Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Int J Cancer ; 145(4): 901-915, 2019 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-30653260

RESUMO

Endothelial lipase (LIPG) is a cell surface associated lipase that displays phospholipase A1 activity towards phosphatidylcholine present in high-density lipoproteins (HDL). LIPG was recently reported to be expressed in breast cancer and to support proliferation, tumourigenicity and metastasis. Here we show that severe oxidative stress leading to AMPK activation triggers LIPG upregulation, resulting in intracellular lipid droplet accumulation in breast cancer cells, which supports survival. Neutralizing oxidative stress abrogated LIPG upregulation and the concomitant lipid storage. In human breast cancer, high LIPG expression was observed in a limited subset of tumours and was significantly associated with shorter metastasis-free survival in node-negative, untreated patients. Moreover, expression of PLIN2 and TXNRD1 in these tumours indicated a link to lipid storage and oxidative stress. Altogether, our findings reveal a previously unrecognized role for LIPG in enabling oxidative stress-induced lipid droplet accumulation in tumour cells that protects against oxidative stress, and thus supports tumour progression.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Lipase/metabolismo , Lipídeos/fisiologia , Estresse Oxidativo/fisiologia , Linhagem Celular Tumoral , Progressão da Doença , Intervalo Livre de Doença , Feminino , Humanos , Metabolismo dos Lipídeos/fisiologia , Lipoproteínas HDL/metabolismo , Células MCF-7 , Pessoa de Meia-Idade , Regulação para Cima/fisiologia
2.
Proc Natl Acad Sci U S A ; 109(21): 8155-60, 2012 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-22570503

RESUMO

Metastasis from primary tumors remains a major problem for tumor therapy. In the search for markers of metastasis and more effective therapies, the tumor metabolome is relevant because of its importance to the malignant phenotype and metastatic capacity of tumor cells. Altered choline metabolism is a hallmark of cancer. More specifically, a decreased glycerophosphocholine (GPC) to phosphocholine (PC) ratio was reported in breast, ovarian, and prostate cancers. Improved strategies to exploit this altered choline metabolism are therefore required. However, the critical enzyme cleaving GPC to produce choline, the initial step in the pathway controlling the GPC/PC ratio, remained unknown. In the present work, we have identified the enzyme, here named EDI3 (endometrial differential 3). Purified recombinant EDI3 protein cleaves GPC to form glycerol-3-phosphate and choline. Silencing EDI3 in MCF-7 cells decreased this enzymatic activity, increased the intracellular GPC/PC ratio, and decreased downstream lipid metabolites. Downregulating EDI3 activity inhibited cell migration via disruption of the PKCα signaling pathway, with stable overexpression of EDI3 showing the opposite effect. EDI3 was originally identified in our screening study comparing mRNA levels in metastasizing and nonmetastasizing endometrial carcinomas. Both Kaplan-Meier and multivariate analyses revealed a negative association between high EDI3 expression and relapse-free survival time in both endometrial (P < 0.001) and ovarian (P = 0.029) cancers. Overall, we have identified EDI3, a key enzyme controlling GPC and choline metabolism. Because inhibition of EDI3 activity corrects the GPC/PC ratio and decreases the migration capacity of tumor cells, it represents a possible target for therapeutic intervention.


Assuntos
Neoplasias da Mama/enzimologia , Colina/metabolismo , Neoplasias do Endométrio/enzimologia , Neoplasias Ovarianas/enzimologia , Fosfolipases/metabolismo , Diester Fosfórico Hidrolases/metabolismo , Animais , Neoplasias da Mama/secundário , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Neoplasias do Endométrio/secundário , Feminino , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias Ovarianas/secundário , Fosfolipases/genética , Diester Fosfórico Hidrolases/genética , Proteína Quinase C-alfa/metabolismo , Transdução de Sinais/fisiologia
3.
Small GTPases ; 10(4): 289-295, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-28301299

RESUMO

PAK1 and PAK4 are members of the p-21 activated kinase family of serine/threonine kinases. PAK1 has previously been implicated in both the formation and disassembly of invasive cell protrusions, termed invadopodia. We recently reported a novel role for PAK4 during invadopodia maturation and confirmed a specific role for PAK1 in invadopodia formation; findings we will review here. Moreover, we found that PAK4 induction of maturation is delivered via interaction with the RhoA regulator PDZ-RhoGEF. We can now reveal that loss of PAK4 expression leads to changes in invadopodia dynamics. Ultimately we propose that PAK4 but not PAK1 is a key mediator of RhoA activity and provide further evidence that modulation of PAK4 expression levels leads to changes in RhoA activity.


Assuntos
Melanoma/metabolismo , Podossomos/metabolismo , Quinases Ativadas por p21/metabolismo , Linhagem Celular , Humanos , Melanoma/genética , Invasividade Neoplásica , Fatores de Troca de Nucleotídeo Guanina Rho/metabolismo , Quinases Ativadas por p21/genética , Proteína rhoA de Ligação ao GTP/metabolismo
4.
Cancer Res ; 77(17): 4589-4601, 2017 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-28652252

RESUMO

Glycerophosphodiesterase EDI3 (GPCPD1; GDE5; GDPD6) has been suggested to promote cell migration, adhesion, and spreading, but its mechanisms of action remain uncertain. In this study, we targeted the glycerol-3-phosphate acyltransferase GPAM along with choline kinase-α (CHKA), the enzymes that catabolize the products of EDI3 to determine which downstream pathway is relevant for migration. Our results clearly showed that GPAM influenced cell migration via the signaling lipid lysophosphatidic acid (LPA), linking it with GPAM to cell migration. Analysis of GPAM expression in different cancer types revealed a significant association between high GPAM expression and reduced overall survival in ovarian cancer. Silencing GPAM in ovarian cancer cells decreased cell migration and reduced the growth of tumor xenografts. In contrast to these observations, manipulating CHKA did not influence cell migration in the same set of cell lines. Overall, our findings show how GPAM influences intracellular LPA levels to promote cell migration and tumor growth. Cancer Res; 77(17); 4589-601. ©2017 AACR.


Assuntos
Movimento Celular , Colina Quinase/metabolismo , Glicerol-3-Fosfato O-Aciltransferase/metabolismo , Neoplasias Ovarianas/mortalidade , Neoplasias Ovarianas/patologia , Animais , Feminino , Humanos , Camundongos , Camundongos Nus , Neoplasias Ovarianas/enzimologia , Prognóstico , Transdução de Sinais , Taxa de Sobrevida , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Oncotarget ; 7(43): 70881-70897, 2016 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-27765920

RESUMO

Cancer cells are thought to use actin rich invadopodia to facilitate matrix degradation. Formation and maturation of invadopodia requires the co-ordained activity of Rho-GTPases, however the molecular mechanisms that underlie the invadopodia lifecycle are not fully elucidated. Previous work has suggested a formation and disassembly role for Rho family effector p-21 activated kinase 1 (PAK1) however, related family member PAK4 has not been explored. Systematic analysis of isoform specific depletion using in vitro and in vivo invasion assays revealed there are differential invadopodia-associated functions. We consolidated a role for PAK1 in the invadopodia formation phase and identified PAK4 as a novel invadopodia protein that is required for successful maturation. Furthermore, we find that PAK4 (but not PAK1) mediates invadopodia maturation likely via inhibition of PDZ-RhoGEF. Our work points to an essential role for both PAKs during melanoma invasion but provides a significant advance in our understanding of differential PAK function.


Assuntos
Melanoma/patologia , Podossomos/patologia , Fatores de Troca de Nucleotídeo Guanina Rho/metabolismo , Neoplasias Cutâneas/patologia , Quinases Ativadas por p21/metabolismo , Actinas , Animais , Linhagem Celular Tumoral , Imunofluorescência , Células HEK293 , Humanos , Invasividade Neoplásica/patologia , Isoformas de Proteínas/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Peixe-Zebra , Quinases Ativadas por p21/genética
6.
Cell Adh Migr ; 8(5): 499-508, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25482527

RESUMO

Endometrial carcinoma differential 3 (EDI3) was the first member of the glycerophosphodiesterase (GDE) protein family shown to be associated with cancer. Our initial work demonstrated that endometrial and ovarian cancer patients with primary tumors overexpressing EDI3 had a higher risk of developing metastasis and decreased survival. Further analysis indicated that EDI3 cleaves glycerophosphocholine to choline and glycerol-3-phosphate, increases the levels of active PKC, and enhances the migratory activity of tumor cells. Despite these initial findings, EDI3 remained mainly uncharacterized. Therefore, to obtain an overview of processes in which EDI3 may be involved, gene array analysis was performed using MCF-7 breast cancer cells after EDI3 knockdown compared with a non-targeting control siRNA. Several biological motifs were altered, including an enrichment of genes involved in integrin-mediated signaling. More specifically, silencing of EDI3 in MCF-7 and OVCAR-3 cells was associated with reduced expression of the key receptor subunit integrin ß1, leading to decreased cell attachment and spreading accompanied by delayed formation of cell protrusions. To confirm these results, we stably overexpressed EDI3 in MCF-7 cells which led to elevated integrin ß1 expression associated with enhanced cell attachment and spreading - two processes critical for metastasis. In conclusion, our data provide further insight into the role of EDI3 during cancer progression.


Assuntos
Adesão Celular/fisiologia , Movimento Celular/fisiologia , Colina/metabolismo , Integrinas/metabolismo , Fosfolipases/metabolismo , Adesão Celular/genética , Movimento Celular/genética , Humanos , Integrinas/genética , Células MCF-7 , Fosfolipases/genética
7.
Cell Cycle ; 11(24): 4499-506, 2012 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-23114620

RESUMO

Recently, EDI3 was identified as a key factor for choline metabolism that controls tumor cell migration and is associated with metastasis in endometrial carcinomas. EDI3 cleaves glycerophosphocholine (GPC) to form choline and glycerol-3-phosphate (G3P). Choline is then further metabolized to phosphatidylcholine (PtdC), the major lipid in membranes and a key player in membrane-mediated cell signaling. The second product, G3P, is a precursor molecule for several lipids with central roles in signaling, for example lysophosphatidic acid (LPA), phosphatidic acid (PA) and diacylglycerol (DAG). LPA activates intracellular signaling pathways by binding to specific LPA receptors, including membrane-bound G protein-coupled receptors and the intracellular nuclear receptor, PPARγ. Conversely, PA and DAG mediate signaling by acting as lipid anchors that bind and activate several signaling proteins. For example, binding of GTPases and PKC to PA and DAG, respectively, increases the activation of signaling networks, mediating processes such as migration, adhesion, proliferation or anti-apoptosis-all relevant for tumor development. We present a concept by which EDI3 either directly generates signaling molecules or provides "membrane anchors" for downstream signaling factors. As a result, EDI3 links choline metabolism to signaling activities resulting in a more malignant phenotype.


Assuntos
Colina/metabolismo , Neoplasias/enzimologia , Neoplasias/metabolismo , Diester Fosfórico Hidrolases/metabolismo , Animais , Diglicerídeos/metabolismo , Humanos , Lisofosfolipídeos/metabolismo , Modelos Biológicos , Ácidos Fosfatídicos/metabolismo , Transdução de Sinais/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA