Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 84
Filtrar
1.
Cell ; 144(2): 253-67, 2011 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-21241894

RESUMO

The study of macroautophagy in mammalian cells has described induction, vesicle nucleation, and membrane elongation complexes as key signaling intermediates driving autophagosome biogenesis. How these components are recruited to nascent autophagosomes is poorly understood, and although much is known about signaling mechanisms that restrain autophagy, the nature of positive inductive signals that can promote autophagy remain cryptic. We find that the Ras-like small G protein, RalB, is localized to nascent autophagosomes and is activated on nutrient deprivation. RalB and its effector Exo84 are required for nutrient starvation-induced autophagocytosis, and RalB activation is sufficient to promote autophagosome formation. Through direct binding to Exo84, RalB induces the assembly of catalytically active ULK1 and Beclin1-VPS34 complexes on the exocyst, which are required for isolation membrane formation and maturation. Thus, RalB signaling is a primary adaptive response to nutrient limitation that directly engages autophagocytosis through mobilization of the core vesicle nucleation machinery.


Assuntos
Autofagia , Células Epiteliais/patologia , Fagossomos/metabolismo , Transdução de Sinais , Proteínas ral de Ligação ao GTP/metabolismo , Proteínas Reguladoras de Apoptose/metabolismo , Proteína Beclina-1 , Linhagem Celular , Classe III de Fosfatidilinositol 3-Quinases/metabolismo , Células Epiteliais/microbiologia , Humanos , Proteínas de Membrana/metabolismo , Complexos Multiproteicos/metabolismo , Salmonella typhimurium/fisiologia , Estresse Fisiológico , Proteínas de Transporte Vesicular/metabolismo
2.
PLoS Comput Biol ; 17(3): e1008870, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33784299

RESUMO

The emerging tumor-on-chip (ToC) approaches allow to address biomedical questions out of reach with classical cell culture techniques: in biomimetic 3D hydrogels they partially reconstitute ex vivo the complexity of the tumor microenvironment and the cellular dynamics involving multiple cell types (cancer cells, immune cells, fibroblasts, etc.). However, a clear bottleneck is the extraction and interpretation of the rich biological information contained, sometime hidden, in the cell co-culture videos. In this work, we develop and apply novel video analysis algorithms to automatically measure the cytotoxic effects on human cancer cells (lung and breast) induced either by doxorubicin chemotherapy drug or by autologous tumor-infiltrating cytotoxic T lymphocytes (CTL). A live fluorescent dye (red) is used to selectively pre-stain the cancer cells before co-cultures and a live fluorescent reporter for caspase activity (green) is used to monitor apoptotic cell death. The here described open-source computational method, named STAMP (spatiotemporal apoptosis mapper), extracts the temporal kinetics and the spatial maps of cancer death, by localizing and tracking cancer cells in the red channel, and by counting the red to green transition signals, over 2-3 days. The robustness and versatility of the method is demonstrated by its application to different cell models and co-culture combinations. Noteworthy, this approach reveals the strong contribution of primary cancer-associated fibroblasts (CAFs) to breast cancer chemo-resistance, proving to be a powerful strategy to investigate intercellular cross-talks and drug resistance mechanisms. Moreover, we defined a new parameter, the 'potential of death induction', which is computed in time and in space to quantify the impact of dying cells on neighbor cells. We found that, contrary to natural death, cancer death induced by chemotherapy or by CTL is transmissible, in that it promotes the death of nearby cancer cells, suggesting the release of diffusible factors which amplify the initial cytotoxic stimulus.


Assuntos
Apoptose/fisiologia , Técnicas de Cocultura/métodos , Linfócitos T Citotóxicos , Microambiente Tumoral/fisiologia , Linhagem Celular Tumoral , Biologia Computacional , Fibroblastos/citologia , Fibroblastos/fisiologia , Humanos , Cinética , Técnicas Analíticas Microfluídicas , Microscopia de Vídeo , Linfócitos T Citotóxicos/citologia , Linfócitos T Citotóxicos/fisiologia
3.
Cell Mol Life Sci ; 78(5): 1943-1955, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33145612

RESUMO

The Hippo signal transduction pathway is an essential regulator of organ size during developmental growth by controlling multiple cellular processes such as cell proliferation, cell death, differentiation, and stemness. Dysfunctional Hippo signaling pathway leads to dramatic tissue overgrowth. Here, we will briefly introduce the Hippo tumor suppressor pathway before focusing on one of its members and the unexpected twists that followed our quest of its functions in its multifarious actions beside the Hippo pathway: the STK38 kinase. In this review, we will precisely discuss the newly identified role of STK38 on regulating the nuclear export machinery by phosphorylating and activating, the major nuclear export receptor XPO1. Finally, we will phrase STK38's role on regulating the subcellular distribution of crucial cellular regulators such as Beclin1 and YAP1 with its implication in cancer.


Assuntos
Carioferinas/metabolismo , Neoplasias/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Transdução de Sinais , Transporte Ativo do Núcleo Celular , Animais , Proteína Beclina-1/metabolismo , Núcleo Celular/metabolismo , Humanos , Fosforilação , Proteína Exportina 1
4.
EMBO Rep ; 20(11): e48150, 2019 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-31544310

RESUMO

STK38 (also known as NDR1) is a Hippo pathway serine/threonine protein kinase with multifarious functions in normal and cancer cells. Using a context-dependent proximity-labeling assay, we identify more than 250 partners of STK38 and find that STK38 modulates its partnership depending on the cellular context by increasing its association with cytoplasmic proteins upon nutrient starvation-induced autophagy and with nuclear ones during ECM detachment. We show that STK38 shuttles between the nucleus and the cytoplasm and that its nuclear exit depends on both XPO1 (aka exportin-1, CRM1) and STK38 kinase activity. We further uncover that STK38 modulates XPO1 export activity by phosphorylating XPO1 on serine 1055, thus regulating its own nuclear exit. We expand our model to other cellular contexts by discovering that XPO1 phosphorylation by STK38 regulates also the nuclear exit of Beclin1 and YAP1, key regulator of autophagy and transcriptional effector, respectively. Collectively, our results reveal STK38 as an activator of XPO1, behaving as a gatekeeper of nuclear export. These observations establish a novel mechanism of XPO1-dependent cargo export regulation by phosphorylation of XPO1's C-terminal auto-inhibitory domain.


Assuntos
Autofagia , Núcleo Celular/metabolismo , Carioferinas/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Proteínas de Transporte/metabolismo , Cromatografia Líquida , Biologia Computacional/métodos , Via de Sinalização Hippo , Humanos , Fosforilação , Ligação Proteica , Mapeamento de Interação de Proteínas , Transporte Proteico , Transdução de Sinais , Espectrometria de Massas em Tandem , Proteína Exportina 1
5.
Soft Matter ; 15(4): 537-545, 2019 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-30516225

RESUMO

We study the competition for space between two cell lines that differ only in the expression of the Ras oncogene. The two cell populations are initially separated and set to migrate antagonistically towards an in-between stripe of free substrate. After contact, their interface moves towards the population of normal cells. We interpret the velocity and traction force data taken before and after contact thanks to a hydrodynamic description of collectively migrating cohesive cell sheets. The kinematics of cells, before and after contact, allows us to estimate the relative material parameters for both cell lines. As predicted by the model, the transformed cell population with larger collective stresses pushes the wild type cell population.


Assuntos
Transformação Celular Neoplásica , Estresse Mecânico , Proteínas ras/metabolismo , Fenômenos Biomecânicos , Movimento Celular , Células HEK293 , Humanos
6.
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
7.
BMC Biol ; 16(1): 109, 2018 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-30285739

RESUMO

BACKGROUND: The WW domain-containing oxidoreductase (WWOX) gene, frequently altered in breast cancer, encodes a tumor suppressor whose function is mediated through its interactions with cancer-related proteins, such as the pro-apoptotic protein p73α. RESULTS: To better understand the involvement of WWOX in breast tumorigenesis, we performed a yeast two-hybrid screen and co-immunoprecipitation assays to identify novel partners of this protein. We characterized the vesicular overexpressed in cancer pro-survival protein 1 (VOPP1) as a new regulator of WWOX. In breast cancer cells, VOPP1 sequestrates WWOX in lysosomes, impairs its ability to associate with p73α, and inhibits WWOX-dependent apoptosis. Overexpressed VOPP1 potentiates cellular transformation and enhances the growth of transplanted tumors in vivo. VOPP1 is overexpressed in breast tumors, especially in tumors that retain WWOX. Moreover, increased expression of VOPP1 is associated with reduced survival of patients with WWOX-positive, but not with WWOX-negative, tumors. CONCLUSIONS: These findings emphasize the importance of the sequestration of WWOX by VOPP1 in addition to WWOX loss in breast tumors and define VOPP1 as a novel oncogene promoting breast carcinogenesis by inhibiting the anti-tumoral effect of WWOX.


Assuntos
Neoplasias da Mama/genética , Transformação Celular Neoplásica/genética , Fatores de Transcrição/genética , Proteínas Supressoras de Tumor/genética , Oxidorredutase com Domínios WW/genética , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Feminino , Humanos , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Oxidorredutase com Domínios WW/metabolismo
8.
EMBO J ; 33(20): 2349-62, 2014 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-25180232

RESUMO

The network of NF-κB-dependent transcription that activates both pro- and anti-inflammatory genes in mammals is still unclear. As NF-κB factors are evolutionarily conserved, we used Drosophila to understand this network. The NF-κB transcription factor Relish activates effector gene expression following Gram-negative bacterial immune challenge. Here, we show, using a genome-wide approach, that the conserved nuclear protein Akirin is a NF-κB co-factor required for the activation of a subset of Relish-dependent genes correlating with the presence of H3K4ac epigenetic marks. A large-scale unbiased proteomic analysis revealed that Akirin orchestrates NF-κB transcriptional selectivity through the recruitment of the Osa-containing-SWI/SNF-like Brahma complex (BAP). Immune challenge in Drosophila shows that Akirin is required for the transcription of a subset of effector genes, but dispensable for the transcription of genes that are negative regulators of the innate immune response. Therefore, Akirins act as molecular selectors specifying the choice between subsets of NF-κB target genes. The discovery of this mechanism, conserved in mammals, paves the way for the establishment of more specific and less toxic anti-inflammatory drugs targeting pro-inflammatory genes.


Assuntos
Montagem e Desmontagem da Cromatina , Proteínas de Drosophila/genética , Imunidade Inata , NF-kappa B/genética , Animais , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Drosophila , Proteínas de Drosophila/metabolismo , Feminino , Masculino , Mutação , NF-kappa B/metabolismo , Proteínas Nucleares , Regiões Promotoras Genéticas/genética , Proteômica , Transativadores/genética , Transativadores/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Ativação Transcricional , Técnicas do Sistema de Duplo-Híbrido
9.
J Cell Sci ; 129(20): 3756-3769, 2016 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-27591259

RESUMO

Coordination between membrane trafficking and actin polymerization is fundamental in cell migration, but a dynamic view of the underlying molecular mechanisms is still missing. The Rac1 GTPase controls actin polymerization at protrusions by interacting with its effector, the Wave regulatory complex (WRC). The exocyst complex, which functions in polarized exocytosis, has been involved in the regulation of cell motility. Here, we show a physical and functional connection between exocyst and WRC. Purified components of exocyst and WRC directly associate in vitro, and interactions interfaces are identified. The exocyst-WRC interaction is confirmed in cells by co-immunoprecipitation and is shown to occur independently of the Arp2/3 complex. Disruption of the exocyst-WRC interaction leads to impaired migration. By using time-lapse microscopy coupled to image correlation analysis, we visualized the trafficking of the WRC towards the front of the cell in nascent protrusions. The exocyst is necessary for WRC recruitment at the leading edge and for resulting cell edge movements. This direct link between the exocyst and WRC provides a new mechanistic insight into the spatio-temporal regulation of cell migration.


Assuntos
Movimento Celular , Extensões da Superfície Celular/metabolismo , Complexos Multiproteicos/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Família de Proteínas da Síndrome de Wiskott-Aldrich/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas do Citoesqueleto/metabolismo , Células HEK293 , Humanos , Ligação Proteica , Subunidades Proteicas/metabolismo
10.
EMBO J ; 32(14): 2039-55, 2013 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-23812009

RESUMO

Remodelling neuronal connections by synaptic activity requires membrane trafficking. We present evidence for a signalling pathway by which synaptic activity and its consequent Ca(2+) influx activate the small GTPase Ral and thereby recruit exocyst proteins to postsynaptic zones. In accord with the ability of the exocyst to direct delivery of post-Golgi vesicles, constitutively active Ral expressed in Drosophila muscle causes the exocyst to be concentrated in the region surrounding synaptic boutons and consequently enlarges the membrane folds of the postsynaptic plasma membrane (the subsynaptic reticulum, SSR). SSR growth requires Ral and the exocyst component Sec5 and Ral-induced enlargement of these membrane folds does not occur in sec5(-/-) muscles. Chronic changes in synaptic activity influence the plastic growth of this membrane in a manner consistent with activity-dependent activation of Ral. Thus, Ral regulation of the exocyst represents a control point for postsynaptic plasticity. This pathway may also function in mammals as expression of activated RalA in hippocampal neurons increases dendritic spine density in an exocyst-dependent manner and increases Sec5 in spines.


Assuntos
Proteínas de Drosophila/metabolismo , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Animais , Animais Geneticamente Modificados , Sinalização do Cálcio , Espinhas Dendríticas/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Drosophila melanogaster/crescimento & desenvolvimento , Drosophila melanogaster/metabolismo , Exocitose , Genes de Insetos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Proteínas Monoméricas de Ligação ao GTP/genética , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Junção Neuromuscular/crescimento & desenvolvimento , Junção Neuromuscular/metabolismo , Neurônios/metabolismo , Transporte Proteico , Ratos , Transdução de Sinais , Membranas Sinápticas/metabolismo , Membranas Sinápticas/ultraestrutura , Proteínas ral de Ligação ao GTP/genética , Proteínas ral de Ligação ao GTP/metabolismo
11.
Nat Methods ; 9(11): 1081-3, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23064519

RESUMO

Characterizing the migration of a population of cells remains laborious and somewhat subjective. Advances in genetics and robotics allow researchers to perform many experiments in parallel, but analyzing the large sets of data remains a bottleneck. Here we describe a rapid, fully automated correlation-based method for cell migration analysis, compatible with standard video microscopy. This method allows for the computation of quantitative migration parameters via an extensive dynamic mapping of cell displacements.


Assuntos
Movimento Celular , Rastreamento de Células/métodos , Processamento de Imagem Assistida por Computador/métodos , Linhagem Celular , Humanos , Microscopia de Vídeo/métodos , Cicatrização
12.
BMC Cancer ; 15: 1020, 2015 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-26715116

RESUMO

BACKGROUND: Aberrant activation of the Wnt/ß-catenin pathway is a major and frequent event in liver cancer, but inhibition of oncogenic ß-catenin signaling has proven challenging. The identification of genes that are synthetically lethal in ß-catenin-activated cancer cells would provide new targets for therapeutic drug design. METHODS: We transfected the parental HuH6 hepatoblastoma cell line with a doxycycline-inducible shRNA against CTNNB1 (gene coding for ß-catenin) to obtain an isogenic cell line pair with or without aberrant ß-catenin signaling. Using this hepatoblastoma isogenic cell line pair, we performed a human kinome-wide siRNA screen to identify synthetic lethal interactions with oncogenic CTNNB1. The phenotypic readouts of the screen were cell proliferation, cell cycle arrest and apoptosis, which were assessed by image-based analysis. In addition, apoptosis was assessed by flow cytometric experiments and immunoblotting. The potential synthetic lethal relationship between candidates genes identified in the screen and oncogenic CTNNB1 was also investigated in a different cellular context, a colorectal HCT116 isogenic cell line pair. RESULTS: We first determined the experimental conditions that led to the efficient expression of shRNA against CTNNB1 and maximal reduction of ß-catenin signaling activity in response to doxycycline treatment. Following high throughput screening in which 687 genes coding for kinases and proteins related to kinases (such as pseudokinases and phosphatases) were targeted, we identified 52 genes required for HuH6 survival. The silencing of five of these genes selectively impaired the viability of HuH6 cells with high ß-catenin signaling: HGS, STRADA, FES, BRAF and PKMYT1. Among these candidates, HGS depletion had the strongest inhibitory effect on cell growth and led to apoptosis specifically in HuH6 with high ß-catenin activity, while HuH6 with low ß-catenin activity were spared. In addition, HGS was identified as a potential synthetic lethal partner of oncogenic CTNNB1 in the HCT116 colorectal isogenic cell line pair. CONCLUSIONS: These results demonstrate the existence of crosstalk between ß-catenin signaling and HGS. Importantly, HGS depletion specifically affected cells with uncontrolled ß-catenin signaling activity in two different types of cancer (Hepatoblastoma HuH6 and colorectal HCT116), and thus may represent a new potential target for novel therapeutic strategies in liver and colorectal cancer.


Assuntos
Complexos Endossomais de Distribuição Requeridos para Transporte/genética , Hepatoblastoma/genética , Neoplasias Hepáticas/genética , Mutação , Fosfoproteínas/genética , RNA Interferente Pequeno/metabolismo , beta Catenina/antagonistas & inibidores , Apoptose , Pontos de Checagem do Ciclo Celular , Linhagem Celular Tumoral , Proliferação de Células , Células HCT116 , Humanos , Fosfotransferases/antagonistas & inibidores , Via de Sinalização Wnt , beta Catenina/genética
13.
Carcinogenesis ; 35(3): 670-82, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24148822

RESUMO

RNA interference has boosted the field of functional genomics, by making it possible to carry out 'loss-of-function' screens in cultured cells. Here, we performed a small interfering RNA screening, in three breast cancer cell lines, for 101 candidate driver genes overexpressed in amplified breast tumors and belonging to eight amplicons on chromosomes 8q and 17q, investigating their role in cell survival/proliferation. This screening identified eight driver genes that were amplified, overexpressed and critical for breast tumor cell proliferation or survival. They included the well-described oncogenic driver genes for the 17q12 amplicon, ERBB2 and GRB7. Four of six other candidate driver genes-RAD21 and EIF3H, both on chromosome 8q23, CHRAC1 on chromosome 8q24.3 and TANC2 on chromosome 17q23-were confirmed to be driver genes regulating the proliferation/survival of clonogenic breast cancer cells presenting an amplification of the corresponding region. Indeed, knockdown of the expression of these genes decreased cell viability, through both cell cycle arrest and apoptosis induction, and inhibited the formation of colonies in anchorage-independent conditions, in soft agar. Strategies for inhibiting the expression of these genes or the function of the proteins they encode are therefore of potential value for the treatment of breast cancers presenting amplifications of the corresponding genomic region.


Assuntos
Neoplasias da Mama/genética , Divisão Celular/genética , Sobrevivência Celular/genética , Transformação Celular Neoplásica/genética , Cromossomos Humanos Par 17 , Cromossomos Humanos Par 8 , RNA Interferente Pequeno/genética , Sequência de Bases , Neoplasias da Mama/patologia , Proteínas de Ciclo Celular , Primers do DNA , Proteínas de Ligação a DNA/genética , Feminino , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Proteínas Nucleares/genética , Nucleoproteínas/genética , Fosfoproteínas/genética , Proteínas/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
14.
Cell Rep Med ; 5(5): 101549, 2024 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-38703767

RESUMO

There is a compelling need for approaches to predict the efficacy of immunotherapy drugs. Tumor-on-chip technology exploits microfluidics to generate 3D cell co-cultures embedded in hydrogels that recapitulate simplified tumor ecosystems. Here, we present the development and validation of lung tumor-on-chip platforms to quickly and precisely measure ex vivo the effects of immune checkpoint inhibitors on T cell-mediated cancer cell death by exploiting the power of live imaging and advanced image analysis algorithms. The integration of autologous immunosuppressive FAP+ cancer-associated fibroblasts impaired the response to anti-PD-1, indicating that tumors-on-chips are capable of recapitulating stroma-dependent mechanisms of immunotherapy resistance. For a small cohort of non-small cell lung cancer patients, we generated personalized tumors-on-chips with their autologous primary cells isolated from fresh tumor samples, and we measured the responses to anti-PD-1 treatment. These results support the power of tumor-on-chip technology in immuno-oncology research and open a path to future clinical validations.


Assuntos
Inibidores de Checkpoint Imunológico , Neoplasias Pulmonares , Medicina de Precisão , Receptor de Morte Celular Programada 1 , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/imunologia , Medicina de Precisão/métodos , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Receptor de Morte Celular Programada 1/metabolismo , Receptor de Morte Celular Programada 1/imunologia , Inibidores de Checkpoint Imunológico/farmacologia , Inibidores de Checkpoint Imunológico/uso terapêutico , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/patologia , Carcinoma Pulmonar de Células não Pequenas/imunologia , Dispositivos Lab-On-A-Chip , Imunoterapia/métodos , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/imunologia , Linhagem Celular Tumoral
15.
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
16.
Cancer Gene Ther ; 30(8): 1144-1155, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37248434

RESUMO

The tumor suppressor gene WWOX is localized in an unstable chromosomal region and its expression is decreased or absent in several types of cancer. A low expression of WWOX is associated with a poor prognosis in breast cancer (BC). It has recently been shown that WWOX contributes to genome stability through its role in the DNA damage response (DDR). In breast cancer cells, WWOX inhibits homologous recombination (HR), and thus promotes the repair of DNA double-stranded breaks (DSBs) by non-homologous end joining (NHEJ). The fine-tuning modulation of HR activity is crucial. Its under or overstimulation inducing genome alterations that can induce cancer. MERIT40 is a positive regulator of the DDR. This protein is indispensable for the function of the multi-protein complex BRCA1-A, which suppresses excessive HR activity. MERIT40 also recruits Tankyrase, a positive regulator of HR, to the DSBs to stimulate DNA repair. Here, we identified MERIT40 as a new molecular partner of WWOX. We demonstrated that WWOX inhibited excessive HR activity induced by overexpression of MERIT40. We showed that WWOX impaired the MERIT40-Tankyrase interaction preventing the role of the complex on DSBs. Furthermore, we found that MERIT40 is overexpressed in BC and that this overexpression is associated to a poor prognosis. These results strongly suggest that WWOX, through its interaction with MERIT40, prevents the deleterious impact of excessive HR on BC development by inhibiting MERIT40-Tankyrase association. This inhibitory effect of WWOX would oppose MERIT40-dependent BC development.


Assuntos
Neoplasias da Mama , Recombinação Homóloga , Feminino , Humanos , Neoplasias da Mama/genética , Quebras de DNA de Cadeia Dupla , Reparo do DNA , Tanquirases/genética , Tanquirases/metabolismo , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo , Oxidorredutase com Domínios WW/genética , Oxidorredutase com Domínios WW/metabolismo
17.
EMBO J ; 27(18): 2375-87, 2008 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-18756269

RESUMO

The Ras family G-proteins RalA and RalB make critical non-overlapping contributions to the generation of a tumorigenic regulatory network, supporting bypass of the normal restraints on both cell proliferation and survival. The Sec6/8 complex, or exocyst, has emerged as a principal direct effector complex for Ral GTPases. Here, we show that RalA and RalB support mitotic progression through mobilization of the exocyst for two spatially and kinetically distinct steps of cytokinesis. RalA is required to tether the exocyst to the cytokinetic furrow in early cytokinesis. RalB is then required for recruitment of the exocyst to the midbody of this bridge to drive abscission and completion of cytokinesis. The collaborative action of RalA and RalB is specified by discrete subcellular compartmentalization and unique pairs of RalGEF proteins that provide inputs from both Ras-family protein-dependent and protein-independent regulatory cues. This suggests that Ral GTPases integrate diverse upstream signals to choreograph multiple roles for the exocyst in mitotic progression.


Assuntos
Citocinese , Regulação da Expressão Gênica , Proteínas ral de Ligação ao GTP/fisiologia , Fator ral de Troca do Nucleotídeo Guanina/metabolismo , Núcleo Celular/metabolismo , Proliferação de Células , Sobrevivência Celular , Células HeLa , Humanos , Cinética , Mitose , Modelos Biológicos , Proteínas de Transporte Vesicular/metabolismo , Proteínas ral de Ligação ao GTP/metabolismo , Proteínas ras/metabolismo
18.
Nat Cell Biol ; 7(12): 1191-201, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16284625

RESUMO

Signalling activity of the Notch receptor, which plays a fundamental role in metazoan cell fate determination, is controlled at multiple levels. We uncovered a Notch signal-controlling mechanism that depends on the ability of the non-visual beta-arrestin, Kurtz (Krz), to influence the degradation and, consequently, the function of the Notch receptor. We identified Krz as a binding partner of a known Notch-pathway modulator, Deltex (Dx), and demonstrated the existence of a trimeric Notch-Dx-Krz protein complex. This complex mediates the degradation of the Notch receptor through a ubiquitination-dependent pathway. Our results establish a novel mode of regulation of Notch signalling and define a new function for non-visual beta-arrestins.


Assuntos
Arrestinas/fisiologia , Proteínas de Drosophila/metabolismo , Receptores Notch/metabolismo , Transdução de Sinais , Animais , Arrestinas/genética , Arrestinas/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/fisiologia , Feminino , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Complexos Multiproteicos , Mutação/fisiologia , Técnicas do Sistema de Duplo-Híbrido , Ubiquitina/metabolismo , Asas de Animais/crescimento & desenvolvimento , beta-Arrestinas
19.
PLoS Biol ; 7(11): e1000235, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19885391

RESUMO

Atypical protein kinase C (aPKC) isoforms have been implicated in cell polarisation and migration through association with Cdc42 and Par6. In distinct migratory models, the Exocyst complex has been shown to be involved in secretory events and migration. By RNA interference (RNAi) we show that the polarised delivery of the Exocyst to the leading edge of migrating NRK cells is dependent upon aPKCs. Reciprocally we demonstrate that aPKC localisation at the leading edge is dependent upon the Exocyst. The basis of this inter-dependence derives from two-hybrid, mass spectrometry, and co-immunoprecipitation studies, which demonstrate the existence of an aPKC-Exocyst interaction mediated by Kibra. Using RNAi and small molecule inhibitors, the aPKCs, Kibra, and the Exocyst are shown to be required for NRK cell migration and it is further demonstrated that they are necessary for the localized activation of JNK at the leading edge. The migration associated control of JNK by aPKCs determines JNK phosphorylation of the plasma membrane substrate Paxillin, but not the phosphorylation of the nuclear JNK substrate, c-jun. This plasma membrane localized JNK cascade serves to control the stability of focal adhesion complexes, regulating migration. The study integrates the polarising behaviour of aPKCs with the pro-migratory properties of the Exocyst complex, defining a higher order complex associated with the localised activation of JNK at the leading edge of migrating cells that determines migration rate.


Assuntos
Movimento Celular , Proteína Quinase 8 Ativada por Mitógeno/metabolismo , Paxilina/metabolismo , Proteína Quinase C/metabolismo , Animais , Western Blotting , Carbazóis/farmacologia , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Membrana Celular/metabolismo , Células Cultivadas , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Exocitose , Adesões Focais/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas de Membrana , Microscopia Confocal , Microtúbulos/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Fosfoproteínas , Fosforilação , Ligação Proteica , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/genética , Proteínas/genética , Proteínas/metabolismo , Interferência de RNA , Ratos , Técnicas do Sistema de Duplo-Híbrido
20.
J Biol Chem ; 285(43): 33073-33081, 2010 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-20709745

RESUMO

Abnormalities in the process of endocytosis are classically linked to malignant transformation through the deficient down-regulation of signaling receptors. The present study describes a non-classical mechanism that does not require internalization by which endocytic proteins affect cell migration and basement membrane invasion. Specifically, we found that the endocytic adaptor epsin binds and regulates the biological properties of the signaling molecule RalBP1 (Ral-binding protein 1). Epsin interacted with the N terminus of RalBP1 via its characteristic epsin N-terminal homology (ENTH) domain. A combination of siRNA-mediated knock-down and transfection of siRNA-resistant constructs in fibrosarcoma cells demonstrated that impairment of the epsin-RalBP1 interaction led to cell migration and basement membrane invasion defects. We found the ENTH domain was necessary and sufficient to sustain normal cell migration and invasion. Because all the epsin endocytic motifs reside in the C-terminal part of the molecule, these results suggest that this novel regulatory circuit does not require endocytosis. In addition, cells depleted of epsin-RalBP1 complex displayed deficient activation of Rac1 and Arf6 suggesting a signaling function for this novel interaction. Further, overexpression of either epsin or RalBP1 enhanced migration and invasion of fibrosarcoma cells. Collectively, our results indicate that epsin regulates RalBP1 function in Rac1- and Arf6-dependent pathways to ultimately affect cell migration and invasion. We propose that the observed up-regulation of both epsin and RalBP1 in certain cancers contributes to their invasive characteristics.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Movimento Celular , Fibrossarcoma/metabolismo , Proteínas Ativadoras de GTPase/metabolismo , Fator 6 de Ribosilação do ADP , Fatores de Ribosilação do ADP/genética , Fatores de Ribosilação do ADP/metabolismo , Transportadores de Cassetes de Ligação de ATP/genética , Proteínas Adaptadoras de Transporte Vesicular/genética , Motivos de Aminoácidos , Animais , Membrana Basal/metabolismo , Membrana Basal/patologia , Linhagem Celular Tumoral , Endocitose/genética , Fibrossarcoma/genética , Fibrossarcoma/patologia , Proteínas Ativadoras de GTPase/genética , Humanos , Camundongos , Células NIH 3T3 , Invasividade Neoplásica/genética , Neuropeptídeos/genética , Neuropeptídeos/metabolismo , Estrutura Terciária de Proteína , Proteínas rac de Ligação ao GTP/genética , Proteínas rac de Ligação ao GTP/metabolismo , Proteínas rac1 de Ligação ao GTP/genética , Proteínas rac1 de Ligação ao GTP/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA