Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
iScience ; 26(7): 107059, 2023 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-37360684

RESUMO

To address the limitation associated with degron based systems, we have developed iTAG, a synthetic tag based on IMiDs/CELMoDs mechanism of action that improves and addresses the limitations of both PROTAC and previous IMiDs/CeLMoDs based tags. Using structural and sequence analysis, we systematically explored native and chimeric degron containing domains (DCDs) and evaluated their ability to induce degradation. We identified the optimal chimeric iTAG(DCD23 60aa) that elicits robust degradation of targets across cell types and subcellular localizations without exhibiting the well documented "hook effect" of PROTAC-based systems. We showed that iTAG can also induce target degradation by murine CRBN and enabled the exploration of natural neo-substrates that can be degraded by murine CRBN. Hence, the iTAG system constitutes a versatile tool to degrade targets across the human and murine proteome.

2.
Cancer Cell ; 37(1): 85-103.e9, 2020 01 13.
Artigo em Inglês | MEDLINE | ID: mdl-31935375

RESUMO

Despite substantial clinical benefit of targeted and immune checkpoint blockade-based therapies in melanoma, resistance inevitably develops. We show cytoskeletal remodeling and changes in expression and activity of ROCK-myosin II pathway during acquisition of resistance to MAPK inhibitors. MAPK regulates myosin II activity, but after initial therapy response, drug-resistant clones restore myosin II activity to increase survival. High ROCK-myosin II activity correlates with aggressiveness, identifying targeted therapy- and immunotherapy-resistant melanomas. Survival of resistant cells is myosin II dependent, regardless of the therapy. ROCK-myosin II ablation specifically kills resistant cells via intrinsic lethal reactive oxygen species and unresolved DNA damage and limits extrinsic myeloid and lymphoid immunosuppression. Efficacy of targeted therapies and immunotherapies can be improved by combination with ROCK inhibitors.


Assuntos
Citoesqueleto/metabolismo , Resistencia a Medicamentos Antineoplásicos , Melanoma/metabolismo , Miosina Tipo II/metabolismo , Animais , Antígeno B7-H1/metabolismo , Ciclo Celular , Linhagem Celular Tumoral , Dano ao DNA , Feminino , Humanos , Imunoterapia , Sistema de Sinalização das MAP Quinases , Masculino , Melanoma/imunologia , Melanoma/terapia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos Nus , Camundongos SCID , Estresse Oxidativo , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas B-raf/genética , Espécies Reativas de Oxigênio , Linfócitos T Reguladores/imunologia , Resultado do Tratamento , Microambiente Tumoral/imunologia , Quinases Associadas a rho/metabolismo
3.
Elife ; 5: e12994, 2016 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-26765561

RESUMO

Rho-associated kinases 1 and 2 (ROCK1/2) are Rho-GTPase effectors that control key aspects of the actin cytoskeleton, but their role in proliferation and cancer initiation or progression is not known. Here, we provide evidence that ROCK1 and ROCK2 act redundantly to maintain actomyosin contractility and cell proliferation and that their loss leads to cell-cycle arrest and cellular senescence. This phenotype arises from down-regulation of the essential cell-cycle proteins CyclinA, CKS1 and CDK1. Accordingly, while the loss of either Rock1 or Rock2 had no negative impact on tumorigenesis in mouse models of non-small cell lung cancer and melanoma, loss of both blocked tumor formation, as no tumors arise in which both Rock1 and Rock2 have been genetically deleted. Our results reveal an indispensable role for ROCK, yet redundant role for isoforms 1 and 2, in cell cycle progression and tumorigenesis, possibly through the maintenance of cellular contractility.


Assuntos
Carcinogênese , Proliferação de Células , Quinases Associadas a rho/metabolismo , Animais , Carcinoma Pulmonar de Células não Pequenas/patologia , Técnicas de Inativação de Genes , Melanoma/patologia , Camundongos , Quinases Associadas a rho/genética
4.
Mol Biol Cell ; 26(22): 4163-70, 2015 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-26310441

RESUMO

Melanoma cells can adopt two functionally distinct forms, amoeboid and mesenchymal, which facilitates their ability to invade and colonize diverse environments during the metastatic process. Using quantitative imaging of single living tumor cells invading three-dimensional collagen matrices, in tandem with unsupervised computational analysis, we found that melanoma cells can switch between amoeboid and mesenchymal forms via two different routes in shape space--an apolar and polar route. We show that whereas particular Rho-family GTPases are required for the morphogenesis of amoeboid and mesenchymal forms, others are required for transitions via the apolar or polar route and not amoeboid or mesenchymal morphogenesis per se. Altering the transition rates between particular routes by depleting Rho-family GTPases can change the morphological heterogeneity of cell populations. The apolar and polar routes may have evolved in order to facilitate conversion between amoeboid and mesenchymal forms, as cells are either searching for, or attracted to, particular migratory cues, respectively.


Assuntos
Melanoma/patologia , Animais , Bovinos , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Forma Celular/fisiologia , Colágeno/metabolismo , Humanos , Mesoderma/metabolismo , Invasividade Neoplásica , Corpos Polares/metabolismo , Transdução de Sinais , Relação Estrutura-Atividade , Proteínas rho de Ligação ao GTP/metabolismo
5.
Cancer Res ; 75(11): 2272-84, 2015 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-25840982

RESUMO

There is an urgent need to identify new therapeutic opportunities for metastatic melanoma. Fragment-based screening has led to the discovery of orally available, ATP-competitive AKT kinase inhibitors, AT13148 and CCT129254. These compounds also inhibit the Rho-kinases ROCK 1 and ROCK 2 and we show they potently inhibit ROCK activity in melanoma cells in culture and in vivo. Treatment of melanoma cells with CCT129254 or AT13148 dramatically reduces cell invasion, impairing both "amoeboid-like" and mesenchymal-like modes of invasion in culture. Intravital imaging shows that CCT129254 or AT13148 treatment reduces the motility of melanoma cells in vivo. CCT129254 inhibits melanoma metastasis when administered 2 days after orthotopic intradermal injection of the cells, or when treatment starts after metastases have arisen. Mechanistically, our data suggest that inhibition of ROCK reduces the ability of melanoma cells to efficiently colonize the lungs. These results suggest that these novel inhibitors of ROCK may be beneficial in the treatment of metastasis.


Assuntos
2-Hidroxifenetilamina/análogos & derivados , Movimento Celular/efeitos dos fármacos , Melanoma/tratamento farmacológico , Inibidores de Proteínas Quinases/administração & dosagem , Pirazóis/administração & dosagem , Quinases Associadas a rho/genética , 2-Hidroxifenetilamina/administração & dosagem , Linhagem Celular Tumoral , Humanos , Melanoma/genética , Melanoma/patologia , Invasividade Neoplásica/genética , Metástase Neoplásica , Fosforilação , Transdução de Sinais/efeitos dos fármacos , Quinases Associadas a rho/antagonistas & inibidores
6.
Nat Cell Biol ; 15(7): 860-71, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23748611

RESUMO

The way in which cells adopt different morphologies is not fully understood. Cell shape could be a continuous variable or restricted to a set of discrete forms. We developed quantitative methods to describe cell shape and show that Drosophila haemocytes in culture are a heterogeneous mixture of five discrete morphologies. In an RNAi screen of genes affecting the morphological complexity of heterogeneous cell populations, we found that most genes regulate the transition between discrete shapes rather than generating new morphologies. In particular, we identified a subset of genes, including the tumour suppressor PTEN, that decrease the heterogeneity of the population, leading to populations enriched in rounded or elongated forms. We show that these genes have a highly conserved function as regulators of cell shape in both mouse and human metastatic melanoma cells.


Assuntos
Forma Celular , Drosophila melanogaster/metabolismo , Genes Supressores de Tumor , Testes Genéticos , Melanoma/secundário , Interferência de RNA , Animais , Drosophila melanogaster/genética , Humanos , Melanoma/genética , Melanoma/metabolismo , Camundongos , PTEN Fosfo-Hidrolase/genética , Fenótipo , Células Tumorais Cultivadas
7.
J Cell Sci ; 124(Pt 8): 1256-67, 2011 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-21444753

RESUMO

Melanoma cells can switch between an elongated mesenchymal-type and a rounded amoeboid-type migration mode. The rounded 'amoeboid' form of cell movement is driven by actomyosin contractility resulting in membrane blebbing. Unlike elongated A375 melanoma cells, rounded A375 cells do not display any obvious morphological front-back polarisation, although polarisation is thought to be a prerequisite for cell movement. We show that blebbing A375 cells are polarised, with ezrin (a linker between the plasma membrane and actin cytoskeleton), F-actin, myosin light chain, plasma membrane, phosphatidylinositol (4,5)-bisphosphate and ß1-integrin accumulating at the cell rear in a uropod-like structure. This structure does not have the typical protruding shape of classical leukocyte uropods, but, as for those structures, it is regulated by protein kinase C. We show that the ezrin-rich uropod-like structure (ERULS) is an inherent feature of polarised A375 cells and not a consequence of cell migration, and is necessary for cell invasion. Furthermore, we demonstrate that membrane blebbing is reduced at this site, leading to a model in which the rigid ezrin-containing structure determines the direction of a moving cell through localised inhibition of membrane blebbing.


Assuntos
Membrana Celular/metabolismo , Movimento Celular , Polaridade Celular , Células/citologia , Proteínas do Citoesqueleto/metabolismo , Melanoma/fisiopatologia , Linhagem Celular Tumoral , Membrana Celular/genética , Células/metabolismo , Proteínas do Citoesqueleto/genética , Humanos , Melanoma/genética , Melanoma/metabolismo
8.
Br J Pharmacol ; 158(2): 610-20, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19732064

RESUMO

BACKGROUND AND PURPOSE: Oxaliplatin is the first platinum-based compound effective in the treatment of colorectal cancer. Oxaliplatin combined with cetuximab for metastatic colorectal cancer is under evaluation. The preliminary results seem controversial, particularly for the use of cetuximab in K-Ras mutated patients. K-Ras mutation is known to affect redox homeostasis. Here we evaluated how the efficacy of oxaliplatin alone or combined with cetuximab varied according to the Ras mutation and redox status in a panel of colorectal tumour cell lines. EXPERIMENTAL APPROACH: Viability was evaluated by methylthiazoletetrazolium assay, reactive oxygen species production by DCFDA and lucigenin on HT29-D4, Caco-2, SW480 and SW620 cell lines. KEY RESULTS: Combination of oxaliplatin and cetuximab was less cytotoxic than oxaliplatin alone in colorectal cells harbouring wild-type Ras and membrane expression of receptors for epidermal growth factor receptor (EGFR), such as HT29-D4 and Caco-2 cells. In contrast, cetuximab did not affect oxaliplatin efficiency in cells harbouring K-Ras(V12) mutation, irrespective of membrane EGFR expression (SW620 and SW480 cells). Transfection of HT29-D4 with K-Ras(V12) decreased oxaliplatin IC(50) and impaired cetuximab sensitivity, without affecting expression of membrane EGFR compared with HT29-D4 control. Oxaliplatin efficacy relies on endogenous production of H(2)O(2). Cetuximab inhibits H(2)O(2) production inhibiting the EGFR/Nox1 NADPH oxidase pathway. Oxaliplatin efficacy was impaired by short hairpin RNA for Nox1 and by catalase (H(2)O(2) scavenger). CONCLUSIONS AND IMPLICATIONS: Cetuximab limited oxaliplatin efficiency by affecting the redox status of cancer cells through Nox1. Such combined therapy might be improved by controlling H(2)O(2) elimination.


Assuntos
Anticorpos Monoclonais/farmacologia , Antineoplásicos/farmacologia , Neoplasias Colorretais/tratamento farmacológico , Compostos Organoplatínicos/farmacologia , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais Humanizados , Antineoplásicos/administração & dosagem , Células CACO-2 , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Cetuximab , Neoplasias Colorretais/genética , Antagonismo de Drogas , Receptores ErbB/genética , Regulação Neoplásica da Expressão Gênica , Genes ras/genética , Humanos , Peróxido de Hidrogênio/metabolismo , Concentração Inibidora 50 , Mutação , NADPH Oxidase 1 , NADPH Oxidases/metabolismo , Compostos Organoplatínicos/administração & dosagem , Oxaliplatina , Oxirredução/efeitos dos fármacos
9.
Int J Cancer ; 122(8): 1757-64, 2008 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-18076063

RESUMO

The catalytic subunit of the NADPH oxidase complex, Nox1 (homologue of gp91phox/Nox2), expressed mainly in intestinal epithelial and vascular smooth muscle cells, functions in innate immune defense and cell proliferation. The molecular mechanisms underlying these functions, however, are not completely understood. We measured Nox1-dependent O2- production during cell spreading on Collagen IV (Coll IV) in colon carcinoma cell lines. Knocking down Nox1 by shRNA, we showed that Nox1-dependent O2- production is activated during cell spreading after 4 hr of adhesion on Collagen IV. Nox1 activation during cell spreading relies on Rac1 activation and arachidonic metabolism. Our results showed that manoalide (a secreted phospholipase A2 inhibitor) and cinnamyl-3,4-dihydroxy-alpha-cyanocinnamate (a 12-lipoxygenase inhibitor) inhibit O2- production, cell spreading and cell proliferation in these colonic epithelial cells. 12-Lipoxygenase inhibition of ROS production and cell spreading can be reversed by adding 12-HETE, a 12-lipoxygenase product, supporting the specific effect observed with cinnamyl-3,4-dihydroxy-alpha-cyanocinnamate. In contrast, Nox1 shRNA and DPI (NADPH oxidase inhibitor) weakly affect cell spreading while inhibiting O2- production and cell proliferation. These results suggest that the 12-lipoxygenase pathway is upstream of Nox1 activation and controls cell spreading and proliferation, while Nox1 specifically affects cell proliferation.


Assuntos
Araquidonato 12-Lipoxigenase/metabolismo , Carcinoma/metabolismo , Carcinoma/patologia , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , NADPH Oxidases/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Ácido 12-Hidroxi-5,8,10,14-Eicosatetraenoico/farmacologia , Western Blotting , Ácidos Cafeicos/farmacologia , Carcinoma/enzimologia , Linhagem Celular Tumoral , Proliferação de Células , Neoplasias do Colo/enzimologia , Ativação Enzimática , Humanos , Inibidores de Lipoxigenase , NADPH Oxidase 1 , Inibidores de Fosfodiesterase/farmacologia , Inibidores de Fosfolipase A2 , RNA/isolamento & purificação , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Terpenos/farmacologia
10.
Biochim Biophys Acta ; 1783(1): 23-33, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18023288

RESUMO

Reactive oxygen species are well-known mediators of various biological responses. Recently, new homologues of the catalytic subunit of NADPH oxidase have been discovered in non-phagocytic cells. These new homologues (Nox1-Nox5) produce low levels of superoxides compared to the phagocytic homologue Nox2/gp91phox. Using Nox1 siRNA, we show that Nox1-dependent superoxide production affects the migration of HT29-D4 colonic adenocarcinoma cells on collagen-I. Nox1 inhibition or down-regulation led to a decrease of superoxide production and alpha 2 beta 1 integrin membrane availability. An addition of arachidonic acid stimulated Nox1-dependent superoxide production and HT29-D4 cell migration. Pharmacological evidences using phospholipase A2, lipoxygenases and protein kinase C inhibitors show that upstream regulation of Nox1 relies on arachidonic acid metabolism. Inhibition of 12-lipoxygenase decreased basal and arachidonic acid induced Nox1-dependent superoxide production and cell migration. Migration and ROS production inhibited by a 12-lipoxygenase inhibitor were restored by the addition of 12(S)-HETE, a downstream product of 12-lipoxygenase. Protein kinase C delta inhibition by rottlerin (and also GO6983) prevented Nox1-dependent superoxide production and inhibited cell migration, while other protein kinase C inhibitors were ineffective. We conclude that Nox1 activation by arachidonic acid metabolism occurs through 12-lipoxygenase and protein kinase C delta, and controls cell migration by affecting integrin alpha 2 subunit turn-over.


Assuntos
Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Movimento Celular , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , NADPH Oxidases/metabolismo , Superóxidos/metabolismo , Araquidonato 12-Lipoxigenase/metabolismo , Ácidos Araquidônicos/farmacologia , Membrana Celular/metabolismo , Movimento Celular/efeitos dos fármacos , Células HT29 , Humanos , Integrina alfa2beta1/metabolismo , NADPH Oxidase 1 , Proteína Quinase C-delta/metabolismo , Superóxidos/antagonistas & inibidores
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA