Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
1.
Oncol Rep ; 35(6): 3593-9, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27004682

RESUMO

Although previous studies have shown that Ras-ERK signaling in mitosis is closed due to the inhibition of signal transduction, the events involved in the molecular mechanisms are still unclear. In the present study, we investigated the Ras-ERK signaling pathway in mitotic COS7 cells. The results demonstrated that treatment with epidermal growth factor (EGF) failed to increase the endocytosis of EGF-EGFR (EGF receptor) complexes in mitotic COS7 cells, although a large amount of endosomes were found in asynchronous COS7 cells. Clathrin expression levels in mitotic COS7 cells were inhibited whereas caveolin expression levels in mitotic COS7 cells were almost unaffected. Y1068 and Y1086 residues of EGFR in the mitotic COS7 cells were activated. However, Grb2 and Shc in the mitotic COS7 cells did not bind to activated EGFR. Ras activity was inhibited in the mitotic COS7 cells whereas its downstream protein, Raf, was obviously phosphorylated by EGF in mitosis. Treatment with phorbol 12-myristate 13-acetate (PMA) also increased the phosphorylation levels of Raf in the mitotic COS7 cells. Nevertheless, Raf phosphorylation in mitosis was significantly inhibited by AG1478. Lastly, activation of EGF-mediated MEK and ERK in the mitotic COS7 cells was obviously inhibited. In summary, our results suggest that the Ras-ERK pathway is inhibited in mitotic COS7 cells which may be the dual result of the difficulty in the transduction of EGF signaling by EGFR or Raf to downstream proteins.


Assuntos
Fator de Crescimento Epidérmico/metabolismo , Receptores ErbB/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Proteínas Proto-Oncogênicas c-raf/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Quinases raf/metabolismo , Animais , Células COS , Caveolinas/biossíntese , Linhagem Celular , Chlorocebus aethiops , Clatrina/biossíntese , Endocitose/fisiologia , Endossomos/fisiologia , Proteína Adaptadora GRB2/metabolismo , Sistema de Sinalização das MAP Quinases , Nocodazol/farmacologia , Fosforilação/efeitos dos fármacos , Quinazolinas/farmacologia , Proteínas Adaptadoras da Sinalização Shc/metabolismo , Acetato de Tetradecanoilforbol/farmacologia , Tirfostinas/farmacologia
2.
Eur J Cell Biol ; 90(8): 678-86, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21616554

RESUMO

Cellular senescence represents a powerful tumor suppressor mechanism to prevent proliferation and invasion of malignant cells. Since tumor cells as well as primary fibroblasts lacking the lysosomal cysteine-type carboxypeptidase cathepsin X exhibit a reduced invasive capacity, we hypothesized that the underlying reason may be the induction of cellular senescence. To investigate the cellular and molecular mechanisms leading to diminished migration/invasion of cathepsin X-deficient cells, we have analyzed murine embryonic fibroblasts (MEF) derived from cathepsin X-deficient mice and neonatal human dermal fibroblasts (NHDF) transfected with siRNAs targeting cathepsin X. Remarkably, both cell types exhibited a flattened and enlarged cell body, a characteristic phenotype of senescent cells. Additional evidence for accelerated senescence was obtained by detection of the common senescence marker ß-galactosidase. Further examination revealed increased expression levels of senescence-associated genes such as p16, p21, p53, and caveolin in these cells along with a reduced proliferation rate. The accelerated cellular senescence induced by cathepsin X deficiency was rescued by simultaneous expression of exogenous cathepsin X. Finally, cell cycle analysis confirmed a marked reduction of the synthesis rate and prolongation of the S-phase, while susceptibility to apoptosis of cathepsin X-deficient cells remained unchanged. In conclusion, cathepsin X deficiency leads to accelerated cellular senescence and consequently to diminished cellular proliferation and migration/invasion implying a potential role of cathepsin X in bypassing cellular senescence.


Assuntos
Catepsinas/biossíntese , Senescência Celular , Animais , Apoptose/genética , Catepsinas/genética , Caveolinas/biossíntese , Caveolinas/genética , Ciclo Celular , Proliferação de Células , Células Cultivadas , Inibidor p16 de Quinase Dependente de Ciclina/biossíntese , Inibidor p16 de Quinase Dependente de Ciclina/genética , Inibidor de Quinase Dependente de Ciclina p21/biossíntese , Inibidor de Quinase Dependente de Ciclina p21/genética , Regulação para Baixo , Ensaio de Imunoadsorção Enzimática , Fibroblastos/citologia , Fibroblastos/metabolismo , Humanos , Camundongos , Camundongos Knockout , Reação em Cadeia da Polimerase , Interferência de RNA , RNA Interferente Pequeno , Proteína Supressora de Tumor p53/biossíntese , Proteína Supressora de Tumor p53/genética , beta-Galactosidase/análise
3.
Tumour Biol ; 32(4): 787-99, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21584795

RESUMO

We aimed to investigate the expression of caveolin-1, -2, -3, and platelet-derived growth factor (PDGF) ß receptor in breast cancer cells and stroma by immunohistochemistry and to analyze their implications. The expression rates of stromal caveolin-2 and PDGF ß receptor increased as the tumor progressed from ductal carcinoma in situ to microinvasive ductal carcinoma, intraductal component of invasive ductal carcinoma (IDC), and IDC (p<0.001). The expression loss of caveolin-1 in tumor stroma of IDC correlated with high tumor stage (p<0.001), high nodal stage (p=0.011), high cancer stage (p=0.005), estrogen receptor negativity (p=0.003), and tumor recurrence (p=0.003). In addition, the expression loss of caveolin-1 in tumor stroma was correlated with a shorter disease-free survival and an overall survival (p<0.001). In conclusion, the loss of stromal caveolin-1 is related to poor prognosis in IDC.


Assuntos
Biomarcadores Tumorais/análise , Neoplasias da Mama/metabolismo , Carcinoma Ductal de Mama/metabolismo , Carcinoma Intraductal não Infiltrante/metabolismo , Caveolina 1/biossíntese , Microambiente Tumoral/fisiologia , Adulto , Biomarcadores Tumorais/metabolismo , Neoplasias da Mama/mortalidade , Neoplasias da Mama/patologia , Carcinoma Ductal de Mama/mortalidade , Carcinoma Ductal de Mama/patologia , Carcinoma Intraductal não Infiltrante/mortalidade , Carcinoma Intraductal não Infiltrante/patologia , Caveolinas/biossíntese , Progressão da Doença , Feminino , Humanos , Imuno-Histoquímica , Estimativa de Kaplan-Meier , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Fator de Crescimento Derivado de Plaquetas/biossíntese , Prognóstico , Análise Serial de Tecidos
4.
Lab Invest ; 89(6): 614-23, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19333235

RESUMO

Here we review the clinical and translational implications of the caveolin gene family for understanding the pathogenesis of human diseases, including breast and prostate cancers, pulmonary hypertension, cardiomyopathy, diabetes, and muscular dystrophy. Detailed phenotypic analysis of caveolin knockout mice has served to highlight the crucial role of a caveolin deficiency in the pathogenesis of many human disease processes. Mutations in the human caveolin genes are associated with a number of established genetic disorders (such as breast cancer, lipodystrophy, muscular dystrophy, and cardiomyopathy), making the caveolins important and novel targets for drug development. The implementation of new strategies for caveolin replacement therapy-including caveolin mimetic peptides-is ongoing.


Assuntos
Caveolinas/fisiologia , Células-Tronco Adultas/metabolismo , Animais , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Cardiomiopatias/metabolismo , Cavéolas/metabolismo , Caveolinas/biossíntese , Caveolinas/genética , Diabetes Mellitus/tratamento farmacológico , Diabetes Mellitus/metabolismo , Humanos , Hipertensão Pulmonar/tratamento farmacológico , Hipertensão Pulmonar/metabolismo , Masculino , Camundongos , Distrofias Musculares/tratamento farmacológico , Distrofias Musculares/metabolismo , Mutação , Peptídeos/uso terapêutico , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/metabolismo , Transdução de Sinais
5.
J Biol Chem ; 283(36): 24659-72, 2008 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-18566454

RESUMO

Determining the role of lipid raft nanodomains in G protein-coupled receptor signaling remains fraught by the lack of assays directly monitoring rafts in native membranes. We thus combined extensive biochemical and pharmacological approaches to a nanoscale strategy based on bioluminescence resonance energy transfer (BRET) to assess the spatial and functional influence of cholesterol-rich liquid-ordered lipid nanodomains on beta2 adrenergic receptor (beta2AR) signaling. The data revealed that whereas beta2AR did not partition within liquid-ordered lipid phase, a pool of G protein and adenylyl cyclase (AC) were sequestered in these domains. Destabilization of the liquid-ordered phase by cholesterol depletion led to a lateral redistribution of Galphas and AC that favored interactions between the receptor and its signaling partners as assessed by BRET. This resulted in an increased basal and agonist-promoted beta2AR-stimulated cAMP production that was partially dampened as a result of constitutive protein kinase A-dependent phosphorylation and desensitization of the receptor. This restraining influence of nanodomains on beta2AR signaling was further substantiated by showing that liquid-ordered lipid phase stabilization using caveolin overexpression or increasing membrane cholesterol amount led to an inhibition of beta2AR-associated signaling. Given the emerging concept that clustering of receptors and effectors into signaling platforms contributes to the efficacy and selectivity of signal transduction, our results support a model whereby cholesterol-promoted liquid-ordered lipid phase-embedding Gs and AC allows their lateral separation from the receptor, thus restraining the basal activity and controlling responsiveness of beta2AR signaling machinery within larger signaling platforms.


Assuntos
Colesterol/metabolismo , Microdomínios da Membrana/metabolismo , Modelos Biológicos , Receptores Adrenérgicos beta 2/metabolismo , Transdução de Sinais/fisiologia , Adenilil Ciclases/metabolismo , Agonistas Adrenérgicos/farmacologia , Caveolinas/biossíntese , Linhagem Celular , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Subunidades alfa de Proteínas de Ligação ao GTP/metabolismo , Humanos , Fosforilação/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
6.
J Neurosci ; 27(37): 9941-50, 2007 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-17855608

RESUMO

It has become widely accepted that along with its ability to directly regulate gene expression, estradiol also influences cell signaling and brain function via rapid membrane-initiated events. Many of these novel signaling processes are dependent on estrogen receptors (ERs) localized to the neuronal membrane. However, the mechanism(s) by which ERs are able to trigger cell signaling when targeted to the neuronal membrane surface has yet to be determined. In hippocampal neurons, we find that caveolin proteins are essential for the regulation of CREB (cAMP response element-binding protein) phosphorylation after estradiol activation of metabotropic glutamate receptor (mGluR) signaling. Furthermore, caveolin-1 (CAV1) and CAV3 differentially regulate the ability of estradiol to activate two discrete signaling pathways. ER alpha activation of mGluR1a is dependent on CAV1, whereas CAV3 is necessary for ER alpha and ER beta activation of mGluR2/3. These results are consistent with previous reports in non-neuronal cells, implicating the importance of caveolin proteins in rapid estrogen signaling. In addition, the functional isolation of distinct estrogen-sensitive signaling pathways by different caveolin proteins suggests novel mechanisms through which the membrane-initiated effects of estradiol are orchestrated.


Assuntos
Caveolinas/biossíntese , Membrana Celular/metabolismo , Neurônios/metabolismo , Receptores de Estrogênio/fisiologia , Animais , Caveolinas/agonistas , Caveolinas/genética , Membrana Celular/efeitos dos fármacos , Membrana Celular/genética , Células Cultivadas , Estradiol/farmacologia , Feminino , Neurônios/efeitos dos fármacos , Ratos , Receptores de Estrogênio/agonistas , Receptores de Estrogênio/biossíntese , Receptores de Estrogênio/genética
7.
Mol Biol (Mosk) ; 41(6): 994-1001, 2007.
Artigo em Russo | MEDLINE | ID: mdl-18318117

RESUMO

Oxidative stimulation induced by hydrogen peroxide (H2O2) on human epithelial cells (HLECs) was performed to observe the effects on cell viability, caveolin expression, and cholesterol depletion in HLECs caused by methyl-beta-cyclodextrin (MbetaCD) was also studied. SRA01/04 HLECs were exposed to H2O2 or MbetaCD of various concentrations and durations. We used a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay to measure the effect of H2O2 on the viability of SRA01/04 HLECs. The distributions of caveolins after oxidative stimulation were probed by fluorescence microscopy and laser scanning confocal microscopy. Immunoblotting was performed to analyze alterations of caveolins expression. We observed that the viability of SRA01/04 HLECs under 0.1 mM H2O2 for 10 min or longer was significantly reduced (*p < 0.05, F = 11.63). Laser scanning microscopy showed immunofluorescent caveolins in SRA01/04 HLECs under 0.1 mM H2O2 for 10 min or longer, caveolins were largely confined to intracellular domains. Western blots showed both membrane and total caveolin protein (22 kDa) levels in SRA01/04 HLECs treated with 0.1, 0.2, 0.5 or 1.0 mM H2O2 for 30 min were significantly reduced, compared with the untreated (*p < 0.05, F = 6.149, or *p < 0.05, F = 14.489 respectively). In addition, the membrane and total caveolin protein level after treated with 0.1 mM (*p < 0.05, F = 6.843, or *p < 0.05, F = 7.944 respectively) H2O2 for different durations also down regulated. Fluorescence microscopy also showed that phosphorylated caveolin-1 was distributed near the focal adhesions of the cells. This study concludes that the responses of HLECs to oxidative stress may include down regulation of caveolin and phosphorylation of caveolin-1 on Tyr14, and that MbetaCD also down regulates caveolin while depleting cholesterol in HLECs.


Assuntos
Caveolinas/biossíntese , Células Epiteliais/metabolismo , Peróxido de Hidrogênio/farmacologia , Cristalino/metabolismo , beta-Ciclodextrinas/farmacologia , Caveolina 1/biossíntese , Caveolina 1/metabolismo , Caveolinas/genética , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Colesterol/metabolismo , Células Epiteliais/efeitos dos fármacos , Humanos , Cristalino/citologia , Estresse Oxidativo , Fosforilação
8.
Infect Immun ; 74(1): 152-9, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16368968

RESUMO

Trypanosoma cruzi infection causes cardiomyopathy and vasculopathy. Previous studies have demonstrated that infection of human umbilical vein endothelial and smooth muscle cells resulted in activation of extracellular signal-regulated kinase (ERK). In the present study, smooth muscle cells were infected with trypomastigotes, and immunoblot analysis revealed an increase in the expression of cyclin D1 and proliferating cell nuclear antigen (PCNA), important mediators of smooth muscle cell proliferation. Interestingly, after infection, the expression of caveolin-1 was reduced in both human umbilical vein endothelial cells and smooth muscle cells. Immunoblot and immunohistochemical analyses of lysates of carotid arteries obtained from infected mice revealed increased expression of PCNA, cyclin D1, its substrate, phospho-Rb (Ser780), and phospho-ERK1/2. The expression of the cyclin-dependent kinase inhibitor p21(Cip1/Waf1), caveolin-1, and caveolin-3 was reduced in carotid arteries obtained from infected mice. There was an increase in the abundance of pre-pro-endothelin-1 mRNA in the carotid artery and aorta from infected mice. The ET(A) receptor was also elevated in infected arteries. ERK activates endothelin-1, which in turn exerts positive feedback activating ERK, and cyclin D1 is a downstream target of both endothelin-1 and ERK. There was significant incorporation of bromodeoxyuridine into smooth muscle cell DNA when treatment was with conditioned medium obtained from infected endothelial cells. Taken together, these data suggest that T. cruzi infection stimulates smooth muscle cell proliferation and is likely a result of the upregulation of the ERK-cyclin D1-endothelin-1 pathway.


Assuntos
Proliferação de Células , Doença de Chagas/metabolismo , Doença de Chagas/patologia , Músculo Liso Vascular/parasitologia , Miócitos de Músculo Liso/parasitologia , Trypanosoma cruzi/fisiologia , Animais , Bromodesoxiuridina/metabolismo , Artérias Carótidas/enzimologia , Caveolinas/biossíntese , Caveolinas/genética , Ciclo Celular/fisiologia , Células Cultivadas , Ciclina D1/fisiologia , Células Endoteliais/metabolismo , Células Endoteliais/parasitologia , Endotelina-1/genética , MAP Quinases Reguladas por Sinal Extracelular/fisiologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos A , Camundongos Endogâmicos C3H , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/patologia , Antígeno Nuclear de Célula em Proliferação/fisiologia , Precursores de RNA/metabolismo , Receptor de Endotelina A/metabolismo
9.
Gynecol Oncol ; 98(3): 409-19, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16005053

RESUMO

OBJECTIVES: The obligate intracellular bacterium Chlamydia trachomatis is frequently found in association with benign proliferative, pre-neoplastic and malignant changes in cervical epithelium. The present study addresses the possible role of C. trachomatis infection of the uterine cervix in modulating human cancer gene expression. METHODS: RNA was extracted from both C. trachomatis infected and non-infected human fibroblast cultures treated with ITFgamma. The extracted RNA was used for cDNA microarrays carrying 33,000 human genes to detect abnormal gene expression induced by Chlamydia. Forty specimens of cervix dissected from the transformation zone had previously tested negative for HPV and positive for C. trachomatis by standard DNA PCR (20). These samples were subjected to RT-PCR to detect the expression of the abnormal genes induced by Chlamydia infection. RESULTS: The ITFgamma-induced, non-replicative Chlamydia-infected fibroblast cultures showed significant modulation of gene expression. The cultures showed a 2-fold decrease in the expression of the gene coding for the tumor suppressor caveolin-1, and increased expression of the oncogene C-myc, a promoter of cervical carcinogenesis. In tissues from the Chlamydia-infected cervical transformation zone, real-time RT-PCR demonstrated a highly significant average 4.7-fold reduction of caveolin-1 mRNA (P < or = 0.0001) and an average 2.1-fold increase in C-myc (P < 0.05). CONCLUSIONS: Human ITFgamma-treated fibroblasts as well as non-neoplastic cervical tissues responded to C. trachomatis with a strong down-regulation of caveolin-1 mRNA and a light up-regulation of C-myc mRNA. These changes were independent of the HPV high-risk types. This study reveals possible mechanisms by which C. trachomatis infection may contribute to neoplastic changes in the transformation of uterine cervix. These possible mechanisms require further evaluation.


Assuntos
Caveolinas/genética , Transformação Celular Neoplásica/genética , Colo do Útero/microbiologia , Chlamydia trachomatis/fisiologia , Genes Supressores de Tumor/fisiologia , Genes myc/genética , Neoplasias do Colo do Útero/genética , Neoplasias do Colo do Útero/microbiologia , Caveolina 1 , Caveolinas/biossíntese , Colo do Útero/metabolismo , Colo do Útero/patologia , Infecções por Chlamydia/complicações , Infecções por Chlamydia/genética , Infecções por Chlamydia/microbiologia , Chlamydia trachomatis/crescimento & desenvolvimento , Feminino , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/microbiologia , Humanos , Interferon gama/farmacologia , Análise de Sequência com Séries de Oligonucleotídeos , Proteínas Proto-Oncogênicas c-myc/biossíntese , Proteínas Proto-Oncogênicas c-myc/genética , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Regulação para Cima
10.
Anticancer Res ; 25(1A): 369-75, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15816560

RESUMO

Constitutive activation of estrogen receptor alpha (ER-alpha) expression is an early event in breast cancer tumorigenesis. However, the mechanism whereby ER-alpha is constitutively activated during transformation of normal mammary cells has not been well established. Previously, we reported that haploinsufficiency of caveolin-1, a major structural protein that forms caveolae, resulted in anchorage-independent growth of a normal mammary epithelial cell line, MCF10A. Here, we further demonstrated that ER-alpha but not ER-beta expression was constitutively activated in these caveolin-1 haploinsufficient cells. Transient treatment of MCF10A cells with beta-methyl-cyclodextrin, a chemical that can displace caveolin-1 from the plasma membrane, also stimulated ER-alpha expression. We further found that the 17beta-estradiol (E2) accelerated anchorage-independent growth of these cells in vitro and promoted their tumorigenesis in nude mice. These results suggest that dysregulation of caveolin-1 is one of the mechanisms by which ER-alpha expression is activated during initiation of breast tumorigenesis.


Assuntos
Neoplasias da Mama/metabolismo , Caveolinas/deficiência , Transformação Celular Neoplásica/efeitos dos fármacos , Transformação Celular Neoplásica/metabolismo , Receptor alfa de Estrogênio/biossíntese , Animais , Mama/efeitos dos fármacos , Mama/metabolismo , Mama/patologia , Neoplasias da Mama/induzido quimicamente , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Caveolina 1 , Caveolinas/antagonistas & inibidores , Caveolinas/biossíntese , Caveolinas/genética , Processos de Crescimento Celular/efeitos dos fármacos , Processos de Crescimento Celular/fisiologia , Transformação Celular Neoplásica/genética , Regulação para Baixo , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Estradiol/farmacologia , Receptor alfa de Estrogênio/genética , Feminino , Humanos , Neoplasias Mamárias Experimentais/induzido quimicamente , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/metabolismo , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Nus , beta-Ciclodextrinas/farmacologia
11.
Development ; 132(5): 1147-60, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15689376

RESUMO

Epidemiological findings suggest that the consequences of a given oncogenic stimulus vary depending upon the developmental state of the target tissue at the time of exposure. This is particularly evident in the mammary gland, where both age at exposure to a carcinogenic stimulus and the timing of a first full-term pregnancy can markedly alter the risk of developing breast cancer. Analogous to this, the biological consequences of activating oncogenes, such as MYC, can be influenced by cellular context both in terms of cell lineage and cellular environment. In light of this, we hypothesized that the consequences of aberrant MYC activation in the mammary gland might be determined by the developmental state of the gland at the time of MYC exposure. To test this hypothesis directly, we have used a doxycycline-inducible transgenic mouse model to overexpress MYC during different stages of mammary gland development. Using this model, we find that the ability of MYC to inhibit postpartum lactation is due entirely to its activation within a specific 72-hour window during mid-pregnancy; by contrast, MYC activation either prior to or following this 72-hour window has little or no effect on postpartum lactation. Surprisingly, we find that MYC does not block postpartum lactation by inhibiting mammary epithelial differentiation, but rather by promoting differentiation and precocious lactation during pregnancy, which in turn leads to premature involution of the gland. We further show that this developmental stage-specific ability of MYC to promote mammary epithelial differentiation is tightly linked to its ability to downregulate caveolin 1 and activate Stat5 in a developmental stage-specific manner. Our findings provide unique in vivo molecular evidence for developmental stage-specific effects of oncogene activation, as well as the first evidence linking MYC with activation of the Jak2-Stat5 signaling pathway.


Assuntos
Epitélio/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Glândulas Mamárias Animais/metabolismo , Proteínas Proto-Oncogênicas c-myc/biossíntese , Proteínas Proto-Oncogênicas c-myc/fisiologia , Animais , Apoptose , Northern Blotting , Western Blotting , Caveolina 1 , Caveolinas/biossíntese , Diferenciação Celular , Linhagem da Célula , Primers do DNA/química , Proteínas de Ligação a DNA/metabolismo , Regulação para Baixo , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Lactação , Camundongos , Camundongos Transgênicos , Proteínas do Leite/metabolismo , Fator de Transcrição STAT3 , Fator de Transcrição STAT5 , Transdução de Sinais , Fatores de Tempo , Transativadores/metabolismo , Regulação para Cima
12.
J Neurochem ; 92(4): 831-9, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15686485

RESUMO

Remodeling and plasticity in the adult brain require cholesterol redistribution and synthesis for the formation of new membrane components. Caveolin-1 is a cholesterol-binding membrane protein involved in cellular cholesterol transport and homeostasis. Evidence presented here demonstrates an up-regulation of caveolin-1 in the hippocampus, which was temporally correlated with an increase in synaptophysin during the reinnervation phase in a mouse model of hippocampal deafferentation. Using an in vitro model of neuronal reactive plasticity, we examined the effect of virally mediated overexpression of caveolin-1 on injured differentiated PC12 cells undergoing terminal remodeling. Three days post lesion, caveolin-1-overexpressing cells revealed increases in synaptophysin and GAP-43, two markers of neurite sprouting and synaptogenesis. Morphologically, caveolin-1-overexpressing cells showed a decrease in primary neurite outgrowth and branching as well as an increase in neurite density. Caveolin-1-overexpressing cells also revealed the presence of terminal swelling and beading along processes, consistent with a possible alteration of microtubules stability. Moreover, a focal enrichment of caveolin-1 immunofluorescence was observed at the bases of axonal and dendritic terminals of mouse primary hippocampal neurons. Altogether, these results indicate that caveolin-1 plays an active role in the regulation of injury-induced synaptic and terminal remodeling in the adult CNS.


Assuntos
Caveolinas/fisiologia , Hipocampo/lesões , Hipocampo/metabolismo , Plasticidade Neuronal/fisiologia , Animais , Caveolina 1 , Caveolinas/biossíntese , Diferenciação Celular/fisiologia , Células Cultivadas , Hipocampo/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neuritos/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Células PC12 , Ratos
13.
Glia ; 49(2): 275-87, 2005 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-15494979

RESUMO

Caveolae represent membrane microdomains acting as integrators of cellular signaling and functional processes. Caveolins are involved in the biogenesis of caveolae and regulate the activity of caveolae-associated proteins. Although caveolin proteins are found in the CNS, the regulation of caveolins in neural cells is poorly described. In the present study, we investigated different modes and mechanisms of caveolin gene regulation in primary rat astrocytes. We demonstrated that activation of cAMP-dependent signaling pathways led to a marked reduction in protein levels of caveolin-1/-2 in cortical astrocytes. Application of transforming growth factor-alpha (TGF-alpha) also resulted in a decrease of caveolin-1/-2 expression. Decreased caveolin protein levels were mirrored by diminished caveolin gene transcription. The repressive effect of TGF-alpha on caveolin-1 expression was MAP kinase-independent and partly mediated through the PI3-kinase pathway. Further downstream, inhibition of histone deacetylases abrogated TGF-alpha effects, suggesting that chromatin remodeling processes could contribute to caveolin-1 repression. Intriguingly, alterations of caveolin gene expression in response to cAMP or TGF-alpha coincided with reciprocal and brain-region specific changes in glial glutamate transporter GLT-1 expression. The reciprocal regulation of caveolin-1 and GLT-1 expression might be gated through a common PI3-kinase dependent pathway triggered by TGF-alpha. Finally, we showed that GLT-1 is located in non-caveolar lipid rafts of cortical astrocytes. In conclusion, this study highlights the occurrence of the reciprocal regulation of caveolin and GLT-1 expression during processes such as astrocyte differentiation via common signaling pathways. We also provide strong evidence that GLT-1 itself is concentrated in lipid rafts, inferring an important role for glial glutamate transporter function.


Assuntos
Astrócitos/metabolismo , Caveolinas/biossíntese , Sistema Nervoso Central/metabolismo , Transportador 2 de Aminoácido Excitatório/metabolismo , Microdomínios da Membrana/metabolismo , Animais , Animais Recém-Nascidos , Astrócitos/citologia , Caveolina 1 , Caveolina 2 , Caveolinas/efeitos dos fármacos , Sistema Nervoso Central/citologia , Córtex Cerebral/citologia , Córtex Cerebral/metabolismo , AMP Cíclico/metabolismo , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Transportador 2 de Aminoácido Excitatório/genética , Regulação da Expressão Gênica/genética , Ácido Glutâmico/metabolismo , Histona Desacetilases/efeitos dos fármacos , Histona Desacetilases/metabolismo , Microdomínios da Membrana/genética , Fosfatidilinositol 3-Quinases/metabolismo , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Fator de Crescimento Transformador alfa/metabolismo , Fator de Crescimento Transformador alfa/farmacologia
14.
Eur J Biochem ; 271(22): 4462-73, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15560787

RESUMO

The proliferation of smooth muscle cells (SMC) is a key event in the development of atherosclerosis. In addition to growth factors or cytokines, we have shown previously that n-3 polyunsaturated fatty acids (PUFAs) act in opposition to n-6 PUFAs by modulating various steps of the inflammatory process. We have investigated the molecular mechanisms by which the incorporation of the n-6 PUFA, arachidonic acid, increases the proliferation of rat SMC treated with interleukin-1beta, while the n-3 PUFAs eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), elicit no mitogenic response. Incorporation of EPA or DHA into SMC, which are then activated by interleukin-1beta to mimic inflammation, decreases promoter activity of the cyclin D1 gene and phosphorylation of the retinoblastoma protein. Together, our data demonstrate that n-3 effects are dependent on the Ras/Raf-1/extracellular signal regulated kinase (ERK)/mitogen-activated protein kinase pathway, and that down-regulation of the cyclin D1 promoter activity is mediated by the specific binding of the early growth response factor-1. Finally, we have shown that the incorporation of EPA and DHA also increased the concentration of caveolin-1 and caveolin-3 in caveolae, which correlated with n-3 PUFA inhibition of SMC proliferation through the mitogen-activated protein kinase pathway. We provide evidence indicating that, in contrast to n-6 PUFAs, n-3 PUFAs exert antiproliferative effects on SMC through the mitogen-activated protein kinase/ERK pathway.


Assuntos
Ciclina D1/biossíntese , Proteínas de Ligação a DNA/biossíntese , Ácidos Graxos Insaturados/farmacologia , Proteínas Imediatamente Precoces/biossíntese , Interleucina-1/farmacologia , Músculo Liso Vascular/efeitos dos fármacos , Fatores de Transcrição/biossíntese , Animais , Bovinos , Caveolina 1 , Caveolina 2 , Caveolinas/biossíntese , Proliferação de Células/efeitos dos fármacos , Ciclina D1/genética , Proteínas de Ligação a DNA/genética , Proteína 1 de Resposta de Crescimento Precoce , Ativação Enzimática , Ácidos Graxos Insaturados/metabolismo , Expressão Gênica/efeitos dos fármacos , Humanos , Proteínas Imediatamente Precoces/genética , Masculino , Camundongos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Músculo Liso Vascular/citologia , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/fisiologia , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Regiões Promotoras Genéticas/efeitos dos fármacos , Ratos , Ratos Wistar , Proteína do Retinoblastoma/metabolismo , Fatores de Transcrição/genética , Quinases raf/metabolismo , Proteínas ras/metabolismo
15.
Cancer Res ; 64(20): 7500-6, 2004 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-15492276

RESUMO

We recently demonstrated that caveolae, vesicular flask-shaped invaginations of the plasma membrane, represent novel therapeutic targets in multiple myeloma. In the present study, we demonstrate that vascular endothelial growth factor (VEGF) triggers Src-dependent phosphorylation of caveolin-1, which is required for p130(Cas) phosphorylation and multiple myeloma cell migration. Conversely, depletion of caveolin-1 by antisense methodology abrogates p130(Cas) phosphorylation and VEGF-triggered multiple myeloma cell migration. The proteasome inhibitor bortezomib both inhibited VEGF-triggered caveolin-1 phosphorylation and markedly decreased caveolin-1 expression. Consequently, bortezomib inhibited VEGF-induced multiple myeloma cell migration. Bortezomib also decreased VEGF secretion in the bone marrow microenvironment and inhibited VEGF-triggered tyrosine phosphorylation of caveolin-1, migration, and survival in human umbilical vascular endothelial cells. Taken together, these studies demonstrate the requirement of caveolae for VEGF-triggered multiple myeloma cell migration and identify caveolin-1 in multiple myeloma cells and human umbilical vascular endothelial cells as a molecular target of bortezomib.


Assuntos
Antineoplásicos/farmacologia , Ácidos Borônicos/farmacologia , Caveolinas/metabolismo , Pirazinas/farmacologia , Fator A de Crescimento do Endotélio Vascular/farmacologia , Medula Óssea/metabolismo , Bortezomib , Caveolina 1 , Caveolinas/biossíntese , Caveolinas/genética , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Movimento Celular/fisiologia , Proteína Substrato Associada a Crk , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Humanos , Mieloma Múltiplo/genética , Mieloma Múltiplo/metabolismo , Mieloma Múltiplo/patologia , Fosforilação , Proteínas/metabolismo , Proteínas Recombinantes/farmacologia , Proteína p130 Retinoblastoma-Like , Transfecção , Tirosina/metabolismo , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Fator A de Crescimento do Endotélio Vascular/metabolismo , Quinases da Família src/metabolismo
16.
Anticancer Drugs ; 15(10): 961-7, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15514565

RESUMO

Caveolins are highly expressed in terminally differentiated cells, but this expression is down-regulated in various cancer cell lines. Exposure to low doses of paclitaxel (taxol) is sufficient to up-regulate caveolin-1, suggesting that a mild cytotoxic stress induces a response implying caveolin and caveolae. Here we show that this up-regulation is sustained even after the cessation of paclitaxel treatment. After exposure to a cytostatic dose of paclitaxel (50 nM), A549 lung cancer cells are blocked in the G2/M cell cycle phase. After removal of paclitaxel, cell death occurs, accompanied with an increase in caveolin expression, suggesting an effect of caveolin in this process. Three days post-paclitaxel treatment, surviving A549 cells were passaged and only a half of them adhered to the culture dish. Adhering cells (still mainly in the G2/M cell cycle phase) were still unable to grow and progressively entered in an apoptotic state. This study suggests that effects of a low dose of paclitaxel were still present even 1 week after drug removal and that caveolin-1 is a good marker of cytotoxicity.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Caveolinas/biossíntese , Divisão Celular/efeitos dos fármacos , Fase G2/efeitos dos fármacos , Paclitaxel/farmacologia , Apoptose , Caveolina 1 , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Humanos , Neoplasias Pulmonares , Regulação para Cima
17.
J Cell Sci ; 117(Pt 22): 5343-51, 2004 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-15466889

RESUMO

MAL, BENE and MAL2 are raft-associated integral membrane proteins of the MAL family of proteins involved in membrane trafficking processes. We show here that the human prostate carcinoma PC-3 cell line expresses the transcripts for the three proteins simultaneously. MAL, BENE and MAL2 co-fractionated with caveolin-1 in the raft fraction of PC-3 cells, and immunofluorescence analysis showed colocalization of these proteins with caveolin-1 in a multivesicular intracellular compartment. Markers of the Golgi apparatus, early and recycling endosomes and lipid droplets were excluded from this compartment. Prostate epithelial cells contain vesicular organelles enriched in raft components named prostasomes that are secreted in the prostate fluid. Interestingly, the prostasome fraction isolated from the culture supernatant of PC-3 cells consisted mainly of 30-130 nm cup-shaped vesicles that were positive for MAL, caveolin-1 and CD59, a glycosylphosphatidylinositol-anchored protein previously found in prostasomes. CD63, an integral membrane protein found in multivesicular bodies/lysosomes and secretory granules was also found in PC-3 cell-derived prostasomes. Prostasome secretion was not inhibited by brefeldin A, a compound that blocks the conventional secretory pathway. However, wortmannin, an inhibitor of phosphatidylinositol-3 kinase, reduced the secretion of prostasomes in PC-3 cells. Our results suggest that MAL family proteins are associated with caveolin-1 in a multivesicular compartment that may be involved in prostasomal secretion in PC-3 cells.


Assuntos
Caveolinas/biossíntese , Proteínas de Membrana Transportadoras/biossíntese , Proteínas da Mielina/biossíntese , Neoplasias da Próstata/metabolismo , Proteolipídeos/biossíntese , Androstadienos/farmacologia , Antígenos CD/biossíntese , Transporte Biológico , Northern Blotting , Western Blotting , Antígenos CD59/química , Caveolina 1 , Caveolinas/química , Linhagem Celular Tumoral , Membrana Celular/metabolismo , DNA Complementar/metabolismo , Detergentes/farmacologia , Eletroforese em Gel de Poliacrilamida , Endossomos/metabolismo , Inibidores Enzimáticos/farmacologia , Células Epiteliais , Glicosilação , Glicosilfosfatidilinositóis/química , Complexo de Golgi/metabolismo , Proteínas de Fluorescência Verde/química , Humanos , Lipídeos/química , Lisossomos/metabolismo , Masculino , Microdomínios da Membrana , Proteínas de Membrana Transportadoras/química , Microscopia Confocal , Microscopia Eletrônica , Microscopia de Fluorescência , Proteínas da Mielina/química , Proteínas Proteolipídicas Associadas a Linfócitos e Mielina , Glicoproteínas da Membrana de Plaquetas/biossíntese , Próstata/metabolismo , Proteolipídeos/química , Tetraspanina 30 , Transfecção , Wortmanina
18.
Clin Cancer Res ; 10(18 Pt 1): 6023-8, 2004 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-15447986

RESUMO

PURPOSE: Spiral computed tomography (CT) can detect lung cancer at an early stage, but the malignant potential is unknown. The question is, as follows: do these small lesions have the same lethal potential as do symptomatic tumors? EXPERIMENTAL DESIGN: We used a cDNA microarray platform and compared the gene expression profile of spiral CT-detected lung carcinomas with a matched case-control population of patients presenting with symptomatic lung cancer. RESULTS: CT-detected and symptomatic tumors have shown a comparable gene expression profile. Correspondence analysis has demonstrated that nine genes were differentially expressed, although with a high variability across the samples that prevented distinguishing the two groups of tumors. Analysis of these nine genes has suggested that early-detected tumors have higher levels of retinoic acid production and higher expression levels of caveolin 2, matrix Gla, and cystatin A, which are already known to be lost during tumor progression. CONCLUSIONS: All of the tumors observed are histologically malignant according to the WHO Classification. Early lung cancers that are detected by screening have a gene expression pattern similar to, but not identical to, that of symptomatic lung carcinomas.


Assuntos
DNA Complementar/metabolismo , Regulação Neoplásica da Expressão Gênica , Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/genética , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Tomografia Computadorizada Espiral/métodos , Idoso , Proteínas de Ligação ao Cálcio/biossíntese , Caveolina 2 , Caveolinas/biossíntese , Cistatinas/biossíntese , Progressão da Doença , Proteínas da Matriz Extracelular/biossíntese , Feminino , Humanos , Imuno-Histoquímica , Neoplasias Pulmonares/metabolismo , Masculino , Pessoa de Meia-Idade , Fenótipo , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Estatística como Assunto , Tomografia Computadorizada por Raios X , Proteína de Matriz Gla
19.
J Lipid Res ; 45(11): 2015-24, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15314095

RESUMO

Peroxisome proliferator-activated receptor gamma (PPARgamma) activation or overexpression induces caveolin-1 (cav-1) expression in several cell types. The objective of this study was to investigate if PPAR agonists could also regulate the cav-1 gene in macrophages and to explore the mechanisms involved. Our experiments demonstrated that rosiglitazone dose- and time-dependently increased cav-1 mRNA and protein in THP-1 macrophages. This induction was not observed in the presence of inhibitors of transcription or de novo protein synthesis. We also showed that the increase in cav-1 elicited by rosiglitazone was not related either to macrophage differentiation or to cellular apoptosis. The inductive effect seems to be dependent on PPAR activation, as the PPAR antagonist GW9662 abolished it. The activation of the liver X receptor with 22(R)-hydroxycholesterol also increased cav-1 mRNA, whereas the inactive (S) isomer did not. Finally, we identified a functional peroxisome proliferator response element in the cav-1 promoter that was activated upon rosiglitazone treatment in THP-1 macrophages.


Assuntos
Caveolinas/biossíntese , PPAR gama/metabolismo , Tiazolidinedionas/farmacologia , Regulação para Cima , Vasodilatadores/farmacologia , Anilidas/farmacologia , Animais , Apoptose , Sequência de Bases , Sítios de Ligação , Western Blotting , Antígeno CD11b/biossíntese , Caspase 3 , Caspases/metabolismo , Caveolina 1 , Caveolinas/metabolismo , Linhagem Celular , Proteínas de Ligação a DNA , Relação Dose-Resposta a Droga , Citometria de Fluxo , Humanos , Hidroxicolesteróis/metabolismo , Ligantes , Receptores X do Fígado , Macrófagos/metabolismo , Dados de Sequência Molecular , Receptores Nucleares Órfãos , Regiões Promotoras Genéticas , Biossíntese de Proteínas , RNA/metabolismo , RNA Mensageiro/metabolismo , Ratos , Receptores Citoplasmáticos e Nucleares/metabolismo , Rosiglitazona , Acetato de Tetradecanoilforbol/farmacologia , Fatores de Tempo , Transcrição Gênica
20.
Br J Cancer ; 91(5): 959-65, 2004 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-15305200

RESUMO

Caveolin-1 and -2 (CAV1, CAV2) are closely linked genes localised to the fragile region of 7q31 (FRA7G), and loss of heterozygosity involving this region has been reported in breast cancer. Several studies have suggested that CAV1 is a negative regulator of HER2/neu signal transduction in vitro. However, the clinical significance of CAV1 in breast cancer has not yet been clarified. We examined quantitatively the mRNA levels of CAV1, CAV2 and HER2/neu in 162 cases of breast cancer using real-time PCR. Caveolin-1 and -2 protein expression was also examined by Western blotting and immunohistochemistry. We then evaluated for correlations between CAV1, CAV2 and HER2/neu gene expression and clinicopathologic factors in the 162 breast cancer cases. Results showed higher HER2/neu mRMA levels and lower CAV1 and CAV2 mRMA levels in breast cancer tissues than in corresponding normal tissues (P<0.001). Caveolin-1 and -2 protein expression levels were also suppressed in cancer tissues compared to normal tissues by Western blotting. Immunohistochemistry revealed that CAV1 and CAV2 proteins were abundantly expressed in mammary gland myoepithelial cells, but only weakly in ductalepithelial cells. Reduced CAV1 mRNA level was significantly associated with increasing tumour size (P=0.041), and negative oestrogen receptor status (P=0.021). There was also a significant association between low CAV2 mRNA level and negative progesterone receptor status (P=0.013), and between high HER2/neu mRNA level and negative hormonal receptor status (ER, P=0.029, PgR, P=0.019). While there was no relationship between HER2/neu and CAV1 mRNA levels, a significant association between CAV1 and CAV2 mRNA levels was observed (P<0.001). Our results indicated that CAV1 suppression correlated closely with that of CAV2 in breast cancer, that CAV1 level was inversely correlated with tumour size, and that CAV1 and CAV2 levels were correlated with hormonal receptor status. Therefore, CAV1 and CAV2 play an important role in tumour progression in breast cancer patients.


Assuntos
Neoplasias da Mama/metabolismo , Caveolinas/biossíntese , Genes erbB-2/genética , RNA Mensageiro/análise , Biomarcadores Tumorais/análise , Western Blotting , Mama/metabolismo , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Caveolina 1 , Caveolina 2 , Caveolinas/genética , Linhagem Celular Tumoral , Progressão da Doença , Feminino , Expressão Gênica , Humanos , Imuno-Histoquímica , Hibridização in Situ Fluorescente , Prognóstico , Receptores de Estrogênio/biossíntese , Receptores de Progesterona/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA