Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
STAR Protoc ; 3(2): 101438, 2022 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-35707685

RESUMO

The various stages of epithelial-mesenchymal transition (EMT) generate phenotypically heterogeneous populations of cells. Here, we detail a dual recombinase lineage tracing system using a transgenic mouse model of metastatic breast cancer to trace and characterize breast cancer cells at different EMT stages. We describe analytical steps to label cancer cells at an early partial or a late full EMT state, followed by tracking their behavior in tumor slice cultures. We then characterize their transcriptome by five-cell RNA sequencing. For complete details on the use and execution of this protocol, please refer to Luond et al. (2021).


Assuntos
Transição Epitelial-Mesenquimal , Neoplasias , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Transição Epitelial-Mesenquimal/genética , Camundongos , Camundongos Transgênicos , Transcriptoma
2.
Dev Cell ; 56(23): 3203-3221.e11, 2021 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-34847378

RESUMO

Epithelial-mesenchymal transition (EMT) is a transient, reversible process of cell de-differentiation where cancer cells transit between various stages of an EMT continuum, including epithelial, partial EMT, and mesenchymal cell states. We have employed Tamoxifen-inducible dual recombinase lineage tracing systems combined with live imaging and 5-cell RNA sequencing to track cancer cells undergoing partial or full EMT in the MMTV-PyMT mouse model of metastatic breast cancer. In primary tumors, cancer cells infrequently undergo EMT and mostly transition between epithelial and partial EMT states but rarely reach full EMT. Cells undergoing partial EMT contribute to lung metastasis and chemoresistance, whereas full EMT cells mostly retain a mesenchymal phenotype and fail to colonize the lungs. However, full EMT cancer cells are enriched in recurrent tumors upon chemotherapy. Hence, cancer cells in various stages of the EMT continuum differentially contribute to hallmarks of breast cancer malignancy, such as tumor invasion, metastasis, and chemoresistance.


Assuntos
Biomarcadores Tumorais/metabolismo , Neoplasias da Mama/patologia , Resistencia a Medicamentos Antineoplásicos , Transição Epitelial-Mesenquimal , Regulação Neoplásica da Expressão Gênica , Neoplasias Pulmonares/secundário , Animais , Antineoplásicos/farmacologia , Apoptose , Biomarcadores Tumorais/genética , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Movimento Celular , Proliferação de Células , Feminino , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Invasividade Neoplásica , Análise de Sequência de RNA , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Proc Natl Acad Sci U S A ; 118(34)2021 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-34408016

RESUMO

During malignant progression, epithelial cancer cells dissolve their cell-cell adhesion and gain invasive features. By virtue of its dual function, ß-catenin contributes to cadherin-mediated cell-cell adhesion, and it determines the transcriptional output of Wnt signaling: via its N terminus, it recruits the signaling coactivators Bcl9 and Pygopus, and via the C terminus, it interacts with the general transcriptional machinery. This duality confounds the simple loss-of-function analysis of Wnt signaling in cancer progression. In many cancer types including breast cancer, the functional contribution of ß-catenin's transcriptional activities, as compared to its adhesion functions, to tumor progression has remained elusive. Employing the mouse mammary tumor virus (MMTV)-PyMT mouse model of metastatic breast cancer, we compared the complete elimination of ß-catenin with the specific ablation of its signaling outputs in mammary tumor cells. Notably, the complete lack of ß-catenin resulted in massive apoptosis of mammary tumor cells. In contrast, the loss of ß-catenin's transcriptional activity resulted in a reduction of primary tumor growth, tumor invasion, and metastasis formation in vivo. These phenotypic changes were reflected by stalled cell cycle progression and diminished epithelial-mesenchymal transition (EMT) and cell migration of breast cancer cells in vitro. Transcriptome analysis revealed subsets of genes which were specifically regulated by ß-catenin's transcriptional activities upon stimulation with Wnt3a or during TGF-ß-induced EMT. Our results uncouple the signaling from the adhesion function of ß-catenin and underline the importance of Wnt/ß-catenin-dependent transcription in malignant tumor progression of breast cancer.


Assuntos
Adesão Celular/fisiologia , Neoplasias Mamárias Animais/metabolismo , Transdução de Sinais/fisiologia , Proteína Wnt3A/metabolismo , beta Catenina/metabolismo , Animais , Apoptose , Ciclo Celular , Movimento Celular , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Neoplasias Mamárias Animais/genética , Camundongos , Camundongos Transgênicos , Invasividade Neoplásica , Metástase Neoplásica , Transcriptoma , Fator de Crescimento Transformador beta/farmacologia , Proteína Wnt3A/genética , beta Catenina/genética
4.
Cancer Res ; 80(17): 3631-3648, 2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32586983

RESUMO

Pygopus 2 (Pygo2) is a coactivator of Wnt/ß-catenin signaling that can bind bi- or trimethylated lysine 4 of histone-3 (H3K4me2/3) and participate in chromatin reading and writing. It remains unknown whether the Pygo2-H3K4me2/3 association has a functional relevance in breast cancer progression in vivo. To investigate the functional relevance of histone-binding activity of Pygo2 in malignant progression of breast cancer, we generated a knock-in mouse model where binding of Pygo2 to H3K4me2/3 was rendered ineffective. Loss of Pygo2-histone interaction resulted in smaller, differentiated, and less metastatic tumors, due, in part, to decreased canonical Wnt/ß-catenin signaling. RNA- and ATAC-sequencing analyses of tumor-derived cell lines revealed downregulation of TGFß signaling and upregulation of differentiation pathways such as PDGFR signaling. Increased differentiation correlated with a luminal cell fate that could be reversed by inhibition of PDGFR activity. Mechanistically, the Pygo2-histone interaction potentiated Wnt/ß-catenin signaling, in part, by repressing the expression of Wnt signaling antagonists. Furthermore, Pygo2 and ß-catenin regulated the expression of miR-29 family members, which, in turn, repressed PDGFR expression to promote dedifferentiation of wild-type Pygo2 mammary epithelial tumor cells. Collectively, these results demonstrate that the histone binding function of Pygo2 is important for driving dedifferentiation and malignancy of breast tumors, and loss of this binding activates various differentiation pathways that attenuate primary tumor growth and metastasis formation. Interfering with the Pygo2-H3K4me2/3 interaction may therefore serve as an attractive therapeutic target for metastatic breast cancer. SIGNIFICANCE: Pygo2 represents a potential therapeutic target in metastatic breast cancer, as its histone-binding capability promotes ß-catenin-mediated Wnt signaling and transcriptional control in breast cancer cell dedifferentiation, EMT, and metastasis.


Assuntos
Desdiferenciação Celular/fisiologia , Regulação Neoplásica da Expressão Gênica/fisiologia , Histonas/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Neoplasias Mamárias Experimentais/patologia , Animais , Progressão da Doença , Feminino , Técnicas de Introdução de Genes , Camundongos , Camundongos Endogâmicos C57BL
5.
Oncogene ; 39(7): 1498-1513, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31666683

RESUMO

An epithelial-mesenchymal transition (EMT) represents a basic morphogenetic process of high cell plasticity underlying embryogenesis, wound healing, cancer metastasis and drug resistance. It involves a profound transcriptional and epigenetic reprogramming of cells. A critical role of epigenetic modifiers and their specific chromatin modifications has been demonstrated during EMT. However, it has remained elusive whether epigenetic control differs between the dynamic cell state transitions of reversible EMT and the fixed differentiation status of irreversible EMT. We have employed varying EMT models of murine breast cancer cells to identify the key players establishing epithelial-mesenchymal cell plasticity during reversible and irreversible EMT. We demonstrate that the Mbd3/NuRD complex and the activities of histone deacetylases (HDACs), and Tet2 hydroxylase play a critical role in keeping cancer cells in a highly metastatic mesenchymal state. Combinatorial interference with their functions leads to mesenchymal-epithelial transition (MET) and efficiently represses metastasis formation by invasive murine and human breast cancer cells in vivo.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Transição Epitelial-Mesenquimal , Histona Desacetilases/metabolismo , Complexo Mi-2 de Remodelação de Nucleossomo e Desacetilase/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Animais , Carcinogênese , Linhagem Celular Tumoral , Proliferação de Células , Dioxigenases , Humanos , Neoplasias Mamárias Experimentais/metabolismo , Neoplasias Mamárias Experimentais/patologia , Camundongos , Metástase Neoplásica
6.
Cancer Cell ; 35(1): 17-32.e6, 2019 01 14.
Artigo em Inglês | MEDLINE | ID: mdl-30645973

RESUMO

Cancer cell plasticity facilitates the development of therapy resistance and malignant progression. De-differentiation processes, such as an epithelial-mesenchymal transition (EMT), are known to enhance cellular plasticity. Here, we demonstrate that cancer cell plasticity can be exploited therapeutically by forcing the trans-differentiation of EMT-derived breast cancer cells into post-mitotic and functional adipocytes. Delineation of the molecular pathways underlying such trans-differentiation has motivated a combination therapy with MEK inhibitors and the anti-diabetic drug Rosiglitazone in various mouse models of murine and human breast cancer in vivo. This combination therapy provokes the conversion of invasive and disseminating cancer cells into post-mitotic adipocytes leading to the repression of primary tumor invasion and metastasis formation.


Assuntos
Adipócitos/citologia , Neoplasias da Mama/tratamento farmacológico , Transdiferenciação Celular/efeitos dos fármacos , Flavonoides/administração & dosagem , Metástase Neoplásica/tratamento farmacológico , Rosiglitazona/administração & dosagem , Células 3T3-L1 , Adipogenia , Animais , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Feminino , Flavonoides/farmacologia , Humanos , Camundongos , Transplante de Neoplasias , Proteínas Proto-Oncogênicas c-met/metabolismo , Rosiglitazona/uso terapêutico , Transdução de Sinais/efeitos dos fármacos , Fator de Crescimento Transformador beta/metabolismo
7.
Small ; 14(8)2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29356374

RESUMO

While nanoparticles are an increasingly popular choice for labeling and tracking stem cells in biomedical applications such as cell therapy, their intracellular fate and subsequent effect on stem cell differentiation remain elusive. To establish an effective stem cell labeling strategy, the intracellular nanocrystal concentration should be minimized to avoid adverse effects, without compromising the intensity and persistence of the signal necessary for long-term tracking. Here, the use of second-harmonic generating barium titanate nanocrystals is reported, whose achievable brightness allows for high contrast stem cell labeling with at least one order of magnitude lower intracellular nanocrystals than previously reported. Their long-term photostability enables to investigate quantitatively at the single cell level their cellular fate in hematopoietic stem cells (HSCs) using both multiphoton and electron microscopy. It is found that the concentration of nanocrystals in proliferative multipotent progenitors is over 2.5-fold greater compared to quiescent stem cells; this difference vanishes when HSCs enter a nonquiescent, proliferative state, while their potency remains unaffected. Understanding the nanoparticle stem cell interaction allows to establish an effective and safe nanoparticle labeling strategy into somatic stem cells that can critically contribute to an understanding of their in vivo therapeutic potential.


Assuntos
Compostos de Bário/química , Células-Tronco Hematopoéticas/citologia , Nanopartículas de Magnetita/química , Nanopartículas/química , Microscopia de Geração do Segundo Harmônico/métodos , Titânio/química , Células-Tronco Adultas/citologia , Animais , Diferenciação Celular/fisiologia , Divisão Celular/fisiologia , Proliferação de Células/fisiologia , Humanos
8.
Mol Biol Cell ; 25(17): 2556-70, 2014 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-24989798

RESUMO

Basal-like breast carcinomas, characterized by unfavorable prognosis and frequent metastases, are associated with epithelial-to-mesenchymal transition. During this process, cancer cells undergo cytoskeletal reorganization and up-regulate membrane-type 1 matrix metalloproteinase (MT1-MMP; MMP14), which functions in actin-based pseudopods to drive invasion by extracellular matrix degradation. However, the mechanisms that couple matrix proteolysis to the actin cytoskeleton in cell invasion have remained unclear. On the basis of a yeast two-hybrid screen for the MT1-MMP cytoplasmic tail-binding proteins, we identify here a novel Src-regulated protein interaction between the dynamic cytoskeletal scaffold protein palladin and MT1-MMP. These proteins were coexpressed in invasive human basal-like breast carcinomas and corresponding cell lines, where they were associated in the same matrix contacting and degrading membrane complexes. The silencing and overexpression of the 90-kDa palladin isoform revealed the functional importance of the interaction with MT1-MMP in pericellular matrix degradation and mesenchymal tumor cell invasion, whereas in MT1-MMP-negative cells, palladin overexpression was insufficient for invasion. Moreover, this invasion was inhibited in a dominant-negative manner by an immunoglobulin domain-containing palladin fragment lacking the dynamic scaffold and Src-binding domains. These results identify a novel protein interaction that links matrix degradation to cytoskeletal dynamics and migration signaling in mesenchymal cell invasion.


Assuntos
Neoplasias da Mama/metabolismo , Carcinoma/metabolismo , Proteínas do Citoesqueleto/fisiologia , Citoesqueleto/metabolismo , Matriz Extracelular/metabolismo , Metaloproteinase 14 da Matriz/metabolismo , Invasividade Neoplásica , Fosfoproteínas/fisiologia , Actinas/metabolismo , Neoplasias da Mama/patologia , Carcinoma/patologia , Linhagem Celular Tumoral , Conectina/metabolismo , Transição Epitelial-Mesenquimal , Feminino , Humanos , Proteínas dos Microfilamentos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteólise
9.
Cell Mol Life Sci ; 71(19): 3685-710, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24794629

RESUMO

The erythropoietin-producing hepatocellular (Eph) receptors comprise the largest family of receptor tyrosine kinases (RTKs). Initially regarded as axon-guidance and tissue-patterning molecules, Eph receptors have now been attributed with various functions during development, tissue homeostasis, and disease pathogenesis. Their ligands, ephrins, are synthesized as membrane-associated molecules. At least two properties make this signaling system unique: (1) the signal can be simultaneously transduced in the receptor- and the ligand-expressing cell, (2) the signaling outcome through the same molecules can be opposite depending on cellular context. Moreover, shedding of Eph and ephrin ectodomains as well as ligand-dependent and -independent receptor crosstalk with other RTKs, proteases, and adhesion molecules broadens the repertoire of Eph/ephrin functions. These integrated pathways provide plasticity to cell-microenvironment communication in varying tissue contexts. The complex molecular networks and dynamic cellular outcomes connected to the Eph/ephrin signaling in tumor-host communication and stem cell niche are the main focus of this review.


Assuntos
Efrinas/metabolismo , Neoplasias/metabolismo , Células-Tronco Neoplásicas/metabolismo , Receptores da Família Eph/metabolismo , Polaridade Celular , Humanos , Integrinas/metabolismo , Metaloproteases/metabolismo , Metástase Neoplásica , Neoplasias/patologia , Células-Tronco Neoplásicas/citologia , Transdução de Sinais , Proteínas rho de Ligação ao GTP/metabolismo
10.
Curr Alzheimer Res ; 11(2): 110-8, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24156269

RESUMO

The prediction of efficacy in long-term treatment of acetylcholinesterase inhibitors (AChEIs) is a major clinical issue, although no consistently strong predictive factors have emerged thus far. The present analyses aimed to identify factors for predicting long-term outcome of galantamine treatment. Analyses were conducted with data from a 24 weeks randomized, double-blind, placebo controlled trial to evaluate the efficacy and the safety of galantamine in the treatment of 303 patients with mild to moderate AD. Patients were divided into responders (4 or more point improvement of ADAScog scores at 24 weeks of treatment) and non-responders. We explored whether patients' background (e.g. sex, age, and duration of disease) and scores of cognitive scales at early stage, are relevant to the long-term response to AChEIs. Predictive values were estimated by the logistic regression model. The responder rate was 31.7%. We found that changes in scores of ADAS-J cog subscales between week 4 and baseline, especially word recognition, can be a good variable to predict subsequent response to galantamine, with approximately 75% of predictive performance. Characteristics of patients, including demographic characteristics, severity of disease and neuropsychological features before treatment were poorly predictive. The present study indicate that initial response to galantamine administration in patients with mild to moderate AD seems to be a reliable predictor of response of consequent galantamine treatment. Patients who show improvement of episodic memory function during the first 4 weeks of galantamine administration may be likely to particularly benefit from galantamine treatment.


Assuntos
Doença de Alzheimer/diagnóstico , Doença de Alzheimer/tratamento farmacológico , Inibidores da Colinesterase/uso terapêutico , Galantamina/uso terapêutico , Método Duplo-Cego , Humanos , Valor Preditivo dos Testes , Estudos Retrospectivos , Resultado do Tratamento
12.
J Cell Biol ; 201(3): 467-84, 2013 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-23629968

RESUMO

Changes in EphA2 signaling can affect cancer cell-cell communication and motility through effects on actomyosin contractility. However, the underlying cell-surface interactions and molecular mechanisms of how EphA2 mediates these effects have remained unclear. We demonstrate here that EphA2 and membrane-anchored membrane type-1 matrix metalloproteinase (MT1-MMP) were selectively up-regulated and coexpressed in invasive breast carcinoma cells, where, upon physical interaction in same cell-surface complexes, MT1-MMP cleaved EphA2 at its Fibronectin type-III domain 1. This cleavage, coupled with EphA2-dependent Src activation, triggered intracellular EphA2 translocation, as well as an increase in RhoA activity and cell junction disassembly, which suggests an overall repulsive effect between cells. Consistent with this, cleavage-prone EphA2-D359I mutant shifted breast carcinoma cell invasion from collective to rounded single-cell invasion within collagen and in vivo. Up-regulated MT1-MMP also codistributed with intracellular EphA2 in invasive cells within human breast carcinomas. These results reveal a new proteolytic regulatory mechanism of cell-cell signaling in cancer invasion.


Assuntos
Neoplasias da Mama/patologia , Carcinoma Ductal de Mama/secundário , Metaloproteinase 14 da Matriz/metabolismo , Receptor EphA2/metabolismo , Sequência de Aminoácidos , Animais , Neoplasias da Mama/enzimologia , Carcinoma Ductal de Mama/enzimologia , Linhagem Celular Tumoral , Forma Celular , Colágeno/metabolismo , Feminino , Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Metástase Linfática , Metaloproteinase 14 da Matriz/genética , Camundongos , Camundongos SCID , Dados de Sequência Molecular , Invasividade Neoplásica , Transplante de Neoplasias , Estrutura Terciária de Proteína , Transporte Proteico , Proteólise , RNA Interferente Pequeno/genética , Receptor EphA2/química , Receptor EphA2/genética , Análise de Célula Única , Imagem com Lapso de Tempo , Análise Serial de Tecidos , Proteína rhoA de Ligação ao GTP/metabolismo
13.
Cell Host Microbe ; 10(6): 577-90, 2011 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-22177562

RESUMO

Kaposi sarcoma (KS), an angioproliferative disease associated with Kaposi sarcoma herpesvirus (KSHV) infection, harbors a diversity of cell types ranging from endothelial to mesenchymal cells of unclear origin. We developed a three-dimensional cell model for KSHV infection and used it to demonstrate that KSHV induces transcriptional reprogramming of lymphatic endothelial cells to mesenchymal cells via endothelial-to-mesenchymal transition (EndMT). KSHV-induced EndMT was initiated by the viral proteins vFLIP and vGPCR through Notch pathway activation, leading to gain of membrane-type-1 matrix metalloproteinase (MT1-MMP)-dependent invasive properties and concomitant changes in viral gene expression. Mesenchymal markers and MT1-MMP were found codistributed with a KSHV marker in the same cells from primary KS biopsies. Our data explain the heterogeneity of cell types within KS lesions and suggest that KSHV-induced EndMT may contribute to KS development by giving rise to infected, invasive cells while providing the virus a permissive cellular microenvironment for efficient spread.


Assuntos
Transição Epitelial-Mesenquimal , Herpesvirus Humano 8/fisiologia , Metaloproteinase 14 da Matriz/metabolismo , Receptores Notch/metabolismo , Sarcoma de Kaposi/enzimologia , Sarcoma de Kaposi/fisiopatologia , Linhagem Celular , Células Endoteliais/citologia , Células Endoteliais/enzimologia , Células Endoteliais/virologia , Regulação Viral da Expressão Gênica , Herpesvirus Humano 8/genética , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/enzimologia , Células-Tronco Mesenquimais/virologia , Sarcoma de Kaposi/metabolismo , Sarcoma de Kaposi/virologia , Transdução de Sinais , Proteínas Virais/genética , Proteínas Virais/metabolismo
14.
Cancer Res ; 70(20): 7851-61, 2010 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-20876804

RESUMO

Aberrant expression and polymorphism of fibroblast growth factor receptor 4 (FGFR4) has been linked to tumor progression and anticancer drug resistance. We describe here a novel mechanism of tumor progression by matrix degradation involving epithelial-to-mesenchymal transition in response to membrane-type 1 matrix metalloproteinase (MT1-MMP, MMP-14) induction at the edge of tumors expressing the FGFR4-R388 risk variant. Both FGFR4 and MT1-MMP were upregulated in tissue biopsies from several human cancer types including breast adenocarcinomas, where they were partially coexpressed at the tumor/stroma border and tumor invasion front. The strongest overall coexpression was found in prostate carcinoma. Studies with cultured prostate carcinoma cell lines showed that the FGFR4-R388 variant, which has previously been associated with poor cancer prognosis, increased MT1-MMP-dependent collagen invasion. In this experimental model, knockdown of FGFR4-R388 or MT1-MMP by RNA interference blocked tumor cell invasion and growth in collagen. This was coupled with impaired phosphorylation of FGFR substrate 2 and Src, upregulation of E-cadherin, and suppression of cadherin-11 and N-cadherin. These in vitro results were substantiated by reduced MT1-MMP content and in vivo growth of prostate carcinoma cells after the FGFR4-R388 gene silencing. In contrast, knockdown of the alternative FGFR4-G388 allele enhanced MT1-MMP and invasive tumor cell growth in vivo and within three-dimensional collagen. These results will help to explain the reported association of the FGFR4-R388 variant with the progression and poor prognosis of certain types of tumors.


Assuntos
Adenocarcinoma/patologia , Neoplasias da Mama/patologia , Matriz Extracelular/patologia , Fatores de Crescimento de Fibroblastos/fisiologia , Neoplasias/patologia , Neoplasias da Próstata/patologia , Receptor Tipo 4 de Fator de Crescimento de Fibroblastos/genética , Adenocarcinoma/genética , Adenocarcinoma/fisiopatologia , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/fisiopatologia , Divisão Celular/genética , DNA Complementar/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Inativação Gênica , Homeostase , Humanos , Linfonodos/patologia , Masculino , Camundongos , Camundongos SCID , Invasividade Neoplásica , Reação em Cadeia da Polimerase , Neoplasias da Próstata/genética , Neoplasias da Próstata/fisiopatologia , RNA Interferente Pequeno/genética , Receptor Tipo 4 de Fator de Crescimento de Fibroblastos/fisiologia , Transdução de Sinais
15.
Proc Natl Acad Sci U S A ; 107(36): 15786-91, 2010 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-20798051

RESUMO

Tumor cells use membrane type 1 matrix metalloproteinase (MT1-MMP) for invasion and metastasis. However, the signaling mechanisms that underlie MT1-MMP regulation in cancer have remained unclear. Using a systematic gain-of-function kinome screen for MT1-MMP activity, we have here identified kinases that significantly enhance MT1-MMP activity in tumor cells. In particular, we discovered an MT1-MMP/FGF receptor-4 (FGFR4) membrane complex that either stimulates or suppresses MT1-MMP and FGFR4 activities, depending on a tumor progression-associated polymorphism in FGFR4. The FGFR4-R388 allele, linked to poor cancer prognosis, increased collagen invasion by decreasing lysosomal MT1-MMP degradation. FGFR4-R388 induced MT1-MMP phosphorylation and endosomal stabilization, and surprisingly, the increased MT1-MMP in return enhanced FGFR4-R388 autophosphorylation. A phosphorylation-defective MT1-MMP was stabilized on the cell surface, where it induced simultaneous FGFR4-R388 internalization and dissociation of cell-cell junctions. In contrast, the alternative FGFR4-G388 variant down-regulated MT1-MMP, and the overexpression of MT1-MMP and particularly its phosphorylation-defective mutant vice versa induced FGFR4-G388 degradation. These results provide a mechanistic basis for FGFR4-R388 function in cancer invasion.


Assuntos
Metaloproteinase 14 da Matriz/metabolismo , Polimorfismo Genético , Receptor Tipo 4 de Fator de Crescimento de Fibroblastos/genética , Endossomos/enzimologia , Estabilidade Enzimática , Humanos , Lisossomos/enzimologia , Fosforilação , Transdução de Sinais
16.
J Cell Biochem ; 105(5): 1211-8, 2008 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-18802920

RESUMO

Membrane type 1 matrix metalloproteinase (MT1-MMP, MMP14) is an efficient extracellular matrix (ECM) degrading enzyme that plays important roles in tissue homeostasis and cell invasion. Like a number of type I membrane proteins, MT1-MMP can be internalized from the cell surface through early and recycling endosomes to late endosomes, and recycled to the plasma membrane. Late endosomes participate in the biogenesis of small (30-100 nm) vesicles, exosomes, which redirect plasma membrane proteins for extracellular secretion. We hypothesized that some of the endosomal MT1-MMP could be directed to exosomes for extracellular release. Using cultured human fibrosarcoma (HT-1080) and melanoma (G361) cells we provide evidence that both the full-length 60 kDa and the proteolytically processed 43 kDa forms of MT1-MMP are secreted in exosomes. The isolated exosomes were identified by their vesicular structure in electron microscopy and by exosomal marker proteins CD9 and tumor susceptibility gene (TSG101). Furthermore, exosomes contained beta1-integrin (CD29). The exosomes were able to activate pro-MMP-2 and degrade type 1 collagen and gelatin, suggesting that the exosomal MT1-MMP was functionally active. The targeting of MT1-MMP in exosomes represents a novel mechanism for cancer cells to secrete membrane type metalloproteolytic activity into the extracellular space.


Assuntos
Exossomos/enzimologia , Espaço Extracelular/enzimologia , Metaloproteinase 14 da Matriz/metabolismo , Antígenos CD/metabolismo , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Proteínas de Ligação a DNA/metabolismo , Complexos Endossomais de Distribuição Requeridos para Transporte , Exossomos/metabolismo , Espaço Extracelular/metabolismo , Humanos , Glicoproteínas de Membrana/metabolismo , Microscopia Imunoeletrônica , Tetraspanina 29 , Fatores de Transcrição/metabolismo
17.
Genes Cells ; 12(12): 1315-27, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18076569

RESUMO

Disabled-1 (Dab1) is an adaptor protein mediating Reelin signaling in neuronal migration during brain development. Cyclin-dependent kinase 5 (Cdk5)-p35 is a proline-directed Ser/Thr kinase also involved in neuronal migration. The interaction between Dab1 and Cdk5 is in need of investigation. Dab1 was phosphorylated at Ser400 and Ser491 by Cdk5 in vivo. We search for proteins that interact with Dab1 in a phosphorylation-dependent manner at these sites, and identified CIN85, an SH3-containing adaptor protein involved in endocytosis, and CPalpha/CPbeta, which are subunits of barbed end F-actin-capping proteins (CP), as proteins bound to unphosphorylated Dab1 by mass spectrometric analysis. It was shown that the PTPAPR sequence of Dab1, conforming to the PxxxPR atypical SH3-binding motif, was the binding site for SH3 domains of CIN85. The results that phosphorylation at Ser491 close to the PTPAPR sequence inhibited association with CIN85 may provide a mechanism regulating the interaction between the PxxxPR motif proteins and SH3 domains of CIN85 family proteins. Together with previous results that CIN85 regulates actin assembly, present results raise the possibility that Cdk5 modulates actin dynamics through regulation of CIN85-Dab1 interaction by the Dab1 phosphorylation.


Assuntos
Moléculas de Adesão Celular Neuronais/metabolismo , Quinase 5 Dependente de Ciclina/fisiologia , Proteínas da Matriz Extracelular/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Serina Endopeptidases/metabolismo , Transdução de Sinais , Animais , Células Cultivadas , Quinase 5 Dependente de Ciclina/genética , Regulação da Expressão Gênica , Camundongos , Proteínas do Tecido Nervoso/química , Fosforilação , Proteína Reelina , Serina/metabolismo
18.
Peptides ; 27(12): 3069-76, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17067722

RESUMO

A novel method, based on the hemolytic screening of a cDNA phage library, was developed to isolate cDNAs encoding grammistins (antibacterial peptide toxins) of the soapfish Pogonoperca punctata. As a result, cDNAs encoding six grammistins were isolated and elucidated for their nucleotide sequences. In common with the grammistins, the precursor protein is composed of a highly conserved signal peptide, a considerably conserved propeptide that is characterized to contain a pair of basic residues (Lys-Arg) at plural positions including the C-terminus and one copy of a mature peptide. This precursor organization is similar to those of dermaseptins, antibacterial peptides from the frog skin.


Assuntos
Bass/genética , Clonagem Molecular , DNA Complementar , Venenos de Peixe/genética , Peptídeos/genética , Toxinas Biológicas/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Dados de Sequência Molecular
19.
Toxicon ; 45(5): 595-601, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15777955

RESUMO

Soapfishes contain peptide toxins (grammistins) in the skin secretion. Two grammistins (Gs 1 and Gs 2) and six grammistins (Pp 1, Pp 2a, Pp 2b, Pp 3, Pp 4a and Pp 4b) have already been isolated from Grammistes sexlineatus and Pogonoperca punctata, respectively. In this study, five grammistins (Gs A-E), together with grammistins Gs 1 and Gs 2, were further isolated from G. sexlineatus by gel filtration and reverse-phase HPLC. Sequence analyses revealed that grammistins Gs A (28 residues) and Gs C (26 residues) are analogous to grammistin Pp 3 and grammistin Gs B (12 residues) to grammistin Pp 1, while grammistins Gs D (13 residues) and Gs E (13 residues) are identical with grammistins Pp 1 and Pp 2b, respectively. Grammistins Gs A-C exhibited antibacterial activity with a broad spectrum against nine species of bacteria in common with the other grammistins but had no hemolytic activity differing from the other grammistins. Grammistins Gs A-E, Gs 1 and Gs 2 could release carboxyfluorescein entrapped within liposomes made of either phosphatidylcholine or phosphatidylglycerol/phosphatidylcholine (3:1), demonstrating their membrane-lytic activity. However, no clear relationship between the membrane-lytic activity and the biological activity of grammistins was recognized.


Assuntos
Antibacterianos/toxicidade , Venenos de Peixe/genética , Venenos de Peixe/toxicidade , Perciformes/metabolismo , Pele/metabolismo , Sequência de Aminoácidos , Animais , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Cromatografia em Gel , Cromatografia Líquida de Alta Pressão , Venenos de Peixe/metabolismo , Fluoresceínas/metabolismo , Bactérias Gram-Positivas/efeitos dos fármacos , Hemólise/efeitos dos fármacos , Lipossomos/metabolismo , Dados de Sequência Molecular , Perciformes/genética , Análise de Sequência de Proteína
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...