Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cancers (Basel) ; 14(7)2022 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-35406433

RESUMO

Sphingosine 1-phosphate (S1P), a bioactive lipid, interacts with five widely expressed G protein-coupled receptors (S1P1-5), regulating a variety of downstream signaling pathways with overlapping but also opposing functions. To date, data regarding the role of S1P5 in cell proliferation are ambiguous, and its role in controlling the growth of untransformed cells remains to be fully elucidated. In this study, we examined the effects of S1P5 deficiency on mouse embryonic fibroblasts (MEFs). Our results indicate that lack of S1P5 expression profoundly affects cell morphology and proliferation. First, S1P5 deficiency reduces cellular senescence and promotes MEF immortalization. Second, it decreases cell size and leads to cell elongation, which is accompanied by decreased cell spreading and migration. Third, it increases proliferation rate, a phenotype rescued by the reintroduction of exogenous S1P5. Mechanistically, S1P5 promotes the activation of FAK, controlling cell spreading and adhesion while the anti-proliferative function of the S1P/S1P5 signaling is associated with reduced nuclear accumulation of activated ERK. Our results suggest that S1P5 opposes the growth-promoting function of S1P1-3 through spatial control of ERK activation and provides new insights into the anti-proliferative function of S1P5.

2.
Med Sci (Paris) ; 36(3): 243-252, 2020 Mar.
Artigo em Francês | MEDLINE | ID: mdl-32228843

RESUMO

Multiple sclerosis (MS) is a disease of the central nervous system with a very debilitating inflammatory component that usually affects young people (years 20-40). This disease is characterized by the progressive destruction of the myelin sheath of the axons by the cells of the immune system, which results in neuronal degeneration. The T and B lymphocytes are the main players in this disease, which can be remittent with relapses or progressive. Among the drugs used to treat MS is the immunosuppressor fingolimod, the targets of which are the sphingosine 1-phosphate receptors. This molecule acts orally by preventing lymphocytes from leaving the thymus and lymph nodes and from reaching inflammatory brain foci. Other immunosuppressive drugs affecting sphingosine 1-phosphate receptors are under development and an intense search for curative drugs and treatments is being conducted.


TITLE: La sclérose en plaques et les médicaments immuno-modulateurs des récepteurs de la sphingosine 1-phosphate. ABSTRACT: La sclérose en plaques (SEP) est une maladie du système nerveux central à composante inflammatoire, très invalidante qui atteint généralement de jeunes adultes (20 à 40 ans). Cette maladie se caractérise par la destruction progressive, par les cellules du système immunitaire, de la gaine de myéline des axones, ce qui aboutit à une dégénérescence neuronale. Les lymphocytes T et B sont les acteurs principaux de cette maladie qui peut être rémittente ou progressive. Parmi les médicaments utilisés dans le cadre de son traitement, le fingolimod, un immunosuppresseur dont les cibles sont les récepteurs de la sphingosine 1-phosphate, administré par voie orale, agit en empêchant les lymphocytes de quitter le thymus et les ganglions lymphatiques, et de rejoindre les foyers inflammatoires cérébraux. Une recherche intense pour développer des traitements et des médicaments curatifs est actuellement en cours et d'autres immunosuppresseurs interagissant avec les récepteurs de sphingosine 1-phosphate sont en cours de développement.


Assuntos
Cloridrato de Fingolimode/uso terapêutico , Imunossupressores/uso terapêutico , Esclerose Múltipla/terapia , Receptores de Esfingosina-1-Fosfato/imunologia , Animais , Descoberta de Drogas/tendências , Humanos , Imunomodulação , Esclerose Múltipla/imunologia
3.
PLoS One ; 14(3): e0213203, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30845158

RESUMO

Although sphingosine-1-phosphate receptor 1 (S1P1) has been shown to trigger several S1P targeted functions such as immune cell trafficking, cell proliferation, migration, or angiogenesis, tools that allow the accurate detection of endogenous S1P1 localization and trafficking remain to be obtained and validated. In this study, we developed and characterized a novel monoclonal S1P1 antibody. Mice were immunized with S1P1 produced in the yeast Pichia pastoris and nine hybridoma clones producing monoclonal antibodies were created. Using different technical approaches including Western blot, immunoprecipitation and immunocytochemistry, we show that a selected clone, hereinafter referred to as 2B9, recognizes human and mouse S1P1 in various cell lineages. The interaction between 2B9 and S1P1 is specific over receptor subtypes, as the antibody does not binds to S1P2 or S1P5 receptors. Using cell-imaging methods, we demonstrate that 2B9 binds to an epitope located at the intracellular domain of S1P1; reveals cytosolic and membrane localization of the endogenous S1P1; and receptor internalization upon S1P or FTY720-P stimulation. Finally, loss of 2B9 signal upon knockdown of endogenous S1P1 by specific small interference RNAs further confirms its specificity. 2B9 was also able to detect S1P1 in human kidney and spinal cord tissue by immunohistochemistry. Altogether, our results suggest that 2B9 could be a useful tool to detect, quantify or localize low amounts of endogenous S1P1 in various physiological and pathological processes.


Assuntos
Anticorpos Monoclonais/imunologia , Neoplasias da Mama/metabolismo , Fibroblastos/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Processamento de Imagem Assistida por Computador/métodos , Receptores de Lisoesfingolipídeo/imunologia , Receptores de Lisoesfingolipídeo/metabolismo , Animais , Neoplasias da Mama/patologia , Células Cultivadas , Feminino , Fibroblastos/citologia , Humanos , Imunização , Rim/metabolismo , Camundongos , Microscopia de Fluorescência , Receptores de Esfingosina-1-Fosfato , Medula Espinal/metabolismo
4.
Biochim Biophys Acta Mol Cell Res ; 1865(11 Pt A): 1630-1639, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30327204

RESUMO

Cytokinesis begins in anaphase with the formation of the central spindle. PRC1 is a microtubule associated protein that plays an essential role in central spindle formation by crosslinking antiparallel microtubules. We have identified PRC1 as a novel binding partner for p27Kip1 (p27). p27 is a cyclin-CDK inhibitor that causes cell cycle arrest in G1. However, p27 has also been involved in the regulation of G2/M progression and cytokinesis, as well as of other cellular processes, including actin and microtubule cytoskeleton dynamics. We found that p27 interferes with the ability of PRC1 to bind to microtubules, without affecting PRC1 dimerization or its capacity to interact with other partners such as KIF4. In this way, p27 inhibited microtubule bundling by PRC1 in vitro and prevented the extensive microtubule bundling phenotype caused by PRC1 overexpression in cells in culture. Finally, co-expression of p27 or a p27 mutant that does not bind cyclin-CDKs inhibited multinucleation induced by PRC1 overexpression. Together, our results suggest that p27 may participate in the regulation of mitotic progression in a CDK-independent manner by modulating PRC1 activity.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Microtúbulos/metabolismo , Fuso Acromático/metabolismo , Imunofluorescência , Expressão Gênica , Células HEK293 , Células HeLa , Humanos , Mitose/genética , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Proteínas Recombinantes
6.
Sci Signal ; 10(472)2017 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-28351953

RESUMO

Sphingosine kinase 1 (SphK1) promotes cell proliferation and survival, and its abundance is often increased in tumors. SphK1 produces the signaling lipid sphingosine 1-phosphate (S1P), which activates signaling cascades downstream five G protein-coupled receptors (S1P1-5) to modulate vascular and immune system function and promote proliferation. We identified a new function of the SphK1-S1P pathway specifically in the control of mitosis. SphK1 depletion in HeLa cells caused prometaphase arrest, whereas its overexpression or activation accelerated mitosis. Increasing the abundance of S1P promoted mitotic progression, overrode the spindle assembly checkpoint (SAC), and led to chromosome segregation defects. S1P was secreted through the transporter SPNS2 and stimulated mitosis by binding to and activating S1P5 on the extracellular side, which then activated the intracellular phosphatidylinositol 3-kinase (PI3K)-AKT pathway. Knockdown of S1P5 prevented the S1P-induced spindle defect phenotype. RNA interference assays revealed that the mitotic kinase Polo-like kinase 1 (PLK1) was an important effector of S1P-S1P5 signaling-induced mitosis in HeLa cells. Our findings identify an extracellular signal and the downstream pathway that promotes mitotic progression and may indicate potential therapeutic targets to inhibit the proliferation of cancer cells.


Assuntos
Segregação de Cromossomos/efeitos dos fármacos , Lisofosfolipídeos/farmacologia , Mitose/efeitos dos fármacos , Receptores de Lisoesfingolipídeo/metabolismo , Transdução de Sinais/efeitos dos fármacos , Esfingosina/análogos & derivados , Animais , Western Blotting , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Células Cultivadas , Células HeLa , Humanos , Camundongos Knockout , Microscopia Confocal , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Interferência de RNA , Receptores de Lisoesfingolipídeo/genética , Esfingosina/farmacologia , Imagem com Lapso de Tempo/métodos , Quinase 1 Polo-Like
7.
Oncotarget ; 8(70): 114414-114415, 2017 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-29383084
8.
Carcinogenesis ; 35(11): 2503-11, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25173885

RESUMO

Gem is a small guanosine triphosphate (GTP)-binding protein within the Ras superfamily, involved in the regulation of voltage-gated calcium channel activity and cytoskeleton reorganization. Gem overexpression leads to stress fiber disruption, actin and cell shape remodeling and neurite elongation in interphase cells. In this study, we show that Gem plays a crucial role in the regulation of cortical actin cytoskeleton that undergoes active remodeling during mitosis. Ectopic expression of Gem leads to cortical actin disruption and spindle mispositioning during metaphase. The regulation of spindle positioning by Gem involves its downstream effector Gmip. Knockdown of Gmip rescued Gem-induced spindle phenotype, although both Gem and Gmip accumulated at the cell cortex. In addition, we implicated RhoA GTPase as an important effector of Gem/Gmip signaling. Inactivation of RhoA by overexpressing dominant-negative mutant prevented normal spindle positioning. Introduction of active RhoA rescued the actin and spindle positioning defects caused by Gem or Gmip overexpression. These findings demonstrate a new role of Gem/Gmip/RhoA signaling in cortical actin regulation during early mitotic stages.


Assuntos
Citoesqueleto de Actina/metabolismo , GTP Fosfo-Hidrolases/biossíntese , Proteínas Ativadoras de GTPase/biossíntese , Proteína rhoA de Ligação ao GTP/biossíntese , Citoesqueleto de Actina/genética , GTP Fosfo-Hidrolases/metabolismo , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Células HeLa , Humanos , Mitose/genética , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Transdução de Sinais/genética , Fuso Acromático/genética , Proteína rhoA de Ligação ao GTP/genética
9.
FASEB J ; 26(12): 5025-34, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22964304

RESUMO

Within the Ras superfamily, Gem is a small GTP-binding protein that plays a role in regulating Ca(2+) channels and cytoskeletal remodeling in interphase cells. Here, we report for the first time that Gem is a spindle-associated protein and is required for proper mitotic progression. Functionally, loss of Gem leads to misaligned chromosomes and prometaphase delay. On the basis of different experimental approaches, we demonstrate that loss of Gem by RNA interference induces spindle elongation, while its enforced expression results in spindle shortening. The spindle length phenotype is generated through deregulation of spindle dynamics on Gem depletion and requires the expression of its downstream effector, the kinesin Kif9. Loss of Kif9 induces spindle abnormalities similar to those observed when Gem expression is repressed by siRNA. We further identify Kif9 as a new regulator of spindle dynamics. Kif9 depletion increases the steady-state levels of spindle α-tubulin by increasing the rate of microtubule polymerization. Overall, this study demonstrates a novel mechanism by which Gem contributes to the mitotic progression by maintaining correct spindle length through the kinesin Kif9.


Assuntos
Segregação de Cromossomos , Cinesinas/metabolismo , Proteínas de Membrana/metabolismo , Mitose , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Fuso Acromático/metabolismo , Linhagem Celular , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células HeLa , Humanos , Immunoblotting , Cinesinas/genética , Proteínas de Membrana/genética , Microscopia de Fluorescência , Microscopia de Vídeo , Microtúbulos/metabolismo , Proteínas Monoméricas de Ligação ao GTP/genética , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Tubulina (Proteína)/metabolismo
10.
J Biol Chem ; 287(35): 29397-405, 2012 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-22700969

RESUMO

Ras GTPases signal by orchestrating a balance among several effector pathways, of which those driven by the GTPases RalA and RalB are essential to Ras oncogenic functions. RalA and RalB share the same effectors but support different aspects of oncogenesis. One example is the importance of active RalA in anchorage-independent growth and membrane raft trafficking. This study has shown a new post-translational modification of Ral GTPases: nondegradative ubiquitination. RalA (but not RalB) ubiquitination increases in anchorage-independent conditions in a caveolin-dependent manner and when lipid rafts are endocytosed. Forcing RalA mono-ubiquitination (by expressing a protein fusion consisting of ubiquitin fused N-terminally to RalA) leads to RalA enrichment at the plasma membrane and increases raft exposure. This study suggests the existence of an ubiquitination/de-ubiquitination cycle superimposed on the GDP/GTP cycle of RalA, involved in the regulation of RalA activity as well as in membrane raft trafficking.


Assuntos
Microdomínios da Membrana/metabolismo , Ubiquitina/metabolismo , Ubiquitinação/fisiologia , Proteínas ral de Ligação ao GTP/metabolismo , Transporte Biológico/fisiologia , Guanosina Difosfato/genética , Guanosina Difosfato/metabolismo , Guanosina Trifosfato/genética , Guanosina Trifosfato/metabolismo , Células HeLa , Humanos , Microdomínios da Membrana/genética , Ubiquitina/genética , Proteínas ral de Ligação ao GTP/genética
11.
Mol Cell ; 42(5): 650-61, 2011 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-21658605

RESUMO

The coordination of the several pathways involved in cell motility is poorly understood. Here, we identify SH3BP1, belonging to the RhoGAP family, as a partner of the exocyst complex and establish a physical and functional link between two motility-driving pathways, the Ral/exocyst and Rac signaling pathways. We show that SH3BP1 localizes together with the exocyst to the leading edge of motile cells and that SH3BP1 regulates cell migration via its GAP activity upon Rac1. SH3BP1 loss of function induces abnormally high Rac1 activity at the front, as visualized by in vivo biosensors, and disorganized and instable protrusions, as revealed by cell morphodynamics analysis. Consistently, constitutively active Rac1 mimics the phenotype of SH3BP1 depletion: slow migration and aberrant cell morphodynamics. Our finding that SH3BP1 downregulates Rac1 at the motile-cell front indicates that Rac1 inactivation in this location, as well as its activation by GEF proteins, is a fundamental requirement for cell motility.


Assuntos
Movimento Celular/fisiologia , Proteínas Ativadoras de GTPase/fisiologia , Proteínas rac1 de Ligação ao GTP/metabolismo , Animais , Regulação para Baixo , Ativação Enzimática , Proteínas Ativadoras de GTPase/genética , Proteínas Ativadoras de GTPase/metabolismo , Inativação Gênica , Centro Organizador dos Microtúbulos/fisiologia , Centro Organizador dos Microtúbulos/ultraestrutura , Ratos , Fatores de Transcrição/metabolismo , Fatores de Transcrição/fisiologia , Proteínas rac1 de Ligação ao GTP/genética , Proteínas ral de Ligação ao GTP/genética , Proteínas ral de Ligação ao GTP/fisiologia
12.
Int J Oncol ; 39(3): 697-707, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21643624

RESUMO

Data relating opioid treatment and modification of cancer cell migration (a prerequisite of metastasis) both in vitro and in vivo are diverging. In the present report we show that opioids increase the migratory activity of bladder cancer cells (T24 and EJ) and we provide a new mechanistic insight, explaining (at least partially) their action: we report that the enhanced opioid-related cell migration is controlled (in the absence of opioid receptors) through their interaction with bradykinin B2 receptors. Indeed, in these cell lines, opioids increase migration, adhesion, spreading and invasion by re-arranging actin cytoskeleton, increasing MMP-2 and -9 secretion and triggering specific intracellular signaling cascades in a non-opioid receptor mediated manner. An interaction, albeit with low affinity, of opioids with the bradykinin B2 receptor is reported, resulting in the increase of migration, while B2 antagonists revert this action. A systematic assay of different human epithelial cancer cell lines confirmed that only the B2-positive/opioid receptor-negative bladder cancer cells present this opioid-related increased migration/invasive phenotype. We suggest that opioid administration in cancer patients should be re-evaluated, keeping in mind that they may have other beneficial (protection) or adverse effects (spreading of cancer cells), in spite of their unique role in pain relief.


Assuntos
Analgésicos Opioides/farmacologia , Movimento Celular/efeitos dos fármacos , Receptor B2 da Bradicinina/metabolismo , Neoplasias da Bexiga Urinária/patologia , Actinas/metabolismo , Adesão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Citoesqueleto/metabolismo , Células HeLa , Humanos , Metaloproteinases da Matriz/metabolismo , Microscopia Confocal , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/genética , Receptor B2 da Bradicinina/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Transfecção , Neoplasias da Bexiga Urinária/metabolismo
13.
Eur J Cell Biol ; 88(2): 91-102, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19004523

RESUMO

Amphiphysins are BIN-amphiphysin-RVS (BAR) domain-containing proteins that influence membrane curvature in sites such as T-tubules in muscular cells, endocytic pits in neuronal as well as non-neuronal cells, and possibly cytoplasmic endosomes. This effect on lipid membranes is fulfilled by diverse amphiphysin 2/BIN1 isoforms, generated by alternative splicing and showing distinct structural and functional properties. In this study, our goal was to characterize the functional role of a ubiquitously expressed amphiphysin 2/BIN1 by the characterization of new molecular partners. We performed a two-hybrid screen with an isoform of amphiphysin 2/BIN1 expressed in HeLa cells. We identified CLIP-170 as an amphiphysin 2/BIN1-interacting molecule. CLIP-170 is a plus-end tracking protein involved in microtubule (MT) stability and recruitment of dynactin. The binding between amphiphysin 2/BIN1 and CLIP-170 is dependent on the N-terminal part of amphiphysin 2 (mostly the BAR domain) and an internal coiled-coil region of CLIP-170. This partnership was confirmed by GST pull-down assay and by co-immunoprecipitation in HeLa cells that express endogenous amphiphysin 2 (mostly isoforms 6, 9 and 10). When overexpressed in HeLa cells, amphiphysin 2/BIN1 leads to the formation of intracellular tubules which can closely align with MTs. After MT depolymerization by nocodazole, amphiphysin 2-stained tubules disappear, and reappear after nocodazole washout. Furthermore, depletion of CLIP-170 by RNAi induced a decrease in the proportion of cells with amphiphysin 2-stained tubules and an increase in the proportion of cells with no tubules. This result suggests the existence of a mechanistic link between the two types of tubules, which is likely to involve the +TIP protein, CLIP-170. Amphiphysin 2/BIN1 may be an anchoring point on membranes for CLIP-170, and consequently for MT. Then, the pushing force of polymerizing MT could help amphiphysin 2/BIN1 in its tubulation potential. We propose that amphiphysin 2/BIN1 participates in the tubulation of traffic intermediates and intracellular organelles first via its intrinsic tubulating potential and second via its ability to bind CLIP-170 and MT.


Assuntos
Membrana Celular/metabolismo , Extensões da Superfície Celular/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Microtúbulos/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Membrana Celular/efeitos dos fármacos , Extensões da Superfície Celular/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Células HeLa , Humanos , Espaço Intracelular/efeitos dos fármacos , Espaço Intracelular/metabolismo , Proteínas Associadas aos Microtúbulos/química , Microtúbulos/efeitos dos fármacos , Proteínas de Neoplasias/química , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/genética , Nocodazol/farmacologia , Ligação Proteica/efeitos dos fármacos , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Estrutura Terciária de Proteína , Transporte Proteico/efeitos dos fármacos , Tubulina (Proteína)/metabolismo , Técnicas do Sistema de Duplo-Híbrido
14.
Int Immunopharmacol ; 8(5): 634-44, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18387505

RESUMO

The opioid system plays a major role in immunomodulation, while its action on cells of the immune system may be opioid receptor-mediated or not. Opioid effects on B-lymphocytes are considered as indirect, attributed to an interplay between distinct cell populations. The aim of the present study was to investigate whether opioid agonists (morphine, alpha(S1)-casomorphin and ethylketocyclazocine) may have a direct action on the secretion of antibodies and cytokines by multiple myeloma-derived cell lines and normal CD19+ B-lymphocytes. Our results show that opioids modulate antibody and cytokine secretion by multiple myeloma cells in a cell line-dependent and opioid receptor-independent manner, while they decrease antibody secretion by normal B-lymphocytes. Furthermore, they decrease the proliferation rate of multiple myeloma cells through opioid receptor activation. Our data suggest two different mechanisms of action of opioids, mediated by different signaling pathways: an early non-opioid receptor-related effect, modulating the constitutive immunoglobulin and cytokine secretion, and a long-term receptor-mediated action on cell growth. These data suggest a further opioid implication in the control of humoral immunity.


Assuntos
Analgésicos Opioides/farmacologia , Linfócitos B/imunologia , Linfócitos B/metabolismo , Antígenos CD19/biossíntese , Antígenos CD19/genética , Linfócitos B/efeitos dos fármacos , Caseínas/farmacologia , Linhagem Celular , Citocinas/biossíntese , Relação Dose-Resposta a Droga , Etilcetociclazocina/farmacologia , Humanos , Imunoglobulinas/biossíntese , Técnicas In Vitro , Interleucinas/biossíntese , Mieloma Múltiplo/imunologia , Mieloma Múltiplo/metabolismo , Fragmentos de Peptídeos/farmacologia , Fosforilação , Receptores Opioides kappa/efeitos dos fármacos , Receptores Opioides mu/efeitos dos fármacos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/biossíntese , Proteínas Quinases p38 Ativadas por Mitógeno/genética
15.
Mol Biol Cell ; 18(4): 1242-52, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17267693

RESUMO

Gem is a protein of the Ras superfamily that plays a role in regulating voltage-gated Ca2+ channels and cytoskeletal reorganization. We now report that GTP-bound Gem interacts with the membrane-cytoskeleton linker protein Ezrin in its active state, and that Gem binds to active Ezrin in cells. The coexpression of Gem and Ezrin induces cell elongation accompanied by the disappearance of actin stress fibers and collapse of most focal adhesions. The same morphological effect is elicited when cells expressing Gem alone are stimulated with serum and requires the expression of ERM proteins. We show that endogenous Gem down-regulates the level of active RhoA and actin stress fibers. The effects of Gem downstream of Rho, i.e., ERM phosphorylation as well as disappearance of actin stress fibers and most focal adhesions, require the Rho-GAP partner of Gem, Gmip, a protein that is enriched in membranes under conditions in which Gem induced cell elongation. Our results suggest that Gem binds active Ezrin at the plasma membrane-cytoskeleton interface and acts via the Rho-GAP protein Gmip to down-regulate the processes dependent on the Rho pathway.


Assuntos
Proteínas do Citoesqueleto/metabolismo , Proteínas Ativadoras de GTPase/metabolismo , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo , Células 3T3 , Actinas/metabolismo , Actinas/ultraestrutura , Animais , Adesão Celular , Membrana Celular/metabolismo , Tamanho Celular , Proteínas do Citoesqueleto/genética , Citoesqueleto/metabolismo , Regulação para Baixo , Proteínas Ativadoras de GTPase/genética , Células HeLa , Humanos , Proteínas Imediatamente Precoces/genética , Proteínas Imediatamente Precoces/metabolismo , Camundongos , Proteínas Monoméricas de Ligação ao GTP/genética , Transdução de Sinais , Técnicas do Sistema de Duplo-Híbrido , Leveduras/genética
16.
Mol Cancer Ther ; 5(5): 1342-51, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16731768

RESUMO

Genomic signaling mechanisms require a relatively long time to get into action and represent the main way through which steroid hormones affect target cells. In addition, steroids may rapidly activate cellular functions by non-genomic signaling mechanisms involving membrane sites. Understanding in depth the molecular mechanisms of the non-genomic action represents an important frontier for developing new and more selective pharmacologic tools for endocrine therapies. In the present study, we report that membrane-impermeable testosterone-bovine serum albumin (BSA) acts synergistically with paclitaxel in modifying actin and tubulin cytoskeleton dynamics in LNCaP (androgen sensitive) and DU-145 (androgen insensitive) human prostate cancer cell lines. In addition, coincubation of either cell line with testosterone-BSA and paclitaxel induced inhibition of cell proliferation and apoptosis. Finally, in vivo experiments in LNCaP and DU-145 tumor xenografts in nude mice showed that both agents decrease tumor mass, whereas testosterone-BSA enhances the effect of paclitaxel. Our findings suggest that chronic activation of membrane androgen receptors in vitro and in vivo facilitates and sustains for a longer time the antitumoral action of cytoskeletal acting agents.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Paclitaxel/farmacologia , Neoplasias da Próstata/tratamento farmacológico , Receptores Androgênicos/metabolismo , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Apoptose/efeitos dos fármacos , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Permeabilidade da Membrana Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Citoesqueleto/efeitos dos fármacos , Citoesqueleto/metabolismo , Relação Dose-Resposta a Droga , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Paclitaxel/uso terapêutico , Neoplasias da Próstata/metabolismo , Soroalbumina Bovina/metabolismo , Soroalbumina Bovina/farmacologia , Testosterona/análogos & derivados , Testosterona/metabolismo , Testosterona/farmacologia , Células Tumorais Cultivadas
17.
J Steroid Biochem Mol Biol ; 98(2-3): 97-110, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16414261

RESUMO

Experimental and epidemiological data suggest a neuroprotective role for estrogen (E(2)). We have recently shown that, in PC12 cells, non-permeable estradiol conjugated to bovine serum albumin (BSA) prevent serum-deprivation induced apoptosis through activation of specific membrane estrogen receptors (mER). In the present study, we explored in detail the early signaling events involved in this anti-apoptotic action, downstream to activation of mER. Our findings suggest that mER is associated to G-proteins, and its activation with non-permeable E(2)-BSA results in the activation of the following downstream pro-survival kinases pathways: (1) the PKB/Akt pathway, (2) the Src-->MEK-->ERK kinases and finally (3) the MAPK-->ERK kinases. Activation of these pro-survival signals leads to CREB phosphorylation and NFkappaB nuclear translocation, two transcription factors controlling the expression of anti-apoptotic Bcl-2 proteins. These data suggest that major pro-survival kinases are involved in the mER-mediated anti-apoptotic effects of estrogen. This is further supported by experiments with specific kinases inhibitors, which partially but significantly reversed the mER-mediated anti-apoptotic effect of E(2)-BSA. Our findings suggest that estrogen act via mER as potent cytoprotective factors, downstream activating pro-survival kinases, assuring thus an efficient and multipotent activation of the anti-apoptotic machinery.


Assuntos
Membrana Celular/metabolismo , Sobrevivência Celular/fisiologia , MAP Quinase Quinase Quinases/metabolismo , Receptores de Estrogênio/metabolismo , Ativação Transcricional/efeitos dos fármacos , Quinases da Família src/metabolismo , Transporte Ativo do Núcleo Celular , Animais , Apoptose/efeitos dos fármacos , Bovinos , Membrana Celular/efeitos dos fármacos , Células Cultivadas , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Estrogênios/metabolismo , Estrogênios/farmacologia , Proteínas de Ligação ao GTP/metabolismo , Mitocôndrias/metabolismo , Modelos Biológicos , Óxido Nítrico Sintase/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Soroalbumina Bovina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Quinase Induzida por NF-kappaB
18.
Mol Cell Biol ; 26(2): 727-34, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16382162

RESUMO

The Ras family GTPases RalA and RalB have been defined as central components of the regulatory machinery supporting tumor initiation and progression. Although it is known that Ral proteins mediate oncogenic Ras signaling and physically and functionally interact with vesicle trafficking machinery, their mechanistic contribution to oncogenic transformation is unknown. Here, we have directly evaluated the relative contribution of Ral proteins and Ral effector pathways to cell motility and directional migration. Through loss-of-function analysis, we find that RalA is not limiting for cell migration in normal mammalian epithelial cells. In contrast, RalB and the Sec6/8 complex or exocyst, an immediate downstream Ral effector complex, are required for vectorial cell motility. RalB expression is required for promoting both exocyst assembly and localization to the leading edge of moving cells. We propose that RalB regulation of exocyst function is required for the coordinated delivery of secretory vesicles to the sites of dynamic plasma membrane expansion that specify directional movement.


Assuntos
Proteínas de Transporte/metabolismo , Movimento Celular/fisiologia , Proteínas ral de Ligação ao GTP/metabolismo , Animais , Células Cultivadas , Proteínas de Membrana/metabolismo , Ratos , Proteínas de Transporte Vesicular
19.
Front Biosci ; 10: 244-56, 2005 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-15574365

RESUMO

Opioids and somatostatin mediate their cellular effects through specific membrane receptors. Three major receptor classes (delta, mu and kappa) were identified for opioids, while for somatostatin, five different receptor classes (SSTR1-5) have been cloned. Through the interaction with their receptors, opioids and somatostatin exert their effects on cell growth, proliferation, differentiation and secretion. Specific actions of each receptor type have been reported, to be implicated in one or more of the cell functions referred above but have been mainly correlated with cell growth control. In several systems the effect of either neuropeptide is the reverse, inducing cell growth rather than antiproliferative and proapoptotic signals. In recent years, a growing number of reports indicate a possible interaction between opioid and somatostatin system. This could occur at the receptor level, through a cross-interaction of either neuropeptide with either receptor type, or receptor hetero-dimerization, and at a post-receptor level, via interaction with specific signaling molecules. These interactions provide new directions for the identification of specific molecules acting at the receptor and post-receptor level, mimicking the effects of both categories of agents.


Assuntos
Regulação Neoplásica da Expressão Gênica , Entorpecentes/química , Neoplasias/metabolismo , Receptores de Somatostatina/metabolismo , Somatostatina/química , Animais , Linhagem Celular Tumoral , Proliferação de Células , Dimerização , Humanos , Neuropeptídeos/química , Receptores Opioides delta/metabolismo , Receptores Opioides kappa/metabolismo , Receptores Opioides mu/metabolismo , Transdução de Sinais
20.
J Clin Endocrinol Metab ; 90(2): 893-903, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15585562

RESUMO

Nongenomic androgen actions imply mechanisms different from the classical intracellular androgen receptor (iAR) activation. We have recently reported the identification of a membrane androgen receptor (mAR) on LNCaP human prostate cancer cells, mediating testosterone signal transduction within minutes. In the present study we provide evidence that activation of mAR by nonpermeable, BSA-coupled testosterone results in 1) inhibition of LNCaP cell growth (with a 50% inhibitory concentration of 5.08 nM, similar to the affinity of testosterone for membrane sites); 2) induction in LNCaP cells of both apoptosis and the proapoptotic Fas protein; and 3) a significant decrease in migration, adhesion, and invasion of iAR-negative DU145 human prostate cancer cells. These actions persisted in the presence of antiandrogen flutamide or after decreasing the content of iAR in LNCaP cells by iAR antisense oligonucleotides. Testosterone-BSA was also effective in inducing apoptosis of DU145 human prostate cancer cells, negative for iAR, but expressing mAR sites. In LNCaP cell-inoculated nude mice, treatment with testosterone-BSA (4.8 mg/kg body weight) for 1 month resulted in a 60% reduction of tumor size compared with that in control animals receiving only BSA, an effect that was not affected by the antiandrogen flutamide. Our findings suggest that activators of mAR may represent a new class of antitumoral agents of prostate cancer.


Assuntos
Apoptose/efeitos dos fármacos , Oligodesoxirribonucleotídeos Antissenso/farmacologia , Neoplasias da Próstata/patologia , Receptores Androgênicos/fisiologia , Animais , Adesão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Membrana Celular/efeitos dos fármacos , Membrana Celular/fisiologia , Movimento Celular/efeitos dos fármacos , Citometria de Fluxo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias da Próstata/fisiopatologia , Receptores Androgênicos/genética , Soroalbumina Bovina , Testosterona/farmacologia , Transplante Heterólogo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...