce transactivation despite an inhibition of MMP activity. Taken together, our results suggest that MMPs act as early as 5 min, and up to around 3 h, to mediate GPCR-induced EGFR transactivation, ERK activation, and stimulation of DNA synthesis.


Assuntos
Bombesina/antagonistas & inibidores , DNA/biossíntese , Dipeptídeos/farmacologia , Receptores ErbB/metabolismo , Lisofosfolipídeos/antagonistas & inibidores , Inibidores de Metaloproteinases de Matriz , Ativação Transcricional , Animais , Bombesina/farmacologia , Divisão Celular , Células Cultivadas , Ativação Enzimática , Lisofosfolipídeos/farmacologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosforilação , Ratos , Receptores da Bombesina/genética , Transdução de Sinais , Transfecção , Tirosina/metabolismo
15.
Cancer Res ; 63(10): 2379-87, 2003 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-12750255

RESUMO

Neuropeptides and their corresponding G protein-coupled receptors are increasingly implicated in the autocrine/paracrine stimulation of growth of human cancers. Using K-Ras mutated human pancreatic ductal adenocarcinoma cell line PANC-1 as a model system, we demonstrate that neurotensin (NT) induced translocation of phosphorylated extracellular signal-regulated kinases (ERK-1 and ERK-2) to the nucleus, rapid dose-dependent activation of dual-specificity mitogen and ERK-1 and ERK-2 kinase-1/2 (MEK-1/2), and striking stimulation of c-Raf-1 but not pan-Ras. Furthermore, treatment of PANC-1 cells with protein kinase C (PKC) inhibitors, GF-1 and Ro 31-8220, completely abrogated NT-induced ERK-1 and ERK-2 activation, and significantly attenuated NT-induced c-Raf-1 stimulation. Interestingly, NT did not stimulate epidermal growth factor receptor transactivation, and epidermal growth factor receptor tyrosine kinase or Src inhibitors did not affect NT-induced ERK activation in PANC-1 cells. Our results indicate that NT potently stimulates c-Raf-1-MEK-ERK in PANC-1 cells through a PKC-dependent signaling pathway. Furthermore, we show that NT-induced DNA synthesis in PANC-1 cells is ERK-dependent. Finally, we demonstrate that NT stimulated clonal growth of PANC-1 cells in semisolid medium, which is abrogated by both GF-1 and the MEK-1/2 inhibitor, U0126. Collectively our results suggest that PKC-mediated stimulation of ERK-1 and ERK-2 play a pivotal role in NT-induced growth of PANC-1 cells harboring activating K-Ras mutation.


Assuntos
Carcinoma Ductal Pancreático/enzimologia , Neurotensina/farmacologia , Neoplasias Pancreáticas/enzimologia , Proteína Quinase C/metabolismo , Butadienos/farmacologia , Carcinoma Ductal Pancreático/patologia , Divisão Celular/efeitos dos fármacos , Divisão Celular/fisiologia , Núcleo Celular/enzimologia , DNA de Neoplasias/biossíntese , DNA de Neoplasias/genética , Proteínas de Ligação a DNA/farmacologia , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Receptores ErbB/genética , Receptores ErbB/metabolismo , Fatores de Ligação de DNA Eritroide Específicos , Quinase 2 de Adesão Focal , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/fisiologia , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Nitrilas/farmacologia , Neoplasias Pancreáticas/patologia , Fosforilação/efeitos dos fármacos , Proteína Quinase C/antagonistas & inibidores , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-raf/metabolismo , Fatores de Transcrição/farmacologia , Ativação Transcricional , Células Tumorais Cultivadas , Quinases da Família src/metabolismo
16.
Artigo em Inglês | MEDLINE | ID: mdl-12620889

RESUMO

PKC, a major target for the tumor-promoting phorbol esters, has been implicated in the signal transduction pathways that mediate important functions in intestinal epithelial cells, including proliferation and carcinogenesis. With the use of IEC-18 cells arrested in G0/G1, addition of phorbol esters resulted in a modest increase in [3H]thymidine incorporation and a slight shift toward the S and G2/M phases of the cell cycle, whereas the combination of EGF and phorbol 12,13-dibutyrate (PDB) synergistically stimulated DNA synthesis. To investigate the effects of receptor-mediated PKC activation on mitogenesis, we demonstrated that ANG II induced ERK activation, a response completely blocked by pretreatment with mitogen/extracellular signal-regulated kinase inhibitors or specific PKC inhibitors. Furthermore, ANG II stimulated an over threefold increase in [3H]thymidine incorporation that was corroborated by flow cytometric analysis of the cell cycle to levels comparable to that achieved by the combination of EGF and PDB. Taken together, our results indicate that receptor-mediated PKC activation, as induced by ANG II, transduces mitogenic signals leading to DNA synthesis and cell proliferation in IEC-18 cells.


Assuntos
Angiotensina II/farmacologia , Células Epiteliais/citologia , Mucosa Intestinal/citologia , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase C/metabolismo , Vasoconstritores/farmacologia , Animais , Carcinógenos/farmacologia , Divisão Celular/efeitos dos fármacos , Linhagem Celular , DNA/biossíntese , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/enzimologia , Fase G1/efeitos dos fármacos , Fase G1/fisiologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/fisiologia , Proteína Quinase 3 Ativada por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Dibutirato de 12,13-Forbol/farmacologia , Ratos , Fase de Repouso do Ciclo Celular/efeitos dos fármacos , Fase de Repouso do Ciclo Celular/fisiologia , Fase S/efeitos dos fármacos , Fase S/fisiologia
17.
J Cell Physiol ; 191(3): 351-61, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12012331

RESUMO

Although the migration of hepatic myofibroblasts (HMFs) contributes to the development of fibrosis, the signals regulating migration of these cells are poorly understood. In this study, we tested the hypothesis that HMF migration is stimulated by platelet-derived growth factor-BB (PDGF-BB) through p38 mitogen-activated protein (MAP) kinase and extracellular signal-regulated kinase (ERK) signaling pathways. This hypothesis was addressed by directly visualizing the migration of cultured human HMFs into a wound. PDGF-BB stimulated membrane ruffling, migration, and proliferation. PDGF-BB also induced activation of p38 MAP kinase, its downstream effector, heat shock protein (HSP) 27, ERK 1 and ERK 2, and p125 focal adhesion kinase (FAK). Selective antagonism of p38 MAP kinase blocked PDGF-BB-stimulated HSP 27 phosphorylation, membrane ruffling, and migration, but did not alter PDGF-BB-induced proliferation. Selective antagonism of ERK kinase inhibited PDGF-BB-induced ERK phosphorylation and proliferation, but did not affect PDGF-BB-stimulated migration. Concentrations of PDGF-BB that stimulated migration and proliferation did not influence myosin-dependent contractility. Neither selective inhibition of p38 MAP kinase nor ERKs altered PDGF-BB-induced activation of FAK. In conclusion, these results provide novel evidence indicating that (1) HMF migration is stimulated by PDGF-BB through the regulation of membrane ruffling by a p38 MAP kinase signaling pathway, (2) whereas p38 MAP kinase mediates PDGF-BB-stimulated migration, but not proliferation, ERKs mediate PDGF-induced proliferation, but not migration, and (3) increases in myosin-dependent contractility are not required for PDGF-BB-stimulated migration.


Assuntos
Fibroblastos/fisiologia , Fígado/fisiologia , Proteínas Quinases Ativadas por Mitógeno/fisiologia , Músculo Liso/fisiologia , Fator de Crescimento Derivado de Plaquetas/farmacologia , Becaplermina , Divisão Celular/fisiologia , Membrana Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Ativação Enzimática/fisiologia , Quinase 1 de Adesão Focal , Proteína-Tirosina Quinases de Adesão Focal , Humanos , Fígado/citologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Músculo Liso/citologia , Miosinas/fisiologia , Proteínas Tirosina Quinases/fisiologia , Proteínas Proto-Oncogênicas c-sis , Proteínas Quinases p38 Ativadas por Mitógeno
18.
Am J Physiol Cell Physiol ; 282(3): C434-50, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11832328

RESUMO

The role of G protein-coupled receptors and their ligands in intestinal epithelial cell signaling and proliferation is poorly understood. Here, we demonstrate that arginine vasopressin (AVP) induces multiple intracellular signal transduction pathways in rat intestinal epithelial IEC-18 cells via a V(1A) receptor. Addition of AVP to these cells induces a rapid and transient increase in cytosolic Ca(2+) concentration and promotes protein kinase D (PKD) activation through a protein kinase C (PKC)-dependent pathway, as revealed by in vitro kinase assays and immunoblotting with an antibody that recognizes autophosphorylated PKD at Ser(916). AVP also stimulates the tyrosine phosphorylation of the nonreceptor tyrosine kinase proline-rich tyrosine kinase 2 (Pyk2) and promotes Src family kinase phosphorylation at Tyr(418), indicative of Src activation. AVP induces extracellular signal-related kinase (ERK)-1 (p44(mapk)) and ERK-2 (p42(mapk)) activation, a response prevented by treatment with mitogen-activated protein kinase kinase (MEK) inhibitors (PD-98059 and U-0126), specific PKC inhibitors (GF-I and Ro-31-8220), depletion of Ca(2+) (EGTA and thapsigargin), selective epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (tyrphostin AG-1478, compound 56), or the selective Src family kinase inhibitor PP-2. Furthermore, AVP acts as a potent growth factor for IEC-18 cells, inducing DNA synthesis and cell proliferation through ERK-, Ca(2+)-, PKC-, EGFR tyrosine kinase-, and Src-dependent pathways.


Assuntos
Arginina Vasopressina/metabolismo , Arginina Vasopressina/farmacologia , Células Epiteliais/metabolismo , Mucosa Intestinal/metabolismo , Transdução de Sinais/fisiologia , Animais , Cálcio/metabolismo , Divisão Celular/efeitos dos fármacos , Divisão Celular/fisiologia , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Meios de Cultura Livres de Soro , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Células Epiteliais/efeitos dos fármacos , Receptores ErbB/metabolismo , Quinase 2 de Adesão Focal , Mucosa Intestinal/citologia , MAP Quinase Quinase 1 , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosforilação , Proteína Quinase C/metabolismo , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Tirosina Quinases/metabolismo , Ratos , Receptores de Vasopressinas/metabolismo , Quinases da Família src/antagonistas & inibidores , Quinases da Família src/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA