Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Publication year range
1.
Nat Commun ; 9(1): 2443, 2018 06 22.
Article in English | MEDLINE | ID: mdl-29934494

ABSTRACT

Cancer cells' ability to migrate through constricting pores in the tissue matrix is limited by nuclear stiffness. MT1-MMP contributes to metastasis by widening matrix pores, facilitating confined migration. Here, we show that modulation of matrix pore size or of lamin A expression known to modulate nuclear stiffness directly impinges on levels of MT1-MMP-mediated pericellular collagenolysis by cancer cells. A component of this adaptive response is the centrosome-centered distribution of MT1-MMP intracellular storage compartments ahead of the nucleus. We further show that this response, including invadopodia formation in association with confining matrix fibrils, requires an intact connection between the nucleus and the centrosome via the linker of nucleoskeleton and cytoskeleton (LINC) complex protein nesprin-2 and dynein adaptor Lis1. Our results uncover a digest-on-demand strategy for nuclear translocation through constricted spaces whereby confined migration triggers polarization of MT1-MMP storage compartments and matrix proteolysis in front of the nucleus depending on nucleus-microtubule linkage.


Subject(s)
1-Alkyl-2-acetylglycerophosphocholine Esterase/metabolism , Cell Movement , Matrix Metalloproteinase 14/metabolism , Microfilament Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Neoplasms/pathology , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Centrosome/metabolism , Humans , Lamin Type A/metabolism , Neoplasm Invasiveness/pathology , Podosomes/metabolism , Proteolysis
2.
PLoS One ; 12(3): e0172603, 2017.
Article in English | MEDLINE | ID: mdl-28273108

ABSTRACT

ICAT (Inhibitor of ß-CAtenin and TCF) is a small acidic protein that negatively regulates ß-catenin co-transcriptional activity by competing with TCF/LEF factors in their binding to ß-catenin superhelical core. In melanoma cells, ICAT competes with LEF1 to negatively regulate the M-MITF and NEDD9 target genes. The structure of ICAT consists of two domains: the 3-helix bundle N-terminal domain binds to ß-catenin Armadillo (Arm) repeats 10-12 and the C-terminal tail binds to Arm repeats 5-9. To elucidate the structural mechanisms governing ICAT/ß-catenin interactions in melanoma cells, three ICAT residues Y15, K19 and V22 in the N-terminal domain, contacting hydrophobic ß-catenin residue F660, were mutated and interaction was assessed by immunoprecipitation. Despite the moderate hydrophobicity of the contact, its removal completely abolished the interaction. In the ICAT C-terminal tail consensus sequence, neutralization of the electrostatic interactions between residues D66, E75 and ß-catenin residues K435, K312, coupled to deletion of the hydrophobic contact between F71 and ß-catenin R386, markedly reduced, but failed to abolish the ICAT-mediated negative regulation of M-MITF and NEDD9 promoters. We conclude that in melanoma cells, anchoring of ICAT N-terminal domain to ß-catenin through the hook made by residue F660, trapped in the pincers formed by ICAT residues Y15 and V22, is crucial for stabilizing the ICAT/ß-catenin complex. This is a prerequisite for binding of the consensus peptide to Arm repeats 5-9 and competition with LEF1. Differences between ICAT and LEF1 in their affinity for ß-catenin may rely on the absence in ICAT of hydrophilic residues between D66 and F71.


Subject(s)
Gene Expression Regulation , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Mutation , Transcriptional Activation , beta Catenin/metabolism , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , Binding, Competitive , Cell Line, Tumor , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Lymphoid Enhancer-Binding Factor 1/metabolism , Melanocytes/metabolism , Microphthalmia-Associated Transcription Factor/genetics , Microphthalmia-Associated Transcription Factor/metabolism , Models, Molecular , Protein Binding , Protein Conformation , Protein Interaction Domains and Motifs
3.
J Cell Biochem ; 118(8): 1984-1993, 2017 08.
Article in English | MEDLINE | ID: mdl-28112429

ABSTRACT

Maintenance of hematopoietic stem cells (HSC) takes place in a highly specialized microenvironment within the bone marrow. Technological improvements, especially in the field of in vivo imaging, have helped unravel the complexity of the niche microenvironment and have completely changed the classical concept from what was previously believed to be a static supportive platform, to a dynamic microenvironment tightly regulating HSC homeostasis through the complex interplay between diverse cell types, secreted factors, extracellular matrix molecules, and the expression of different transmembrane receptors. To add to the complexity, non-protein based metabolites have also been recognized as a component of the bone marrow niche. The objective of this review is to discuss the current understanding on how the different extracellular matrix components of the niche regulate HSC fate, both during embryonic development and in adulthood. Special attention will be provided to the description of non-protein metabolites, such as lipids and metal ions, which contribute to the regulation of HSC behavior. J. Cell. Biochem. 118: 1984-1993, 2017. © 2017 Wiley Periodicals, Inc.


Subject(s)
Bone Marrow Cells/metabolism , Cellular Microenvironment/genetics , Extracellular Matrix Proteins/genetics , Extracellular Matrix/chemistry , Hematopoietic Stem Cells/metabolism , Stem Cell Niche/genetics , Animals , Bone Marrow Cells/cytology , Chemokine CXCL12/genetics , Chemokine CXCL12/metabolism , Dinoprostone/metabolism , Embryo, Mammalian , Embryonic Development/genetics , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Extracellular Matrix Proteins/metabolism , Gene Expression Regulation , Hematopoietic Stem Cells/cytology , Homeostasis , Humans , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Signal Transduction
4.
Int J Hematol ; 105(2): 141-152, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27905003

ABSTRACT

Mobilized peripheral blood (PB) is the most common source of hematopoietic stem cells (HSC) for autologous transplantation. Granulocyte colony stimulating factor (G-CSF) is the most commonly used mobilization agent, yet despite its widespread use, a considerable number of patients still fail to mobilize. Recently, a greater understanding of the interactions that regulate HSC homeostasis in the bone marrow (BM) microenvironment has enabled the development of new molecules that mobilize HSC through specific inhibition, modulation or perturbation of these interactions. AMD3100 (plerixafor), a small molecule that selectively inhibits the chemokine receptor CXCR4 is approved for mobilization in combination with G-CSF in patients with Non-Hodgkin's lymphoma and multiple myeloma. Nevertheless, identifying mobilization strategies that not only enhance HSC number, but are rapid and generate an optimal "mobilized product" for improved transplant outcomes remains an area of clinical importance. In recent times, new agents based on recombinant proteins, peptides and small molecules have been identified as potential candidates for therapeutic HSC mobilization. In this review, we describe the most recent developments in HSC mobilization agents and their potential impact in HSC transplantation.


Subject(s)
Hematopoietic Stem Cell Mobilization/methods , Animals , Benzylamines , Bone Marrow/chemistry , Cyclams , Granulocyte Colony-Stimulating Factor/pharmacology , Heterocyclic Compounds/pharmacology , Humans , Receptors, CXCR4/antagonists & inhibitors
5.
Cancer Res ; 74(7): 1983-95, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24514042

ABSTRACT

Inhibitor of ß-catenin and TCF (ICAT) inhibits ß-catenin transcriptional activity by competing with T-cell factor/lymphoid enhancer factor. We documented high ICAT levels in human melanoma cells, in which ß-catenin signaling is frequently deregulated, finding a correlation with the capacity to form metastases in nude mice. Ectopic expression of ICAT in melanoma cells did not affect their proliferation but increased cell motility and Matrigel invasion of metastatic cells in a manner relying upon stable ICAT-ß-catenin interaction. This effect was associated with conversion of an elongated/mesenchymal phenotype to a round/amoeboid phenotype in the absence of similar effects on elongated morphology of nonmetastatic melanoma cells. Transition from mesenchymal to amoeboid movement was associated with decreased levels of NEDD9 and activated Rac1, a positive regulator of mesenchymal movement. Ectopic ICAT promoted colonization of melanoma cells in the lungs of nude mice, suggesting an increase in metastatic potential. Together, our results showed that by downregulating Rac signaling in metastatic melanoma cells, ICAT increased their invasive motility by promoting a morphologic variation that facilitates a favorable adaptation to their microenvironment.


Subject(s)
Intracellular Signaling Peptides and Proteins/physiology , Melanoma/pathology , Adaptor Proteins, Signal Transducing/physiology , Animals , Cell Line, Tumor , Cell Movement , Humans , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Melanoma/mortality , Melanoma/secondary , Mice , Mice, Nude , Neoplasm Invasiveness , Phosphoproteins/physiology , beta Catenin/physiology
6.
Med Sci (Paris) ; 29(3): 287-92, 2013 Mar.
Article in French | MEDLINE | ID: mdl-23544383

ABSTRACT

During development, neural crest cells-derived melanoblasts migrate along the dorso-lateral axis into the dermis, then cross the basal layer to reach the epidermis and differentiate into melanocytes. They finally colonize the hair follicles to become resident pigmented cells. Neoplastic transformation converts melanocytes into highly invasive melanoma cells, which can adopt two modes of interconvertible migration (mesenchymal and amoeboid). Through analysis of the coat color phenotype of natural mouse mutants and genetically modified animals, many of the genes regulating migration were identified. Deciphering of cell membrane protrusions and signaling molecules involved in melanoma cell motility was further achieved through 2D and 3D culture systems. Here, we summarize how these data allow a better understanding of the complex mechanisms controlling migration of normal and pathological cells of the melanocytic lineage.


Subject(s)
Cell Movement , Embryonic Development , Melanocytes/physiology , Animals , Cell Movement/genetics , Cells, Cultured , Humans , Melanoma/pathology , Neural Crest/cytology
7.
Pigment Cell Melanoma Res ; 26(3): 316-25, 2013 May.
Article in English | MEDLINE | ID: mdl-23433358

ABSTRACT

During embryonic development in vertebrates, the neural crest-derived melanoblasts migrate along the dorsolateral axis and cross the basal membrane separating the dermis from the epidermis to reach their final location in the interfollicular epidermis and epidermal hair follicles. Neoplastic transformation converts melanocytes into highly invasive and metastatic melanoma cells. In vitro, these cells extend various types of protrusions and adopt two interconvertible modes of migration, mesenchymal and amoeboid, driven by different signalling molecules. In this review, we describe the major contributions of natural mouse mutants, mouse models generated by genetic engineering and in vitro culture systems, to identification of the genes, signalling pathways and mechanisms regulating the migration of normal and pathological cells of the melanocyte lineage, at both the cellular and molecular levels.


Subject(s)
Cell Movement , Melanocytes/pathology , Melanoma/pathology , Animals , Embryonic Development , Humans , Models, Biological , Neoplasm Invasiveness
SELECTION OF CITATIONS
SEARCH DETAIL
...