Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Exp Dermatol ; 21(7): 551-4, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22716255

ABSTRACT

Depth of invasion, a quantifier of vertical growth, is a major cutaneous melanoma staging factor. Stromal penetrance requires pericellular proteolysis regulated by the serine protease and matrix metalloproteinase cascades. The serine protease inhibitor SERPINE1, a poor prognosis biomarker in various cancers, promotes tumor progression likely by titrating the extent and local of plasmin-initiated matrix remodelling. SERPINE1 in human melanoma was assessed using tissue arrays that included primary/metastatic tumors and normal skin. SERPINE1 was basal layer-restricted in the normal epidermis. SERPINE1 immunoreactivity was evident in 27/28 primary (96%) and 24/26 metastatic tumors (92%); cutaneous metastases (80%) had significantly elevated SERPINE1 levels compared with low signals characteristic of lymph node lesions. Moderate SERPINE1 expression was a general finding in primary melanoma, whereas reduced or increased SERPINE1 immunolocalization typified metastatic deposits. The amplitude of SERPINE1 expression may impact melanoma site-specific dissemination, with cutaneous metastases representing a high-SERPINE1 tumor subtype.


Subject(s)
Biomarkers, Tumor/metabolism , Melanoma/metabolism , Melanoma/secondary , Plasminogen Activator Inhibitor 1/metabolism , Skin Neoplasms/metabolism , Epidermis/metabolism , Humans , Immunohistochemistry , Lymphatic Metastasis , Skin Neoplasms/pathology , Skin Neoplasms/secondary
2.
Mol Cancer ; 10: 114, 2011 Sep 14.
Article in English | MEDLINE | ID: mdl-21917148

ABSTRACT

BACKGROUND: The initial use of BRAF targeted therapeutics in clinical trials has demonstrated encouraging responses in melanoma patients, although a rise in drug-resistant cells capable of advancing malignant disease has been described. The current study uses BRAFV600E expressing WM793 melanoma cells to derive data aimed at investigating the molecular determinant of cell invasion following treatment with clinical BRAF inhibitors. FINDINGS: Small-molecule inhibitors targeting BRAF reduced MEK1/2-ERK1/2 pathway activation and cell survival; yet, viable cell subpopulations persisted. The residual cells exhibited an elongated cell shape, prominent actin stress fibers and retained the ability to invade 3-D dermal-like microenvironments. BRAF inhibitor treatments were associated with reduced expression of RND3, an antagonist of RHOA activation, and elevated RHOA-dependent signaling. Restoration of RND3 expression or RHOA knockdown attenuated the migratory ability of residual cells without affecting overall cell survival. The invasive ability of BRAF inhibitor treated cells embedded in collagen gels was diminished following RND3 re-expression or RHOA depletion. Conversely, melanoma cell movement in the absence of BRAF inhibition was unaffected by RND3 expression or RHOA depletion. CONCLUSION: These data reveal a novel switch in the requirement for RND3 and RHOA in coordinating the movement of residual WM793 cells that are initially refractive to BRAF inhibitor therapy. These results have important clinical implications because they suggest that combining BRAF inhibitors with therapies that target the invasion of drug-resistant cells could aid in controlling disease relapse.


Subject(s)
Melanoma/metabolism , Mutant Proteins/metabolism , Proto-Oncogene Proteins B-raf/metabolism , Signal Transduction , rho GTP-Binding Proteins/metabolism , rhoA GTP-Binding Protein/metabolism , Amino Acid Substitution , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Drug Resistance, Neoplasm , Gene Knockdown Techniques , Humans , Imidazoles/pharmacology , Indoles/pharmacology , Melanoma/pathology , Mutant Proteins/antagonists & inhibitors , Mutant Proteins/genetics , Neoplasm Invasiveness , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/genetics , RNA Interference , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Spheroids, Cellular/drug effects , Spheroids, Cellular/pathology , Sulfonamides/pharmacology , rho GTP-Binding Proteins/genetics , rhoA GTP-Binding Protein/genetics
3.
Int J Cell Biol ; 2011: 562481, 2011.
Article in English | MEDLINE | ID: mdl-21837240

ABSTRACT

Cellular migration, over simple surfaces or through complex stromal barriers, requires coordination between detachment/re-adhesion cycles, involving structural components of the extracellular matrix and their surface-binding elements (integrins), and the precise regulation of the pericellular proteolytic microenvironment. It is now apparent that several proteases and protease inhibitors, most notably urokinase plasminogen activator (uPA) and plasminogen activator inhibitor type-1 (PAI-1), also interact with several cell surface receptors transducing intracellular signals that significantly affect both motile and proliferative programs. These events appear distinct from the original function of uPA/PAI-1 as modulators of the plasmin-based proteolytic cascade. The multifaceted interactions of PAI-1 with specific matrix components (i.e., vitronectin), the low-density lipoprotein receptor-related protein-1 (LRP1), and the uPA/uPA receptor complex have dramatic consequences on the migratory phenotype and may underlie the pathophysiologic sequalae of PAI-1 deficiency and overexpression. This paper focuses on the increasingly intricate role of PAI-1 as a major mechanistic determinant of the cellular migratory phenotype.

4.
Matrix Biol ; 28(2): 101-9, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19379667

ABSTRACT

Anastellin is an angiogenesis inhibitor derived from the first type III repeat of fibronectin (FN). Anastellin binds to fibronectin and promotes the polymerization of soluble fibronectin into a highly polymerized form termed superfibronectin. In addition, anastellin also causes remodeling of pre-existing fibronectin matrix and modulates cell signaling pathways in both endothelial cells and fibroblasts. In the present study, we address the relationship of anastellin's effects on fibronectin matrix to its effects on p38 MAP kinase (MAPK) activation. Using a mutant form of anastellin which binds to fibronectin matrix, but does not stimulate formation of superfibronectin, we demonstrate that the activation of p38 MAPK by anastellin is not dependent on the formation of superfibronectin. The mutant form of anastellin does stimulate matrix remodeling, but experiments using FN(-/-) cells show that the effect of anastellin on p38-MAPK activation is completely independent of fibronectin. Anastellin was able to activate p38 MAPK on cells in suspension as well as on cells null for beta1 integrins, suggesting that anastellin activity did not require ligation of integrins. These data suggest that the activation of p38 MAPK by anastellin is independent of anastellin's effects on fibronectin matrix organization.


Subject(s)
Extracellular Matrix/metabolism , Fibronectins/metabolism , Peptide Fragments/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Analysis of Variance , Enzyme Activation/physiology , Enzyme-Linked Immunosorbent Assay , Fibroblasts , Fibronectins/genetics , Humans , Immunoblotting , Microscopy, Fluorescence
5.
Cancer Res ; 69(6): 2224-33, 2009 Mar 15.
Article in English | MEDLINE | ID: mdl-19244113

ABSTRACT

The depth of cell invasion into the dermis is a clinical determinant for poor prognosis in cutaneous melanoma. The signaling events that promote the switch from a noninvasive to invasive tumor phenotype remain obscure. Activating mutations in the serine/threonine kinase B-RAF are prevalent in melanoma. Mutant B-RAF is required for melanoma cell invasion. The expression of Rnd3, a Rho family GTPase, is regulated by mutant B-RAF, although its role in melanoma progression is unknown. In this study, we determined the functional contribution of Rnd3 to invasive melanoma. Endogenous Rnd3 was targeted for knockdown using a doxycycline-inducible short hairpin RNA system in invasive human melanoma cells. Depletion of Rnd3 promoted prominent actin stress fibers and enlarged focal adhesions. Mechanistically, stress fiber formation induced by Rnd3 knockdown required the specific involvement of RhoA and ROCK1/2 activity but not RhoB or RhoC. Rnd3 expression in human melanoma cell lines was strongly associated with elevated extracellular signal-regulated kinase phosphorylation and invasive behavior in a three-dimensional dermal-like environment. A functional role for Rnd3 was shown in the invasive outgrowth of melanoma tumor spheroids. Knockdown of Rnd3 reduced the invasive outgrowth of spheroids embedded in collagen gels. Additionally, Rnd3 depletion inhibited collective and border cell movement out from spheroids in a ROCK1/2-dependent manner. Collectively, these findings implicate Rnd3 as a major suppressor of RhoA-mediated actin cytoskeletal organization and in the acquisition of an invasive melanoma phenotype.


Subject(s)
Actins/metabolism , Cell Movement/physiology , Melanoma/pathology , rho GTP-Binding Proteins/metabolism , Cell Cycle/physiology , Cell Growth Processes/physiology , Cytoskeleton/enzymology , Cytoskeleton/metabolism , Cytoskeleton/pathology , Humans , Melanoma/enzymology , Melanoma/genetics , Melanoma/metabolism , Neoplasm Invasiveness , Proto-Oncogene Proteins B-raf/metabolism , RNA, Small Interfering/genetics , Spheroids, Cellular , Transfection , Tumor Cells, Cultured , rho GTP-Binding Proteins/biosynthesis , rho GTP-Binding Proteins/deficiency , rho GTP-Binding Proteins/genetics , rhoA GTP-Binding Protein/metabolism
6.
Mol Biol Cell ; 19(2): 498-508, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18045987

ABSTRACT

The actin cytoskeleton controls multiple cellular functions, including cell morphology, movement, and growth. Accumulating evidence indicates that oncogenic activation of the mitogen-activated protein kinase kinase/extracellular signal-regulated kinase 1/2 (MEK/ERK1/2) pathway is accompanied by actin cytoskeletal reorganization. However, the signaling events contributing to actin cytoskeleton remodeling mediated by aberrant ERK1/2 activation are largely unknown. Mutant B-RAF is found in a variety of cancers, including melanoma, and it enhances activation of the MEK/ERK1/2 pathway. We show that targeted knockdown of B-RAF with small interfering RNA or pharmacological inhibition of MEK increased actin stress fiber formation and stabilized focal adhesion dynamics in human melanoma cells. These effects were due to stimulation of the Rho/Rho kinase (ROCK)/LIM kinase-2 signaling pathway, cumulating in the inactivation of the actin depolymerizing/severing protein cofilin. The expression of Rnd3, a Rho antagonist, was attenuated after B-RAF knockdown or MEK inhibition, but it was enhanced in melanocytes expressing active B-RAF. Constitutive expression of Rnd3 suppressed the actin cytoskeletal and focal adhesion effects mediated by B-RAF knockdown. Depletion of Rnd3 elevated cofilin phosphorylation and stress fiber formation and reduced cell invasion. Together, our results identify Rnd3 as a regulator of cross talk between the RAF/MEK/ERK and Rho/ROCK signaling pathways, and a key contributor to oncogene-mediated reorganization of the actin cytoskeleton and focal adhesions.


Subject(s)
Actins/metabolism , Cytoskeleton/enzymology , Focal Adhesions/enzymology , Proto-Oncogene Proteins B-raf/metabolism , rho GTP-Binding Proteins/metabolism , Actin Depolymerizing Factors/metabolism , Cell Line, Tumor , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Lim Kinases/metabolism , MAP Kinase Signaling System , Melanoma/metabolism , Melanoma/pathology , Mitogen-Activated Protein Kinase Kinases/metabolism , Mutant Proteins/metabolism , Neoplasm Invasiveness , Phosphorylation , Stress Fibers/enzymology , rho-Associated Kinases/metabolism
7.
Cancer Res ; 65(1): 148-56, 2005 Jan 01.
Article in English | MEDLINE | ID: mdl-15665290

ABSTRACT

The formation of a microvascular endothelium plays a critical role in the growth and metastasis of established tumors. The ability of a fragment from the first type III repeat of fibronectin (III(1C)), anastellin, to suppress tumor growth and metastasis in vivo has been reported to be related to its antiangiogenic properties, however, the mechanism of action of anastellin remains unknown. Utilizing cultures of human dermal microvascular endothelial cells, we provide evidence that anastellin inhibits signaling pathways which regulate the extracellular signal-regulated (ERK) mitogen-activated protein kinase pathway and subsequent expression of cell cycle regulatory proteins. Addition of anastellin to primary microvascular endothelial cells resulted in a complete inhibition of serum-dependent proliferation. Growth inhibition correlated with a decrease in serum-dependent expression of cyclin D1, cyclin A and the cyclin-dependent kinase, cdk4, key regulators of cell cycle progression through G(1) phase. Consistent with a block in G(1)-S transition, anastellin inhibited serum-dependent incorporation of [(3)H]-thymidine into S-phase nuclei. Addition of anastellin to serum-starved microvessel cells resulted in a time-dependent and dose-dependent decrease in basal levels of phosphorylated MEK/ERK and blocked serum-dependent activation of ERK. Adenoviral infection with Ad.DeltaB-Raf:ER, an inducible estrogen receptor-B-Raf fusion protein, restored levels of active ERK in anastellin-treated cells, rescued levels of cyclin D1, cyclin A, and cdk4, and rescued [(3)H]-thymidine incorporation. These data suggest that the antiangiogenic properties of anastellin observed in mouse models of human cancer may be due to its ability to block endothelial cell proliferation by modulating ERK signaling pathways and down-regulating cell cycle regulatory gene expression required for G(1)-S phase progression.


Subject(s)
Endothelium, Vascular/physiology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Fibronectins/pharmacology , Peptide Fragments/pharmacology , Adult , Apoptosis/drug effects , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , G1 Phase/drug effects , Humans , Kinetics , Microcirculation/physiology , Recombinant Proteins/pharmacology , Skin/blood supply
8.
J Cell Sci ; 116(Pt 22): 4663-74, 2003 Nov 15.
Article in English | MEDLINE | ID: mdl-14576359

ABSTRACT

The fibronectin matrix contains cryptic sites which are thought to modulate cellular biological responses. One of these sites, located in fibronectin's first type III repeat (III1c), influences signaling pathways that are relevant to cytoskeletal organization and cell cycle progression. The purpose of this study was to identify possible mechanisms responsible for the effects of III1c on cell behavior. Recombinant peptides representing various type III repeats of fibronectin were compared for their effects on fibronectin matrix organization and activation of intracellular signaling pathways. III1c and III13 but not III11c or III10 bound to monolayers of human skin fibroblasts in a dose- and time-dependent manner and were localized to the extracellular matrix. Binding of III13, but not III1c, to matrix was sensitive to heparitinase, suggesting that the association of III1c with the matrix was not dependent on heparan sulfate proteoglycans. Quantitative and morphological assessment indicated that, in contrast to previously published reports, the binding of III1c to cell layers did not result in the loss or disruption of matrix fibronectin. Binding of III1c but not III13 to the extracellular matrix did result in the loss of a conformationally sensitive epitope present within the EDA type III module of cellular fibronectin. III1c-induced loss of the EDA epitope did not require the presence of cells, occurred within 1 hour and was associated with the activation of p38 mitogen-activated protein kinase (MAPK) followed by the formation of filopodia. Maximal phosphorylation of p38 MAPK occurred within 1 hour, whereas cytoskeletal changes did not appear until 12 hours later. These findings are consistent with a model in which the binding of III1c to the extracellular matrix results in a conformational remodeling of the fibronectin matrix, which has both short- and long-term effects on cell physiology.


Subject(s)
Cell Adhesion/physiology , Cytoskeleton/metabolism , Extracellular Matrix/physiology , Fibroblasts/metabolism , Fibronectins/metabolism , Cell Cycle/physiology , Cells, Cultured , Epidermis/metabolism , Fibroblasts/cytology , Heparan Sulfate Proteoglycans/metabolism , Humans , Microscopy, Fluorescence , Mitogen-Activated Protein Kinases/metabolism , Peptides/metabolism , Phosphorylation , Polysaccharide-Lyases/metabolism , Protein Structure, Tertiary , Pseudopodia/metabolism , Recombinant Fusion Proteins/metabolism , Signal Transduction , p38 Mitogen-Activated Protein Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...