Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Semin Cancer Biol ; 28: 39-50, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24631836

RESUMO

Exosomes are small vesicles that derive from endosomes and are delivered by many cells, including tumor cells that are a particular rich source of exosomes. Exosomes are suggested to be the most potent intercellular communicators. Being recovered in all body fluids, they can communicate with neighboring as well as distant cells. The latter was first described for dendritic cell exosomes that can initiate T cell activation. However, tumor exosomes (TEX) may impede this crosstalk. Besides with hematopoietic cells, TEX communicate with the tumor cell itself, but also with host stroma cells and endothelial cells. This crosstalk received much attention as there is strong evidence that TEX account for angiogenesis and premetastatic niche formation, which may proceed directly via binding and uptake of TEX by cells in the premetastatic organ or indirectly via TEX being taken up by hematopoietic progenitors in the bone marrow (BM), which mature toward lineages with immunosuppressive features or are forced toward premature release from the BM and homing into premetastatic organs. Knowing these deleterious activities of TEX, it becomes demanding to search for modes of therapeutic interference. I here introduce our hypothesis that metastasis formation may be hampered by tailored exosomes that outsmart TEX. The essential prerequisites are an in depth knowledge on TEX binding, uptake, binding-initiated signal transduction and uptake-promoted target cell reprogramming.


Assuntos
Exossomos/metabolismo , Neoplasias/metabolismo , Neoplasias/patologia , Animais , Transporte Biológico/fisiologia , Comunicação Celular/fisiologia , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/patologia , Humanos , Metástase Neoplásica/patologia , Transdução de Sinais/fisiologia , Células Estromais/metabolismo , Células Estromais/patologia
2.
Cell Commun Signal ; 13: 29, 2015 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-26054340

RESUMO

BACKGROUND: Claudin-7 (cld7), a tight junction (TJ) component, is also found basolaterally and in the cytoplasm. Basolaterally located cld7 is enriched in glycolipid-enriched membrane domains (GEM), where it associates with EpCAM (EpC). The conditions driving cld7 out of TJ into GEM, which is associated with a striking change in function, were not defined. Thus, we asked whether cld7 serines or palmitoylation affect cld7 location and protein, particularly EpCAM, associations. RESULTS: HEK cells were transfected with EpCAM and wild type cld7 or cld7, where serine phopsphorylation or the palmitoylation sites (AA184, AA186) (cld7(mPalm)) were mutated. Exchange of individual serine phosphorylation sites did not significantly affect the GEM localization and the EpCAM association. Instead, cld7(mPalm) was poorly recruited into GEM. This has consequences on migration and invasiveness as palmitoylated cld7 facilitates integrin and EpCAM recruitment, associates with cytoskeletal linker proteins and cooperates with MMP14, CD147 and TACE, which support motility, matrix degradation and EpCAM cleavage. On the other hand, only cld7(mPalm) associates with TJ proteins. CONCLUSION: Cld7 palmitoylation prohibits TJ integration and fosters GEM recruitment. Via associated molecules, palmitoylated cld7 supports motility and invasion.


Assuntos
Movimento Celular , Claudinas/metabolismo , Lipoilação , Microdomínios da Membrana/metabolismo , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Adesão Celular , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Linhagem Celular , Claudinas/genética , Molécula de Adesão da Célula Epitelial , Humanos , Mutação , Metástase Neoplásica/patologia , Junções Íntimas/metabolismo , Transfecção
3.
Int J Cancer ; 133(4): 855-66, 2013 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-23390083

RESUMO

Pancreatic cancer has a dismal prognosis because of early metastatic spread, a suggested feature of cancer-initiating cells (CIC). To control for a functional contribution of the pancreatic CIC-marker EpCAM, we explored metastasis formation by a stable EpCAM-knockdown (ASML-EpC(kd)) of the rat pancreatic adenocarcinoma line BSp73ASML (ASML(wt)). As EpCAM associates with claudin-7, an ASML-claudin-7-knockdown (ASML-cld7(kd)) was included to differentiate between EpC- and EpC-cld7-mediated effects. The metastatic capacity of ASML-EpC(kd) and more pronounced ASML-cld7(kd) cells is strikingly reduced. EpC-associated cld7 interferes with EpC-mediated cell-cell adhesion and supports migration. This requires cld7 phosphorylation and formation of an EpC-cld7-tetraspanin-alpha6beta4 complex in glycolipid-enriched membrane domains (GEM), where cld7 associates via the tetraspanin-alpha6beta4 complex with phosphorylated ezrin. The association of cld7 with alpha6beta4 and cytoskeleton strongly stimulates tumor cell migration. However, EpC does not actively contribute. Instead, GEM-located cld7 associates with presenilin-2, which facilitates EpC cleavage and thereby tumor cell proliferation. Finally, the EpC-cld7 complex promotes drug resistance. Both EpC and cld7 support MAPK and JNK activation, such that in ASML-EpC(kd) and ASML-cld7(kd) cells an undue expansion of proapoptotic molecules is observed. Only cld7 promotes activation of the PI3K/Akt pathway by a strong downregulation of Pten. Accordingly, cisplatin treatment prolongs the survival time of ASML-cld7(kd)-bearing rats. Taken together, cld7 supports tumorigenic features of EpC by provoking EpC cleavage and thereby its cotranscription factor activity. On the other hand, only cld7 is directly engaged in motility and apoptosis resistance. Thus, at least in concern of migrating CIC, it is cld7 that acts as a CIC biomarker.


Assuntos
Antígenos de Neoplasias/fisiologia , Moléculas de Adesão Celular/fisiologia , Resistencia a Medicamentos Antineoplásicos , Metástase Linfática , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/patologia , Animais , Apoptose , Western Blotting , Adesão Celular , Molécula de Adesão da Célula Epitelial , Citometria de Fluxo , Imunoprecipitação , Fosforilação , Ratos
4.
Oncotarget ; 7(21): 30659-77, 2016 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-27120791

RESUMO

In epithelial cells claudin7 (cld7) is a major component of tight junctions, but is also recovered from glycolipid-enriched membrane microdomains (GEM). In tumor cells, too, cld7 exists in two stages. Only GEM-located cld7, which is palmitoylated, promotes metastasis. Searching for the underlying mechanism(s) revealed the following.The metastatic capacity of the rat pancreatic adenocarcinoma cell line ASML is lost by a knockdown (kd) of cld7 and is not regained by rescuing cld7 with a mutated palmitoylation site (cld7mPalm). ASML-cld7kd and ASML-cld7mPalm cells show reduced motility and invasiveness. This is due to cld7, but not cld7mPalm associating with α6ß4, ezrin, uPAR and MMP14, which jointly support motility and invasion. Palmitoylated cld7 also is engaged in drug resistance by repressing Pten, allowing activation of the antiapoptotic PI3K/Akt pathway. An association of cld7mPalm with the major Pten phosphorylating kinases does not restore apoptosis resistance as phosphorylated Pten is not guided towards GEM to compete with non-phosphorylated Pten. The pathway whereby palmitoylated cld7 supports expression of several EMT genes and nuclear translocation of EMT transcription factors remains to be unraveled. An association with Notch, reduced in ASML-cld7mPalm cells, might be the starting point. Finally, GEM-located, palmitoylated cld7 associates with several components of vesicle transport machineries engaged in exosome biogenesis.Taken together, prerequisites for cld7 acting as a cancer-initiating cell marker are GEM location and palmitoylation, which support a multitude of associations and integration into exosomes. The latter suggests palmitoylated cld7 contributing to message transfer via exosomes.


Assuntos
Claudinas/metabolismo , Glicolipídeos/metabolismo , Microdomínios da Membrana/metabolismo , Proteínas de Membrana/metabolismo , Fatores de Transcrição/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Animais , Linhagem Celular Tumoral , Movimento Celular/genética , Claudinas/genética , Citosol/metabolismo , Exossomos/metabolismo , Técnicas de Silenciamento de Genes , Lipoilação , Metástase Neoplásica , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Transdução de Sinais
5.
Oncotarget ; 6(4): 2046-63, 2015 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-25514462

RESUMO

In colorectal cancer (CoCa) EpCAM is frequently associated with claudin-7. There is evidence that tumor-promoting EpCAM activities are modulated by the association with claudin-7. To support this hypothesis, claudin-7 was knocked-down (kd) in HT29 and SW948 cells. HT29-cld7kd and SW948-cld7kd cells display decreased anchorage-independent growth and the capacity for holoclone-, respectively, sphere-formation is reduced. Tumor growth is delayed and cld7kd cells poorly metastasize. In line with this, migratory and invasive potential of cld7kd clones is strongly impaired, migration being inhibited by anti-CD49c, but not anti-EpCAM, although motility is reduced in EpCAM siRNA-treated cells. This is due to claudin-7 recruiting EpCAM in glycolipid-enriched membrane fractions towards claudin-7-associated TACE and presenilin2, which cleave EpCAM. The cleaved intracellular domain, EpIC, promotes epithelial-mesenchymal transition (EMT)-associated transcription factor expression, which together with fibronectin and vimentin are reduced in claudin-7kd cells. But, uptake of HT29wt and SW948wt exosomes by the claudin-7kd lines sufficed for transcription factor upregulation and for restoring motility. Thus, claudin-7 contributes to motility and invasion and is required for recruiting EpCAM towards TACE/presenilin2. EpIC generation further supports motility by promoting a shift towards EMT. Notably, EMT features of cld7-competent metastatic CoCa cells can be transferred via exosomes to poorly metastatic cells.


Assuntos
Movimento Celular , Claudinas/metabolismo , Neoplasias Colorretais/metabolismo , Transição Epitelial-Mesenquimal , Animais , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Western Blotting , Adesão Celular , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Claudinas/genética , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Molécula de Adesão da Célula Epitelial , Exossomos/metabolismo , Células HT29 , Humanos , Camundongos SCID , Microscopia Confocal , Invasividade Neoplásica , Interferência de RNA , Esferoides Celulares/metabolismo , Transplante Heterólogo
6.
Mol Oncol ; 7(5): 917-28, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23727360

RESUMO

C4.4A is a metastasis-associated molecule that functions appear to rely on associated alph6beta4 integrin. To corroborate the impact of the C4.4A-alpha6beta4 integrin association on metastasis formation, C4.4A was knocked-down in a highly metastatic rat pancreatic adenocarcinoma (ASML, ASML-C4.4Akd). Metastasis formation by ASML-C4.4Akd cells after intrafootpad application was strongly retarded in draining nodes and lung colonization was rare. Furthermore, cisplatin treatment significantly prolonged the survival time only of ASML-C4.4Akd-bearing rats. ASML-C4.4Akd cells display reduced migratory activity and impaired matrix protein degradation due to inefficient MMP14 activation; loss of drug-resistance is due to mitigated PI3K/Akt pathway activation. These losses of function rely on the laminin receptor C4.4A recruiting activated alpha6beta4 integrin into rafts, where C4.4A cooperates with alpha6beta4 and via alpha6beta4 with MMP14. Within this raft-located complex, MMP14 provokes focalized matrix degradation and mostly alpha6beta4 integrin promotes BAD phosphorylation and upregulated Bcl2 and BclXl expression. Thus, metastasis-promoting activities of C4.4A are not genuine characteristics of C4.4A. Instead, the raft-located laminin receptor C4.4A recruits alpha6beta4 integrin and supports via the alpha6beta4 integrin MMP14 activation. Thereby C4.4A acts as a linker to facilitate several steps in the metastatic cascade. Taking the restricted C4.4A expression in non-transformed tissue, this knowledge should pave the way toward the use of C4.4A as a therapeutic target.


Assuntos
Moléculas de Adesão Celular/metabolismo , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Moléculas de Adesão Celular/genética , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Cisplatino/farmacologia , Feminino , Citometria de Fluxo , Imunofluorescência , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/metabolismo , Imuno-Histoquímica , Integrina alfa6beta4/genética , Integrina alfa6beta4/metabolismo , Laminina/genética , Laminina/metabolismo , Metaloproteinase 14 da Matriz/genética , Metaloproteinase 14 da Matriz/metabolismo , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Calinina
7.
Exp Cell Res ; 312(19): 3857-71, 2006 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-17010335

RESUMO

For the invasive migration of tumor cells, at least two mechanisms are currently discussed: (1) the mesenchymal mode depending on extracellular proteolysis and (2) the proteolysis-independent amoeboid mode depending on the activity of the Rho kinase ROCK. The ability of tumor cells to switch between different modes of motility has been shown to limit the efficiency of agents aimed to reduce invasion. Here we show by combining 2D and 3D migration assays that human mammary tumor cells exhibited a strongly reduced migration velocity as compared to their normal counterparts indicating that high invasiveness is not necessarily correlated with high migratory capacity in 2D assays. This reduced migration was apparently due to significant differences in actin organization, decreased persistence of lamellipodia by 50% and increased cell substrate adhesion. These differences resulted from a 2.5-fold higher activity of ROCK and were mediated by its downstream effectors myosin light chain kinase and cofilin. Thus, inhibition of ROCK activity caused a marked increase in 2D migration efficiency by 40%, without, however, affecting 3D invasion. A massive reduction of invasion by 60% was achieved by the simultaneous inhibition of the ROCK-dependent amoeboid and the extracellular proteolysis-dependent mesenchymal mode. These results may point to a new efficient strategy for blocking tumor cell invasion in vivo.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Fatores de Despolimerização de Actina/metabolismo , Actinas/metabolismo , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Neoplasias da Mama/fisiopatologia , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Feminino , Adesões Focais , Humanos , Técnicas In Vitro , Cadeias Leves de Miosina/metabolismo , Invasividade Neoplásica/fisiopatologia , Fenótipo , Fosforilação , Pseudópodes/fisiologia , Transdução de Sinais , Proteínas rac1 de Ligação ao GTP/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo , Quinases Associadas a rho
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA