Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Cell Sci ; 133(12)2020 06 23.
Artigo em Inglês | MEDLINE | ID: mdl-32467329

RESUMO

Recent developments in techniques for tissue clearing and size reduction have enabled optical imaging of whole organs and the study of rare tumorigenic events in vivo The adult mammary gland provides a unique model for investigating physiological or pathological processes such as morphogenesis or epithelial cell dissemination. Here, we establish a new pipeline to study rare cellular events occurring in the mammary gland, by combining orthotopic transplantation of mammary organoids with the uDISCO organ size reduction and clearing method. This strategy allows us to analyze the behavior of individually labeled cells in regenerated mammary gland. As a proof of concept, we analyzed the localization of rare epithelial cells overexpressing atypical protein kinase C iota (also known as PRKCI, referred to here as aPKCι) with an N-terminal eGFP fusion (GFP-aPKCι+) in the normal mammary gland. Using this analytical pipeline, we were able to visualize epithelial aPKCι+ cells escaping from the normal mammary epithelium and disseminating into the surrounding stroma. This technical resource should benefit mammary development and tumor progression studies.


Assuntos
Glândulas Mamárias Humanas , Organoides , Animais , Células Epiteliais , Epitélio , Humanos , Glândulas Mamárias Animais , Morfogênese
2.
Proc Natl Acad Sci U S A ; 116(48): 24108-24114, 2019 11 26.
Artigo em Inglês | MEDLINE | ID: mdl-31699818

RESUMO

Metastasis is the main cause of cancer-related deaths. How a single oncogenic cell evolves within highly organized epithelium is still unknown. Here, we found that the overexpression of the protein kinase atypical protein kinase C ι (aPKCi), an oncogene, triggers basally oriented epithelial cell extrusion in vivo as a potential mechanism for early breast tumor cell invasion. We found that cell segregation is the first step required for basal extrusion of luminal cells and identify aPKCi and vinculin as regulators of cell segregation. We propose that asymmetric vinculin levels at the junction between normal and aPKCi+ cells trigger an increase in tension at these cell junctions. Moreover, we show that aPKCi+ cells acquire promigratory features, including increased vinculin levels and vinculin dynamics at the cell-substratum contacts. Overall, this study shows that a balance between cell contractility and cell-cell adhesion is crucial for promoting basally oriented cell extrusion, a mechanism for early breast cancer cell invasion.


Assuntos
Neoplasias da Mama/metabolismo , Isoenzimas/fisiologia , Proteína Quinase C/fisiologia , Vinculina/metabolismo , Neoplasias da Mama/patologia , Adesão Celular , Linhagem Celular Tumoral , Separação Celular , Humanos , Junções Intercelulares/metabolismo , Isoenzimas/genética , Isoenzimas/metabolismo , Invasividade Neoplásica , Proteína Quinase C/genética , Proteína Quinase C/metabolismo
3.
Annu Rev Cell Dev Biol ; 32: 555-576, 2016 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-27501444

RESUMO

Metastasis is responsible for most cancer-associated deaths. Accumulating evidence based on 3D migration models has revealed a diversity of invasive migratory schemes reflecting the plasticity of tumor cells to switch between proteolytic and nonproteolytic modes of invasion. Yet, initial stages of localized regional tumor dissemination require proteolytic remodeling of the extracellular matrix to overcome tissue barriers. Recent data indicate that surface-exposed membrane type 1-matrix metalloproteinase (MT1-MMP), belonging to a group of membrane-anchored MMPs, plays a central role in pericellular matrix degradation during basement membrane and interstitial tissue transmigration programs. In addition, a large body of work indicates that MT1-MMP is targeted to specialized actin-rich cell protrusions termed invadopodia, which are responsible for matrix degradation. This review describes the multistep assembly of actin-based invadopodia in molecular details. Mechanisms underlying MT1-MMP traffic to invadopodia through endocytosis/recycling cycles, which are key to the invasive program of carcinoma cells, are discussed.


Assuntos
Metaloproteinase 14 da Matriz/metabolismo , Neoplasias/enzimologia , Neoplasias/patologia , Animais , Polaridade Celular , Humanos , Modelos Biológicos , Invasividade Neoplásica , Podossomos/metabolismo
4.
Sci Rep ; 6: 24925, 2016 04 27.
Artigo em Inglês | MEDLINE | ID: mdl-27116935

RESUMO

During their metastatic spread, cancer cells need to remodel the extracellular matrix in order to migrate through stromal compartments adjacent to the primary tumor. Dissemination of breast carcinoma cells is mediated by membrane type 1-matrix metalloproteinase (MT1-MMP/MMP14), the main invadopodial matrix degradative component. Here, we identify MT1-MMP as a novel interacting partner of dual-specificity LIM Kinase-1 and -2 (LIMK1/2), and provide several evidence for phosphorylation of tyrosine Y573 in the cytoplasmic domain of MT1-MMP by LIMK. Phosphorylation of Y573 influences association of F-actin binding protein cortactin to MT1-MMP-positive endosomes and invadopodia formation and matrix degradation. Moreover, we show that LIMK1 regulates cortactin association to MT1-MMP-positive endosomes, while LIMK2 controls invadopodia-associated cortactin. In turn, LIMK1 and LIMK2 are required for MT1-MMP-dependent matrix degradation and cell invasion in a three-dimensional type I collagen environment. This novel link between LIMK1/2 and MT1-MMP may have important consequences for therapeutic control of breast cancer cell invasion.


Assuntos
Movimento Celular , Quinases Lim/metabolismo , Metaloproteinase 14 da Matriz/metabolismo , Processamento de Proteína Pós-Traducional , Tirosina/metabolismo , Linhagem Celular Tumoral , Cortactina/metabolismo , Humanos , Fosforilação , Ligação Proteica , Mapeamento de Interação de Proteínas
5.
J Cell Sci ; 129(8): 1605-18, 2016 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-26945059

RESUMO

Here, we identify the LIM protein lipoma-preferred partner (LPP) as a binding partner of a specific protein phosphatase 2A (PP2A) heterotrimer that is characterised by the regulatory PR130/B″α1 subunit (encoded by PPP2R3A). The PR130 subunit interacts with the LIM domains of LPP through a conserved Zn²âº-finger-like motif in the differentially spliced N-terminus of PR130. Isolated LPP-associated PP2A complexes are catalytically active. PR130 colocalises with LPP at multiple locations within cells, including focal contacts, but is specifically excluded from mature focal adhesions, where LPP is still present. An LPP-PR130 fusion protein only localises to focal adhesions upon deletion of the domain of PR130 that binds to the PP2A catalytic subunit (PP2A/C), suggesting that PR130-LPP complex formation is dynamic and that permanent recruitment of PP2A activity might be unfavourable for focal adhesion maturation. Accordingly, siRNA-mediated knockdown of PR130 increases adhesion of HT1080 fibrosarcoma cells onto collagen I and decreases their migration in scratch wound and Transwell assays. Complex formation with LPP is mandatory for these PR130-PP2A functions, as neither phenotype can be rescued by re-expression of a PR130 mutant that no longer binds to LPP. Our data highlight the importance of specific, locally recruited PP2A complexes in cell adhesion and migration dynamics.


Assuntos
Proteínas do Citoesqueleto/metabolismo , Proteínas com Domínio LIM/metabolismo , Proteína Fosfatase 2/metabolismo , Domínio Catalítico/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Adesões Focais/genética , Humanos , Ligação Proteica , Proteína Fosfatase 2/genética , RNA Interferente Pequeno/genética
6.
Nat Commun ; 6: 6919, 2015 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-25897521

RESUMO

Cells release multiple, distinct forms of extracellular vesicles including structures known as microvesicles, which are known to alter the extracellular environment. Despite growing understanding of microvesicle biogenesis, function and contents, mechanisms regulating cargo delivery and enrichment remain largely unknown. Here we demonstrate that in amoeboid-like invasive tumour cell lines, the v-SNARE, VAMP3, regulates delivery of microvesicle cargo such as the membrane-type 1 matrix metalloprotease (MT1-MMP) to shedding microvesicles. MT1-MMP delivery to nascent microvesicles depends on the association of VAMP3 with the tetraspanin CD9 and facilitates the maintenance of amoeboid cell invasion. VAMP3-shRNA expression depletes shed vesicles of MT1-MMP and decreases cell invasiveness when embedded in cross-linked collagen matrices. Finally, we describe functionally similar microvesicles isolated from bodily fluids of ovarian cancer patients. Together these studies demonstrate the importance of microvesicle cargo sorting in matrix degradation and disease progression.


Assuntos
Regulação Neoplásica da Expressão Gênica/fisiologia , Invasividade Neoplásica , Neoplasias/irrigação sanguínea , Neovascularização Patológica/metabolismo , Animais , Linhagem Celular , Linhagem Celular Tumoral , Corantes Fluorescentes , Humanos , Melanoma , Transporte Proteico , Proteína 3 Associada à Membrana da Vesícula/genética , Proteína 3 Associada à Membrana da Vesícula/metabolismo
7.
Proc Natl Acad Sci U S A ; 111(18): E1872-9, 2014 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-24753582

RESUMO

Dissemination of carcinoma cells requires the pericellular degradation of the extracellular matrix, which is mediated by membrane type 1-matrix metalloproteinase (MT1-MMP). In this article, we report a co-up-regulation and colocalization of MT1-MMP and atypical protein kinase C iota (aPKCι) in hormone receptor-negative breast tumors in association with a higher risk of metastasis. Silencing of aPKC in invasive breast-tumor cell lines impaired the delivery of MT1-MMP from late endocytic storage compartments to the surface and inhibited matrix degradation and invasion. We provide evidence that aPKCι, in association with MT1-MMP-containing endosomes, phosphorylates cortactin, which is present in F-actin-rich puncta on MT1-MMP-positive endosomes and regulates cortactin association with the membrane scission protein dynamin-2. Thus, cell line-based observations and clinical data reveal the concerted activity of aPKC, cortactin, and dynamin-2, which control the trafficking of MT1-MMP from late endosome to the plasma membrane and play an important role in the invasive potential of breast-cancer cells.


Assuntos
Neoplasias da Mama/metabolismo , Isoenzimas/metabolismo , Metaloproteinase 14 da Matriz/metabolismo , Proteína Quinase C/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Adulto , Idoso , Transporte Biológico Ativo , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Carcinoma Ductal de Mama/genética , Carcinoma Ductal de Mama/metabolismo , Carcinoma Ductal de Mama/patologia , Linhagem Celular Tumoral , Cortactina/metabolismo , Grânulos Citoplasmáticos/metabolismo , Progressão da Doença , Dinamina II/metabolismo , Endossomos/metabolismo , Matriz Extracelular/metabolismo , Feminino , Humanos , Isoenzimas/antagonistas & inibidores , Isoenzimas/genética , Metaloproteinase 14 da Matriz/genética , Pessoa de Meia-Idade , Invasividade Neoplásica , Fosforilação , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/genética , Interferência de RNA , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Neoplásico/genética , RNA Neoplásico/metabolismo , RNA Interferente Pequeno/genética , Regulação para Cima
8.
Carcinogenesis ; 35(2): 396-406, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24072773

RESUMO

Protein kinase C iota (PKCι), a serine/threonine kinase required for cell polarity, proliferation and migration, is commonly up- or downregulated in cancer. PKCι is a human oncogene but whether this is related to its role in cell polarity and what repertoire of oncogenes acts in concert with PKCι is not known. We developed a panel of candidate oncogene expressing Madin-Darby canine kidney (MDCK) cells and demonstrated that H-Ras, ErbB2 and phosphatidylinositol 3-kinase transformation led to non-polar spheroid morphogenesis (dysplasia), whereas MDCK spheroids expressing c-Raf or v-Src were largely polarized. We show that small interfering RNA (siRNA)-targeting PKCι decreased the size of all spheroids tested and partially reversed the aberrant polarity phenotype in H-Ras and ErbB2 spheroids only. This indicates distinct requirements for PKCι and moreover that different thresholds of PKCι activity are required for these phenotypes. By manipulating PKCι function using mutant constructs, siRNA depletion or chemical inhibition, we have demonstrated that PKCι is required for polarization of parental MDCK epithelial cysts in a 3D matrix and that there is a threshold of PKCι activity above and below which, disorganized epithelial morphogenesis results. Furthermore, treatment with a novel PKCι inhibitor, CRT0066854, was able to restore polarized morphogenesis in the dysplastic H-Ras spheroids. These results show that tightly regulated PKCι is required for normal-polarized morphogenesis in mammalian cells and that H-Ras and ErbB2 cooperate with PKCι for loss of polarization and dysplasia. The identification of a PKCι inhibitor that can restore polarized morphogenesis has implications for the treatment of Ras and ErbB2 driven malignancies.


Assuntos
Polaridade Celular , Transformação Celular Neoplásica/patologia , Cistos/patologia , Células Epiteliais/patologia , Isoenzimas/metabolismo , Morfogênese/fisiologia , Proteína Quinase C/metabolismo , Esferoides Celulares/patologia , Animais , Transformação Celular Neoplásica/metabolismo , Células Cultivadas , Cistos/metabolismo , Cães , Células Epiteliais/metabolismo , Genes ras/fisiologia , Humanos , Isoenzimas/antagonistas & inibidores , Isoenzimas/genética , Rim/metabolismo , Rim/patologia , Fosfatidilinositol 3-Quinase/metabolismo , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/genética , RNA Interferente Pequeno/genética , Receptor ErbB-2/metabolismo , Esferoides Celulares/metabolismo
9.
J Cell Biol ; 203(6): 1063-79, 2013 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-24344185

RESUMO

Remodeling of the extracellular matrix by carcinoma cells during metastatic dissemination requires formation of actin-based protrusions of the plasma membrane called invadopodia, where the trans-membrane type 1 matrix metalloproteinase (MT1-MMP) accumulates. Here, we describe an interaction between the exocyst complex and the endosomal Arp2/3 activator Wiskott-Aldrich syndrome protein and Scar homolog (WASH) on MT1-MMP­containing late endosomes in invasive breast carcinoma cells. We found that WASH and exocyst are required for matrix degradation by an exocytic mechanism that involves tubular connections between MT1-MMP­positive late endosomes and the plasma membrane in contact with the matrix. This ensures focal delivery of MT1-MMP and supports pericellular matrix degradation and tumor cell invasion into different pathologically relevant matrix environments. Our data suggest a general mechanism used by tumor cells to breach the basement membrane and for invasive migration through fibrous collagen-enriched tissues surrounding the tumor.


Assuntos
Exocitose , Proteínas dos Microfilamentos/fisiologia , Proteínas de Transporte Vesicular/fisiologia , Adenocarcinoma/patologia , Neoplasias da Mama/patologia , Endossomos/metabolismo , Matriz Extracelular/metabolismo , Matriz Extracelular/ultraestrutura , Feminino , Humanos , Metaloproteinase 14 da Matriz/metabolismo , Proteínas dos Microfilamentos/metabolismo , Modelos Biológicos , Invasividade Neoplásica , Metástase Neoplásica/patologia , Metástase Neoplásica/ultraestrutura , Proteínas de Transporte Vesicular/metabolismo
10.
Sci Signal ; 6(293): ra82, 2013 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-24045153

RESUMO

Atypical protein kinase Cι (PKCι) has roles in cell growth, cellular polarity, and migration, and its abundance is frequently increased in cancer. We identified a protein interaction surface containing a dibasic motif (RIPR) that bound a distinct subset of PKCι substrates including lethal giant larvae 2 (LLGL2) and myosin X, but not other substrates such as Par3. Further characterization demonstrated that Arg471 in this motif was important for binding to LLGL2, whereas Arg474 was critical for interaction with myosin X, indicating that multiple complexes could be formed through this motif. A somatic mutation of the dibasic motif (R471C) was the most frequent mutation of PKCι in human cancer, and the intact dibasic motif was required for normal polarized epithelial morphogenesis in three-dimensional cysts. Thus, the R471C substitution is a change-of-function mutation acting at this substrate-specific recruitment site to selectively disrupt the polarizing activity of PKCι.


Assuntos
Polaridade Celular/genética , Proteínas do Citoesqueleto/metabolismo , Isoenzimas/química , Isoenzimas/genética , Modelos Moleculares , Proteínas de Neoplasias/genética , Conformação Proteica , Proteína Quinase C/química , Proteína Quinase C/genética , Motivos de Aminoácidos/genética , Sequência de Aminoácidos , Diamino Aminoácidos/genética , Análise de Variância , Animais , Proteínas do Citoesqueleto/genética , Cães , Células HEK293 , Humanos , Immunoblotting , Células Madin Darby de Rim Canino , Espectrometria de Massas , Microscopia de Fluorescência , Dados de Sequência Molecular , Mutação/genética , Fosforilação , Alinhamento de Sequência , Especificidade por Substrato
11.
J Cell Sci ; 126(Pt 11): 2381-91, 2013 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-23549785

RESUMO

Aberrant signalling of receptor tyrosine kinases (RTKs), such as c-Met, the receptor for hepatocyte growth factor (HGF), has been implicated in the oncogenesis of various tumours including non-small cell lung carcinoma (NSCLC). Through its pro-migratory properties, c-Met has been implicated specifically in the process of tumour metastasis, demanding a better understanding of the underlying signalling pathways. Various players downstream of c-Met have been well characterised, including the extracellular-signal-regulated kinases (ERKs) 1 and 2. In a small interfering RNA (siRNA)-based high-throughput wound healing screen performed in A549 lung carcinoma cells, we identified ERK2 but not ERK1 as a strong mediator of HGF-induced motility. This finding was confirmed in several NSCLC cell lines as well as in HeLa cells. One known substrate for ERK kinases in cell migration, the focal adhesion protein paxillin, was also one of the hits identified in the screen. We demonstrate that HGF stimulation results in a time-dependent phosphorylation of paxillin on serine 126, a process that can be blocked by inhibition of the ERK1/2 upstream kinase mitogen-activated protein kinase/ERK kinase 1 (MEK1) or inhibition of glycogen synthase kinase 3 (GSK3). Further, we show that paxillin turnover at focal adhesions is increased upon stimulation by HGF, an effect that is dependent on serine residues 126 (GSK3 site) and 130 (ERK site) within paxillin. In line with the isoform-specific requirement of ERK2 for HGF-mediated migration in lung tumour cell models, ERK2 but not ERK1 is shown to be responsible for paxillin serine 126 phosphorylation and its increased turnover at focal adhesions.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/metabolismo , Movimento Celular , Fator de Crescimento de Hepatócito/metabolismo , Neoplasias Pulmonares/metabolismo , Sistema de Sinalização das MAP Quinases , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Adesões Focais/genética , Adesões Focais/metabolismo , Quinase 3 da Glicogênio Sintase/genética , Quinase 3 da Glicogênio Sintase/metabolismo , Células HeLa , Fator de Crescimento de Hepatócito/farmacologia , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , MAP Quinase Quinase 1/genética , MAP Quinase Quinase 1/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/genética , Paxilina/genética , Paxilina/metabolismo , Proteínas Proto-Oncogênicas c-met/genética , Proteínas Proto-Oncogênicas c-met/metabolismo
12.
Biochem J ; 451(2): 329-42, 2013 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-23418854

RESUMO

The aPKC [atypical PKC (protein kinase C)] isoforms ι and ζ play crucial roles in the formation and maintenance of cell polarity and represent attractive anti-oncogenic drug targets in Ras-dependent tumours. To date, few isoform-specific chemical biology tools are available to inhibit aPKC catalytic activity. In the present paper, we describe the identification and functional characterization of potent and selective thieno[2,3-d]pyrimidine-based chemical inhibitors of aPKCs. A crystal structure of human PKCι kinase domain bound to a representative compound, CRT0066854, reveals the basis for potent and selective chemical inhibition. Furthermore, CRT0066854 displaces a crucial Asn-Phe-Asp motif that is part of the adenosine-binding pocket and engages an acidic patch used by arginine-rich PKC substrates. We show that CRT0066854 inhibits the LLGL2 (lethal giant larvae 2) phosphorylation in cell lines and exhibits phenotypic effects in a range of cell-based assays. We conclude that this compound can be used as a chemical tool to modulate aPKC activity in vitro and in vivo and may guide the search for further aPKC-selective inhibitors.


Assuntos
Proteína Quinase C/antagonistas & inibidores , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/farmacologia , Pirimidinas/química , Tiofenos/farmacologia , Adenosina/metabolismo , Trifosfato de Adenosina/química , Trifosfato de Adenosina/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Cristalografia por Raios X , Proteínas do Citoesqueleto/metabolismo , Cães , Avaliação Pré-Clínica de Medicamentos/métodos , Ensaios de Triagem em Larga Escala , Humanos , Concentração Inibidora 50 , Isoenzimas/antagonistas & inibidores , Mimetismo Molecular , Dados de Sequência Molecular , Fosforilação , Proteína Quinase C/química , Proteína Quinase C/metabolismo , Inibidores de Proteínas Quinases/síntese química , Inibidores de Proteínas Quinases/metabolismo , Pirimidinas/farmacologia , Tiofenos/química
13.
J Cell Sci ; 125(Pt 16): 3733-8, 2012 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-22553211

RESUMO

In migrating NRK cells, aPKCs control the dynamics of turnover of paxillin-containing focal adhesions (FA) determining migration rate. Using a proteomic approach (two-dimensional fluorescence difference gel electrophoresis), dynein intermediate chain 2 (dynein IC2) was identified as a protein that is phosphorylated inducibly during cell migration in a PKC-regulated manner. By gene silencing and co-immunoprecipitation studies, we show that dynein IC2 regulates the speed of cell migration through its interaction with paxillin. This interaction is controlled by serine 84 phosphorylation, which lies on the aPKC pathway. The evidence presented thus links aPKC control of migration to the dynein control of FA turnover through paxillin.


Assuntos
Adesão Celular/fisiologia , Movimento Celular/fisiologia , Dineínas/metabolismo , Paxilina/metabolismo , Sequência de Aminoácidos , Animais , Células Cultivadas , Dineínas do Citoplasma , Rim/citologia , Rim/enzimologia , Dados de Sequência Molecular , Fosforilação , Ratos
14.
Int J Cancer ; 131(12): 2852-62, 2012 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-22511072

RESUMO

Drugs targeting protein kinase C (PKC) show promising therapeutic activity. However, little is known about the expression patterns of the 11 PKC genes in human tumors, and the clinical significance of most PKC genes is unknown. We used qRT-PCR assays to quantify mRNA levels of the 11 PKC genes in 458 breast tumors from patients with known clinical/pathological status and long-term outcome. The proportion of tumors in which the expression of the different genes was altered varied widely, from 9.6% for PKN2 to 40.2% for PKCι/λ. In breast tumors, overexpression was the main alteration observed for PKCι/λ (33.4%), PKCδ (29.5%) and PKCζ (9.6%), whereas underexpression was the main alteration observed for PKCα (27.3%), PKCε (11.6%), PKCη (8.7%) and PKN2 (8.1%). Both overexpression and underexpression were observed for PKCß (underexpression 15.5%, overexpression 13.8%), PKCθ (underexpression 14.8%, overexpression 10.0%) and PKN1 (underexpression 6.6%, overexpression 7.4%). Several links were found between different PKC genes; and also between the expression patterns of PKC genes and several classical pathological and clinical parameters. PKCι/λ alone was found to have prognostic significance (p = 0.043), whereas PKCα showed a trend towards an influence on relapse-free survival (p = 0.052). PKCι/λ retained its prognostic significance in Cox multivariate regression analysis (p = 0.031). These results reveal very complex expression patterns of PKC genes in breast tumors, and suggest that their expression should be considered together when evaluating anti-tumoral drugs. PKCι/λ seems to be the most promising therapeutic target in breast cancer.


Assuntos
Neoplasias da Mama/genética , Família Multigênica , Proteína Quinase C/genética , Neoplasias da Mama/patologia , Feminino , Regulação Enzimológica da Expressão Gênica , Humanos , Prognóstico , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
15.
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
16.
J Cell Sci ; 123(Pt 16): 2725-32, 2010 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-20647370

RESUMO

Members of the PKC superfamily have been implicated in various migratory models and in particular in spatially restricted processes. However, defining the precise local events that underlie the PKC-dependent processes is constrained by the unspecific nature of interventions. Here we address this problem in relation to atypical PKC (aPKC) action, which in conjunction with the exocyst complex controls the polarised delivery of promigratory signals. A drug-dependent recruitment approach was employed to manipulate the local recruitment of signals to the leading edge of migrating cells, under conditions where the aPKC-exocyst control is globally abrogated. We found that activation of ERK but not JNK at focal adhesions recovers the majority of the migratory loss attributed to ERK action, demonstrating a necessary role for active plasma membrane ERK in the downstream signalling of aPKC-dependent migration. The data further show that restored focal adhesion dynamics are a contributing mechanism through which localized ERK activity influences this aPKC-exocyst-dependent migration.


Assuntos
Movimento Celular/fisiologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Proteína Quinase C/metabolismo , Animais , Membrana Celular/enzimologia , Células Cultivadas , Ativação Enzimática , Adesões Focais/enzimologia , Rim/citologia , MAP Quinase Quinase 1/metabolismo , Sistema de Sinalização das MAP Quinases , Paxilina/metabolismo , Fosforilação , Ligação Proteica , Ratos , Transdução de Sinais , Sirolimo/análogos & derivados , Sirolimo/farmacologia
17.
Nat Rev Mol Cell Biol ; 11(2): 103-12, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20094051

RESUMO

Networks of signal transducers determine the conversion of environmental cues into cellular actions. Among the main players in these networks are protein kinases, which can acutely and reversibly modify protein functions to influence cellular events. One group of kinases, the protein kinase C (PKC) family, have been increasingly implicated in the organization of signal propagation, particularly in the spatial distribution of signals. Examples of where and how various PKC isoforms direct this tier of signal organization are becoming more evident.


Assuntos
Proteína Quinase C/metabolismo , Transdução de Sinais , Animais , Comunicação Celular , Movimento Celular , Humanos , Isoenzimas/genética , Isoenzimas/metabolismo , Proteína Quinase C/genética , Transporte Proteico
18.
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
19.
Curr Opin Cell Biol ; 21(2): 268-79, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19233632

RESUMO

Intervention in protein kinase C (PKC) has a chequered history, partly because of the poor selectivity of many inhibitors and partly a reflection of the sometimes antagonistic action of related PKC isoforms. Recent advances in targeting PKC isoforms have come from structural work on isolated kinase domains that have provided opportunities to drive selectivity through structure-based avenues. The promise of isoform selective inhibitors and the rationale for their development are discussed in the broader context of the PKC inhibitor arsenal.


Assuntos
Isoenzimas/antagonistas & inibidores , Proteína Quinase C/antagonistas & inibidores , Animais , Domínio Catalítico , Ensaios Clínicos como Assunto , Ativação Enzimática , Inibidores Enzimáticos/química , Inibidores Enzimáticos/uso terapêutico , Humanos , Isoenzimas/genética , Isoenzimas/metabolismo , Estrutura Molecular , Família Multigênica , Neoplasias/tratamento farmacológico , Oligonucleotídeos Antissenso/uso terapêutico , Proteína Quinase C/genética , Proteína Quinase C/metabolismo , Transdução de Sinais/fisiologia
20.
Mol Cell Biol ; 26(23): 8953-63, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17000765

RESUMO

Ral GTPase activity is a crucial cell-autonomous factor supporting tumor initiation and progression. To decipher pathways impacted by Ral, we have generated null and hypomorph alleles of the Drosophila melanogaster Ral gene. Ral null animals were not viable. Reduced Ral expression in cells of the sensory organ lineage had no effect on cell division but led to postmitotic cell-specific apoptosis. Genetic epistasis and immunofluorescence in differentiating sensory organs suggested that Ral activity suppresses c-Jun N-terminal kinase (JNK) activation and induces p38 mitogen-activated protein (MAP) kinase activation. HPK1/GCK-like kinase (HGK), a MAP kinase kinase kinase kinase that can drive JNK activation, was found as an exocyst-associated protein in vivo. The exocyst is a Ral effector, and the epistasis between mutants of Ral and of msn, the fly ortholog of HGK, suggest the functional relevance of an exocyst/HGK interaction. Genetic analysis also showed that the exocyst is required for the execution of Ral function in apoptosis. We conclude that in Drosophila Ral counters apoptotic programs to support cell fate determination by acting as a negative regulator of JNK activity and a positive activator of p38 MAP kinase. We propose that the exocyst complex is Ral executioner in the JNK pathway and that a cascade from Ral to the exocyst to HGK would be a molecular basis of Ral action on JNK.


Assuntos
Apoptose , Drosophila melanogaster/citologia , Drosophila melanogaster/embriologia , Proteínas de Ligação ao GTP/metabolismo , Proteína Quinase 9 Ativada por Mitógeno/metabolismo , Alelos , Animais , Diferenciação Celular , Linhagem da Célula , Drosophila melanogaster/enzimologia , Drosophila melanogaster/genética , Embrião não Mamífero , Ativação Enzimática , Epistasia Genética , Proteínas de Ligação ao GTP/genética , Deleção de Genes , Regulação da Expressão Gênica no Desenvolvimento , Genes Essenciais , Genes de Insetos , Imuno-Histoquímica , MAP Quinase Quinase 4/metabolismo , Microscopia de Vídeo , Proteínas Serina-Treonina Quinases/metabolismo , Órgãos dos Sentidos/embriologia , Órgãos dos Sentidos/crescimento & desenvolvimento , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...