Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Cycle ; 17(3): 288-297, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29160745

RESUMO

The purpose of this research has been deciphering the Warburg paradox, the biochemical enigma unsolved since 1923. We solved it by demonstrating that its specific character, i.e. the forced aerobic lactate exportation, represents a crucial metabolic device to counteract the cytotoxic effect produced by an excess of pyruvate at the connection of glycolysis with the Krebs cycle. This solution was verified by exposing cancer cells of different histogenesis to pyruvate concentrations higher than the physiological ones, after showing that these concentrations are totally innocuous when injected into mice. The mechanism of the pyruvate cytotoxicity relies on the saturation of the respiratory chain, leading to a negative shift of the cytosolic NADP/NADPH ratio and the consequent restriction of the purine synthesis and the related cell apoptosis. The reducing equivalents generated by glycolysis and by cytosolic metabolism compete each other for their disposal trough the respiratory chain; this makes it that the cytotoxicity of pyruvate is inversely related to the mitochondrial number and efficiency of various cell types. Thus, the cytotoxicity is high in anaplastic cancer stem cells, whose mitochondria are extremely few and immature (cristae-poor); on the contrary, no inhibition is brought about in adult differentiated cells, physiologically rich of mature mitochondria. All this generates the pyruvate anticancer selectivity, together with the lack of a general toxicity, making pyruvate represent an ideal candidate for a radical non toxical anticancer treatment.


Assuntos
Antineoplásicos/farmacologia , Glicólise/efeitos dos fármacos , Animais , Humanos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Neoplasias/metabolismo , Neoplasias/patologia , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/metabolismo , Hipóxia Tumoral/efeitos dos fármacos
2.
Cell Cycle ; 16(3): 280-285, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27841718

RESUMO

We previously showed that cellular RedOx state governs the G1-S transition of AH130 hepatoma, a tumor spontaneously reprogrammed to the embryonic stem cell stage. This transition is impaired when the mithocondrial electron transport system is blocked by specific inhibitors (antimycin A) or the respiratory chain is saturated by adding to the cells high concentrations of pyruvate. The antimycin A or pyruvate block is removed by the addition of adequate concentrations of folate (F). This suggests that the G1-S transition of AH130 cells depends on a respiration-linked step of DNA synthesis related to folate metabolism. In the study reported here, we characterized the effects of methotrexate (MTX), an inhibitor of dihydofolate-reductase, on the G1-S transition of hepatoma cells, in the absence or the presence of exogenously added F, dihydrofolate (FH2) or tetrahydrofolate (FH4). MTX, at 1 µM or higher concentrations, inhibited G1-S transition. This inhibition was completely removed by exogenous folates. Surprisingly, 10 nM MTX stimulated G1-S transition. The addition of F, but not FH2 or FH4, significantly increased this effect. Furthermore, 10 nM MTX removed the block of the G1-S transition operated by antimycin A or pyruvate, an effect which was enhanced in the presence of F. Finally, the stimulatory effect of 10 nM MTX was inhibited in the presence of serine. Our findings indicated that, under certain conditions, MTX may stimulate, rather than inhibiting, the cycling of cancer cells exhibiting a stem cell-like phenotype, such as AH130 cells. This may impact the therapeutic use of MTX and of folates as supportive care.


Assuntos
Ciclo Celular/efeitos dos fármacos , Metotrexato/farmacologia , Neoplasias/patologia , Aminoácidos/farmacologia , Animais , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Ácido Fólico/metabolismo , Masculino , Redes e Vias Metabólicas/efeitos dos fármacos , Ratos Wistar
3.
Oncotarget ; 6(31): 31985-96, 2015 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-26396171

RESUMO

We defined the stem cell profile of K562 line, demonstrating the expression of the Embryonic Transcription Factors Oct3/4, Sox2, Klf4 and Nanog. This profile was associated with a high vulnerability to the physiological oxidizable substrate pyruvate. remarkably, this substrate was shown to be innocuous, even at the highest doses, to normal differentiated cells. This vulnerability is based on a complex metabolic trim centered on the cellular redox state expressed by the NADP/NADPH ratio geared by the mitochondrial respiratory chain. Flow cytometry revealed that the inhibition of this chain by antimycin A produced cell accumulation in the S phase of cell cycle and apoptosis. This block negatively interferes with the aerobic synthesis of purines, without affecting the anaerobic synthesis of pyrimidines. This imbalance was reproduced by using two antifolate agents, LY309887 and raltitrexed (TDX), inhibitors of purine or pyrimidine synthesis, respectively. All this revealed the apparent paradox that low doses of TDX stimulated, instead of inhibiting, leukemia cell growth. This paradox might have significant impact on therapy with regard to the effects of TDX during the intervals of administration, when the drug concentrations become so low as to promote maintenance of dormant cancer cells in hypoxic tissue niches.


Assuntos
Antineoplásicos/farmacologia , Fase G1/efeitos dos fármacos , Leucemia/patologia , Redes e Vias Metabólicas/efeitos dos fármacos , Células-Tronco Neoplásicas/patologia , Fase S/efeitos dos fármacos , Citometria de Fluxo , Imunofluorescência , Antagonistas do Ácido Fólico/farmacologia , Humanos , Células K562 , Fator 4 Semelhante a Kruppel , Leucemia/tratamento farmacológico , Leucemia/metabolismo , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/metabolismo , Piruvatos/farmacologia , Células Tumorais Cultivadas
4.
Cell Cycle ; 13(2): 268-78, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24200964

RESUMO

We have previously shown that peculiar metabolic features of cell adaptation and survival in hypoxia imply growth restriction points that are typical of embryonic stem cells and disappear with differentiation. Here we provide evidence that such restrictions can be exploited as specific antiblastic targets by physiological factors such as pyruvate, tetrahydrofolate, and glutamine. These metabolites act as powerful cytotoxic agents on cancer stem cells (CSCs) when supplied at doses that perturb the biochemical network, sustaining the resumption of aerobic growth after the hypoxic dormant state. Experiments were performed in vivo and in vitro using CSCs obtained from various anaplastic tumors: human melanoma, leukemia, and rat hepatoma cells. Pretreatment of melanoma CSCs with pyruvate significantly reduces their self-renewal in vitro and tumorigenicity in vivo. The metabolic network underlying the cytotoxic effect of the physiological factors was thoroughly defined, principally using AH130 hepatoma, a tumor spontaneously reprogrammed to the embryonic stem stage. This network, based on a tight integration of aerobic glycolysis, cellular redox state, and folate metabolism, is centered on the cellular NADP/NADPH ratio that controls the redox pathway of folate utilization in purine synthesis. On the whole, this study indicates that pyruvate, FH 4, and glutamine display anticancer activity, because CSCs are committed to survive and maintain their stemness in hypoxia. When CSC need to differentiate and proliferate, they shift from anaerobic to aerobic status, and the few mitochondria available makes them susceptible to the injury of the above physiological factors. This vulnerability might be exploited for novel therapeutic treatments.


Assuntos
Carcinoma Hepatocelular/patologia , Neoplasias Hepáticas Experimentais/patologia , Células-Tronco Neoplásicas/efeitos dos fármacos , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/metabolismo , Hipóxia Celular , Linhagem Celular Tumoral/efeitos dos fármacos , Ciclo do Ácido Cítrico , Glutamina/metabolismo , Glutamina/farmacologia , Humanos , Leucemia/patologia , Neoplasias Hepáticas Experimentais/tratamento farmacológico , Neoplasias Hepáticas Experimentais/metabolismo , Melanoma/patologia , Redes e Vias Metabólicas , Mitose , NADP/metabolismo , Células-Tronco Neoplásicas/patologia , Oxirredução , Ácido Pirúvico/metabolismo , Ácido Pirúvico/farmacologia , Ratos , Ratos Wistar , Tetra-Hidrofolatos/metabolismo , Tetra-Hidrofolatos/farmacologia
5.
Cell Cycle ; 12(2): 353-64, 2013 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-23287475

RESUMO

One undisputed milestone of traditional oncology is neoplastic progression, which consists of a progressive selection of dedifferentiated cells driven by a chance sequence of genetic mutations. Recently it has been demonstrated that the overexpression of well-defined transcription factors reprograms somatic cells to the pluripotent stem status. The demonstration raises crucial questions as to whether and to what extent this reprogramming contributes to tumorigenesis, and whether the epigenetic changes involved in it are reversible. Here, we show for the first time that a tumor produced in vivo by a chemical carcinogen is the product of the interaction between neoplastic progression and reprogramming. The experimental model employed the prototype of ascites tumors, the Yoshida AH130 hepatoma and other neoplasias, including human melanoma. AH130 hepatoma was started in the liver by the carcinogen o-aminoazotoluene. This compound binds to and abolishes the p53 protein, producing a genomic instability that promotes both the neoplastic progression and the hepatoma reprogramming. Eventually this tumor contained 100% CD133(+) elements and pO(2)-dependent percentages of the three embryonic transcription factors Nanog, Klf4 and c-Myc. Once transferred into aerobic cultures, the minor cellular fraction expressing this triad generates various types of adherent cells, which are progressively substituted by non-tumorigenic elements committed to fibromuscular, neuronal and glial differentiation. This reprogramming appears to be accomplished stepwise, with the assembly of the triad into a sophisticated transcriptional, oxygen-dependent circuitry, in which Nanog and Klf4 antagonistically regulate c-Myc, and hence, cell hypoxia survival and cell cycle activation.


Assuntos
Desdiferenciação Celular/fisiologia , Transformação Celular Neoplásica/metabolismo , Regulação Neoplásica da Expressão Gênica/fisiologia , Fatores de Transcrição Kruppel-Like/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Fatores de Transcrição/metabolismo , Animais , Desdiferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Transformação Celular Neoplásica/efeitos dos fármacos , Primers do DNA/genética , Citometria de Fluxo , Instabilidade Genômica/efeitos dos fármacos , Humanos , Fator 4 Semelhante a Kruppel , Neoplasias Hepáticas Experimentais/induzido quimicamente , Masculino , Microscopia Eletrônica , Proteína Homeobox Nanog , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase em Tempo Real , Proteína Supressora de Tumor p53/metabolismo , o-Aminoazotolueno/metabolismo , o-Aminoazotolueno/toxicidade
6.
Bioelectrochemistry ; 78(2): 176-80, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19726240

RESUMO

A novel tethered bilayer lipid micromembrane (tBLmicroM) was prepared and characterized. It consists of a mercury cap electrodeposited on a platinum microelectrode, about 20 microm in diameter. The micromembrane was prepared by tethering to the mercury cap a thiolipid monolayer and by then self-assembling a lipid monolayer on top of it. The thiolipid consisted of a disulfidated tetraoxyethylene hydrophilic spacer covalently linked to two phytanyl chains. Upon incorporating OmpF porin in the tBLmicroM, its single-channel activity was recorded by the patch-clamp technique, and its particular features described. An electrochemical impedance spectrum of the tBLmicroM incorporating OmpF porin is also reported. To the best of our knowledge, this tBLmicroM is the first metal-supported biomimetic micromembrane capable of incorporating non-engineered channel proteins in a functionally active state from their detergent solutions, and of allowing the recording of single-channel activity and of impedance spectra of these proteins via ion translocation into the hydrophilic spacer. The limited spaciousness of the spacer prevents a statistical analysis based on current-amplitude or blockage-time histograms. Nonetheless, the robustness, stability, ease of preparation and disposability of the present tBLmicroM may open the way to the realization of a channel-protein microarray platform allowing a high throughput drug screening.


Assuntos
Biomimética/métodos , Microdomínios da Membrana/química , Microdomínios da Membrana/metabolismo , Mercúrio/química , Porinas/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Eletrodos , Canais de Potássio Éter-A-Go-Go/metabolismo , Gramicidina/metabolismo , Interações Hidrofóbicas e Hidrofílicas , Bicamadas Lipídicas/química , Bicamadas Lipídicas/metabolismo , Meliteno/metabolismo , Proteínas Musculares/metabolismo , Técnicas de Patch-Clamp , Transporte Proteico , Proteolipídeos/metabolismo
7.
Cell Cycle ; 7(2): 176-87, 2008 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-18256528

RESUMO

Tumors are ecosystems which develop from stem cells endowed with unlimited self-renewal and genetic instability, under the effects of mutagenesis and natural selection imposed by environmental changes. While changes and variations made possible by genetic instability are practically unlimited, the microenvironment progressively reduces those possibilities in the struggle for life imposed by hypoxia and nutrient shortage typical of tumor environments. This entails the tendency to evolve a convergent phenotype resistant to microenvironmental restrictions (first of all hypoxia), which progressively dominates the clonal selection. It is shown that adaptation to hypoxia, rather than being a peculiarity of cancer stem cells, is also a characteristic of normal hematopoietic stem cells, and may thus be described as a general feature of the stem cell phenotype. The metabolic orientation of this phenotype closely resembles the orientation of highly anaplastic ascites hepatomas, showing that, in restricted environments, stem cell recruitment to growth is limited by mitochondrial reoxidation of reducing equivalents produced in folate redox steps connected with purine synthesis. Finally, a review of earlier research into glucose metabolism in cancer leads to the reinterpretation of Warburg's aerobic glycolysis as a defence mechanism which disposes of glycolytic products able to negatively interfere with the crucial role of mitochondrial respiration in cell recruitment for growth.


Assuntos
Hipóxia , Neoplasias/metabolismo , Células-Tronco Neoplásicas/metabolismo , Animais , Glicólise , Humanos
8.
J Phys Chem B ; 112(4): 1315-9, 2008 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-18181608

RESUMO

The HERG potassium channel was incorporated in a mercury-supported tethered bilayer lipid membrane (tBLM) obtained by anchoring a thiolipid monolayer to the mercury surface and by self-assembling a lipid monolayer on top of it from a lipid film spread on the surface of an electrolyte solution. HERG was then incorporated in this tBLM from its micellar solution in Triton X-100, thus avoiding the use of vesicles in the preparation of the tBLM and of proteoliposomes in channel incorporation. The HERG "inward" current following a repolarization step was obtained by subtracting the current recorded upon addition of the specific inhibitor WAY from that recorded prior to this addition. This current was compared with that reported in the literature by the patch-clamp technique.


Assuntos
Canais de Potássio Éter-A-Go-Go/química , Mercúrio/química , Linhagem Celular , Humanos , Bicamadas Lipídicas/química , Estrutura Molecular
9.
Cancer Res ; 67(6): 2402-7, 2007 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-17363556

RESUMO

We worked out an experimental protocol able to purge the stem cell compartment of the SH-SY5Y neuroblastoma clone. This protocol was based on the prolonged treatment of the wild-type cell population with either hypoxia or the antiblastic etoposide. Cell fate was monitored by immunocytochemical and electrophysiologic (patch-clamp) techniques. Both treatments produced the progressive disappearance of neuronal type (N) cells (which constitute the bulk of the tumor), leaving space for a special category of epithelial-like substrate-adherent cells (S(0)). The latter represent a minimal cell component of the untreated population and are endowed with immunocytochemical markers (p75, c-kit, and CD133) and the electrophysiologic "nude" profile, typical of the neural crest stem cells. S(0) cells displayed a highly clonogenic potency and a substantial plasticity, generating both the N component and an alternative subpopulation terminally committed to the fibromuscular lineage. Unlike the N component, this lineage was highly insensitive to the apoptotic activity of hypoxia and etoposide and developed only when the neuronal option was abolished. Under these conditions, the fibromuscular progeny of S(0) expanded and progressed up to the exhaustion of the staminal compartment and to the extinction of the tumor. When combined, hypoxia and etoposide cooperated in abolishing the N cell generation and promoting the conversion of the tumor described. This synergy might mirror a natural condition in the ischemic areas occurring in cancer. These results have relevant implications for the understanding of the documented tendency of neuroblastomas to regress from a malignant to a benign phenotype, either spontaneously or on antiblastic treatment.


Assuntos
Etoposídeo/farmacologia , Hipóxia , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/metabolismo , Neuroblastoma/terapia , Antineoplásicos Fitogênicos/farmacologia , Linhagem Celular Tumoral , Células Clonais , Eletrofisiologia , Humanos , Imuno-Histoquímica , Células-Tronco Neoplásicas/patologia , Neuroblastoma/tratamento farmacológico , Neuroblastoma/metabolismo , Neuroblastoma/patologia
10.
Stem Cells ; 25(5): 1119-25, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17255519

RESUMO

We showed that resistance to severe hypoxia defines hierarchical levels within normal hematopoietic populations and that hypoxia modulates the balance between generation of progenitors and maintenance of hematopoietic stem cells (HSC) in favor of the latter. This study deals with the effects of hypoxia (0.1% oxygen) in vitro on Friend's murine erythroleukemia (MEL) cells, addressing the question of whether a clonal leukemia cell population comprise functionally different cell subsets characterized by different hypoxia resistance. To identify leukemia stem cells (LSC), we used the Culture Repopulating Ability (CRA) assay we developed to quantify in vitro stem cells capable of short-term reconstitution (STR). Hypoxia strongly inhibited the overall growth of MEL cell population, which, despite its clonality, comprised progenitors characterized by markedly different hypoxia-resistance. These included hypoxia-sensitive colony-forming cells and hypoxia-resistant STR-type LSC, capable of repopulating secondary liquid cultures of CRA assays, confirming what was previously shown for normal hematopoiesis. STR-type LSC were found capable not only of surviving in hypoxia but also of being mostly in cycle, in contrast with the fact that almost all hypoxia-surviving cells were growth-arrested and with what we previously found for HSC. However, quiescent LSC were also detected, capable of delayed culture repopulation with the same efficiency as STR-like LSC. The fact that even quiescent LSC, believed to sustain minimal residual disease in vivo, were found within the MEL cells indicates that all main components of leukemia cell populations may be present within clonal cell lines, which are therefore suitable to study the sensitivity of individual components to treatments. Disclosure of potential conflicts of interest is found at the end of this article.


Assuntos
Leucemia Eritroblástica Aguda/patologia , Células-Tronco Neoplásicas/patologia , Animais , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Hipóxia Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Células Clonais , Ensaio de Unidades Formadoras de Colônias , Fluoruracila/farmacologia , Camundongos , Células-Tronco Neoplásicas/efeitos dos fármacos
11.
Stem Cells ; 24(2): 443-53, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16100002

RESUMO

We explored the stem cell compartment of the SH-SY5Y neuroblastoma (NB) clone and its development by a novel approach, integrating clonal and immunocytochemical investigations with patch-clamp measurements of ion currents simultaneously expressed on single cells. The currents selected were the triad IHERG, IKDR, INa, normally expressed at varying mutual ratios during development of neural crest stem cells, from which NB derives upon neoplastic transformation. These ratios could be used as electrophysiological clusters of differentiation (ECDs), identifying otherwise indistinguishable stages in maturation. Subcloning procedures allowed the isolation of highly clonogenic substrate-adherent (S-type) cells that proved to be p75- and nestinpositive and were characterized by a nude electrophysiological profile (ECDS0). These cells expressed negligible levels of the triad and manifested the capacity of generating the two following lineages: first, a terminally differentiating, smooth muscular lineage, positive for calponin and smooth muscle actin, whose electrophysiological profile is characterized by a progressive diminution of IHERG, the increase of IKDR and INa, and the acquisition of IKIR (ECDS2); second, a neuronal abortive pathway (NF-68 positive), characterized by a variable expression of IHERG and IKDR and a low expression of INa (ECDNS). This population manifested a vigorous amplification, monopolizing the stem cell compartment at the expense of the smooth muscular lineage to such an extent that neuronal-like (N-type) cells must be continuously removed if the latter are to develop.


Assuntos
Crista Neural/fisiologia , Neuroblastoma/patologia , Células-Tronco/química , Células-Tronco/fisiologia , Antígenos de Diferenciação/análise , Diferenciação Celular , Linhagem Celular Tumoral , Linhagem da Célula , Proliferação de Células , Células Clonais , Eletrofisiologia , Humanos , Imuno-Histoquímica , Modelos Biológicos , Mutação , Técnicas de Patch-Clamp , Fatores de Tempo
12.
Biol Chem ; 386(9): 919-29, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16164417

RESUMO

The macrophage colony-stimulating factor (M-CSF, CSF-1) regulates survival, proliferation and differentiation of mononuclear phagocytes, as well as macrophage motility and morphology. The latter features are usually regulated by ECM-mediated activation of integrins and subsequent tyrosine phosphorylation of cellular proteins, including focal adhesion kinase (FAK). FAK is phosphorylated by downstream receptor tyrosine kinases as well. We addressed the question whether M-CSF regulates FAK tyrosine phosphorylation in macrophages, and found that M-CSF induces FAK phosphorylation at all known tyrosine residues. This phosphorylation was dependent on Src. Extracellularly-regulated kinase (ERK), Jun N-terminal kinase (JNK) and phosphatidylinositol-3-kinase (PI3K) were found to be negatively involved in M-CSF-induced FAK phosphorylation, as their inhibition resulted in FAK hyper-phosphorylation. Following M-CSF treatment, FAK and the active forms of M-CSFR and Src were redistributed to the cytoskeleton, where active ERK, JNK and PI3K were detectable. Immunofluorescence showed the presence of FAK and its active form in focal complexes following M-CSF treatment. Moreover, cell spreading and adhesion were impaired when FAK tyrosine phosphorylation was abrogated by either transfection with FRNK, a dominant negative form of FAK, or treatment with a number of inhibitors of upstream FAK-activating signals. These results point to a relevant role for FAK in the regulation of cell spreading and adhesion in macrophages.


Assuntos
Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Fator Estimulador de Colônias de Macrófagos/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos/enzimologia , Animais , Western Blotting , Fracionamento Celular , Linhagem Celular , Células Cultivadas , Humanos , Imunoprecipitação , Macrófagos/citologia , Camundongos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosforilação/efeitos dos fármacos , Proteínas Tirosina Quinases/metabolismo , Domínios de Homologia de src
13.
Mol Biol Cell ; 16(6): 2972-83, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15800067

RESUMO

Adhesive receptors of the integrin family are primarily involved in cell-extracellular matrix adhesion. Additionally, integrins trigger multiple signaling pathways that are involved in cell migration, proliferation, survival, and differentiation. We previously demonstrated that the activation of integrins containing the beta(1) subunit leads to a selective increase in potassium currents carried by the human ether-a-go-go-related gene (hERG) channels in neuroblastoma and leukemia cells; this current activation modulates adhesion-dependent differentiation in these cells. We hypothesized that the cross-talk between integrins and hERG channels could be traced back to the assembly of a macromolecular signaling complex comprising the two proteins. We tested this hypothesis in both SH-SY5Y neuroblastoma cells and in human embryonic kidney 293 cells stably transfected with hERG1 and, therefore, expressing only the full-length hERG1 protein on the plasma membrane. The beta(1) integrin and hERG1 coprecipitate in these cells and colocalize in both intracellular and surface membrane compartments. The two proteins also coprecipitate with caveolin-1, suggesting the localization of the complex in lipid rafts/caveolae. hERG1-transfected cells undergo an activation of hERG currents after beta(1) integrin-mediated adhesion to fibronectin; concomitant with this activation, the focal adhesion kinase associates with the hERG1 protein and becomes tyrosine phosphorylated. Using hERG1-specific inhibitors, we show that the tyrosine phosphorylation of focal adhesion kinase is strictly dependent on hERG channel activity. Similarly, the activity of the small GTPase Rac1 turned out to be dependent on hERG currents. On the whole, these data indicate that the hERG1 protein associates with beta(1) integrins and modulates adhesion receptor signaling.


Assuntos
Adesão Celular , Canais de Potássio Éter-A-Go-Go/metabolismo , Cadeias beta de Integrinas/metabolismo , Transdução de Sinais , Técnicas de Cultura de Células , Linhagem Celular , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Canal de Potássio ERG1 , Fibronectinas/metabolismo , Citometria de Fluxo , Imunofluorescência , Humanos , Cadeias beta de Integrinas/análise , Membranas Intracelulares/metabolismo , Rim/citologia , Rim/embriologia , Modelos Biológicos , Neuroblastoma/patologia , Técnicas de Patch-Clamp , Fosforilação , Testes de Precipitina , Transfecção , Proteínas rac1 de Ligação ao GTP/metabolismo
14.
Cancer Res ; 64(2): 606-11, 2004 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-14744775

RESUMO

The acquisition of the capacity to invade surrounding tissues confers a more malignant phenotype to tumor cells and is necessary for the establishment of metastases. The understanding of the molecular mechanisms underlying cell invasion in human solid tumors such as colorectal cancers could provide not only more sensitive prognostic analyses but also novel molecular targets for cancer therapy. We report in this article that K(+) ion channels belonging to the HERG family are important determinants for the acquisition of an invasive phenotype in colorectal cancers. The herg1 gene and HERG1 protein are expressed in many colon cancer cell lines, and the activity of HERG channels modulates colon cancer cell invasiveness. Moreover, the amount of HERG1 protein expressed on the plasma membrane is directly related to the invasive phenotype of colon cancer cells. Finally, both the herg1 gene and HERG1 protein were expressed in a high percentage of primary human colorectal cancers, with the highest incidence occurring in metastatic cancers, whereas no expression could be detected either in normal colonic mucosa or in adenomas.


Assuntos
Proteínas de Transporte de Cátions , Neoplasias do Colo/genética , Neoplasias do Colo/patologia , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Proteínas de Ligação a DNA , Regulação Neoplásica da Expressão Gênica/genética , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Canais de Potássio/genética , Transativadores , Linhagem Celular Tumoral , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go , Humanos , Imuno-Histoquímica , Invasividade Neoplásica , Técnicas de Patch-Clamp , Canais de Potássio/fisiologia , Regulador Transcricional ERG
15.
Gene Expr Patterns ; 3(6): 767-76, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14643686

RESUMO

Deciphering the expression pattern of K+ channel encoding genes during development can help in the understanding of the establishment of cellular excitability and unravel the molecular mechanisms of neuromuscular diseases. We focused our attention on genes belonging to the erg family, which is deeply involved in the control of neuromuscular excitability in Drosophila flies and possibly other organisms. Both in situ hybridisation and RNase Protection Assay experiments were used to study the expression pattern of mouse (m)erg1, m-erg2 and m-erg3 genes during mouse embryo development, to allow the pattern to be compared with their expression in the adult. M-erg1 is first expressed in the heart and in the central nervous system (CNS) of embryonic day 9.5 (E9.5) embryos; the gene appears in ganglia of the peripheral nervous system (PNS) (dorsal root (DRG) and sympathetic (SCG) ganglia, mioenteric plexus), in the neural layer of retina, skeletal muscles, gonads and gut at E13.5. In the adult m-erg1 is expressed in the heart, various structures of the CNS, DRG and retina. M-erg2 is first expressed at E9.5 in the CNS, thereafter (E13.5) in the neural layer of retina, DRG, SCG, and in the atrium. In the adult the gene is present in some restricted areas of the CNS, retina and DRG. M-erg3 displayed an expression pattern partially overlapping that of m-erg1, with a transitory expression in the developing heart as well. A detailed study of the mouse adult brain showed a peculiar expression pattern of the three genes, sometimes overlapping in different encephalic areas.


Assuntos
Camundongos/embriologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Canais de Potássio/metabolismo , Animais , Encéfalo/embriologia , Encéfalo/metabolismo , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go , Regulação da Expressão Gênica no Desenvolvimento , Hibridização In Situ , Camundongos/genética , Camundongos/metabolismo , Camundongos Endogâmicos C57BL , Canais de Potássio/genética , RNA Mensageiro/análise , RNA Mensageiro/metabolismo
16.
Biochem Biophys Res Commun ; 302(1): 101-8, 2003 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-12593854

RESUMO

From the adrenergic SH-SY5Y human neuroblastoma clone, we isolated a subclone (21S) endowed with a glial-oriented phenotype. At difference from the parental clone, 21S cells responded to depolarizing stimuli with overshooting action potentials, whose repolarization phase was composed of an initial rapid episode, followed by a long-lasting plateau and a slow return to the resting potential (V(REST)). The action potential depolarization phase was sustained by a TTX-sensitive Na(+) current, while the first repolarizing episode was produced by the scanty delayed rectifier potassium current (I(KDR)) expressed in 21S cells. The bulk of repolarization, including the after-hyperpolarization, was sustained by the human eag related (HERG) potassium current (I(HERG)) that also governs V(REST) in 21S cells. This double role of I(HERG), together with the poor expression of I(KDRs), represents a novel finding in electrophysiology, as well as gives a clue to identify a new excitable element of the complex cellular population of neuroblastoma.


Assuntos
Proteínas de Transporte de Cátions , Proteínas de Ligação a DNA , Coração/fisiologia , Neuroblastoma/fisiopatologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Canais de Potássio/fisiologia , Transativadores , Western Blotting , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go , Imunofluorescência , Humanos , Neuroblastoma/patologia , Técnicas de Patch-Clamp , Regulador Transcricional ERG , Células Tumorais Cultivadas
17.
J Biol Chem ; 278(5): 2947-55, 2003 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-12431979

RESUMO

The role of K(+) channel activity during cell cycle progression has become a research topic of considerable interest. Blocking of K(+) channels inhibits the proliferation of many cell types, although the mechanism of this inhibition is unclear. There is speculation that K(+) channels differentially regulate the electrical potential of the plasma membrane (V(m)) during proliferation. We have demonstrated that in tumor cells the value of V(m) is clamped to rather depolarized values by K(+) channels belonging to the HERG family. We report here that tumor cell lines preferentially express the herg1 gene and a truncated, N-deleted form that corresponds to herg1b. This alternative transcript is also expressed in human primary acute myeloid leukemias. Both HERG1 and HERG1B proteins are expressed on the plasma membrane of tumor cells and can form heterotetramers. The expression of HERG protein isoforms is strongly cell cycle-dependent, accounting for variations in HERG currents along the mitotic cycle. Moreover, the blocking of HERG channels dramatically impairs cell growth of HERG-bearing tumor cells. These results suggest that modulated expression of different K(+) channels is the molecular basis of a novel mechanism regulating neoplastic cell proliferation.


Assuntos
Proteínas de Transporte de Cátions , Ciclo Celular/genética , Proteínas de Ligação a DNA , Regulação Neoplásica da Expressão Gênica , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Canais de Potássio/genética , Transativadores , Transcrição Gênica , Sequência de Bases , Ciclo Celular/efeitos dos fármacos , Divisão Celular , Primers do DNA , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Dados de Sequência Molecular , Neuroblastoma/patologia , Isoformas de Proteínas/genética , Proteínas Recombinantes/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Deleção de Sequência , Transcrição Gênica/efeitos dos fármacos , Regulador Transcricional ERG , Transfecção , Tretinoína/farmacologia , Células Tumorais Cultivadas
18.
Oncogene ; 21(23): 3670-6, 2002 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-12032835

RESUMO

We had previously shown that murine macrophages expressing v-Fes, the oncogenically activated counterpart of the c-Fes cytoplasmic tyrosine kinase, proliferate independently of Macrophage Colony-Stimulating Factor (MCSF) and that the Extracellular signal-Regulated Kinase (ERK) pathway mediates the mitogenic effect of v-Fes. In this study, the response of c-fes- and v-fes-overexpressing cells to MCSF was investigated. A critical modulation of the activation of Mitogen-activated ERK Kinase (MEK) and ERK based on the MCSF dose was characterized. ERK activation was increased by MCSF doses capable to elicit a mitogenic response (2-5 U/ml). On the contrary, MCSF doses as low as 0.05 U/ml markedly reduced ERK phosphorylation and nuclear content and moderately but significantly reduced cell proliferation. The reduction of MEK and ERK phosphorylation was very rapid, suggesting the involvement of cytosolic phosphatases. Among these, phospho-tyrosine protein phosphatases and phosphoserine/threonine protein phosphatase-2A were found involved. These findings represent the first observation that different doses of the same growth factor, MCSF in particular, can exert opposite effects on cell proliferation by switching on or off ERK signaling.


Assuntos
Fator Estimulador de Colônias de Macrófagos/farmacologia , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas Tirosina Quinases , Animais , Arsenicais/farmacologia , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Relação Dose-Resposta a Droga , Citometria de Fluxo , Humanos , MAP Quinase Quinase 1 , Macrófagos/enzimologia , Camundongos , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Ácido Okadáico/farmacologia , Fosforilação/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-fes , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo
19.
J Neurosci ; 22(9): 3414-25, 2002 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-11978818

RESUMO

Native rat lactotrophs express thyrotrophin-releasing hormone-dependent K+ currents consisting of fast and slow deactivating components that are both sensitive to the class III anti-arrhythmic drugs that block the eag-related gene (ERG) K+ current (I(ERG)). Here we describe in MMQ prolactin-releasing pituitary cells the isolation of the slowly deactivating long-lasting component (I(ERGS)), which, unlike the fast component (I(ERGF)), is insensitive to verapamil 2 microm but sensitive to a novel scorpion toxin (ErgTx-2) that hardly affects I(ERGF). The time constants of I(ERGS) activation, deactivation, and recovery from inactivation are more than one order of magnitude greater than in I(ERGF), and the voltage-dependent inactivation is left-shifted by approximately 25 mV. The very slow MMQ firing frequency (approximately 0.2 Hz) investigated in perforated patch is increased approximately four times by anti-arrhythmic agents, by ErgTx-2, and by the abrupt I(ERGS) deactivation. Prolactin secretion in the presence of anti-arrhythmics is three- to fourfold higher in comparison with controls. We provide evidence from I(ERGS) and I(ERGF) simulations in a firing model cell to indicate that only I(ERGS) has an accommodating role during the experimentally observed very slow firing. Thus, we suggest that I(ERGS) potently modulates both firing and prolactin release in lactotroph cells.


Assuntos
Proteínas de Transporte de Cátions , Proteínas de Membrana Transportadoras , Hipófise/fisiologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Canais de Potássio/metabolismo , Potássio/metabolismo , Prolactina/metabolismo , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Antiarrítmicos/farmacologia , Encéfalo/metabolismo , Bloqueadores dos Canais de Cálcio/farmacologia , Linhagem Celular , Simulação por Computador , Relação Dose-Resposta a Droga , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go , Modelos Neurológicos , Técnicas de Patch-Clamp , Hipófise/citologia , Hipófise/efeitos dos fármacos , Canais de Potássio/efeitos dos fármacos , Canais de Potássio/genética , RNA/genética , RNA/metabolismo , Ratos , Venenos de Escorpião/isolamento & purificação , Venenos de Escorpião/farmacologia , Tretinoína/metabolismo , Verapamil/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...