Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
Biochim Biophys Acta ; 1863(9): 2221-33, 2016 09.
Article in English | MEDLINE | ID: mdl-27188792

ABSTRACT

Class III ß-tubulin (TUBB3) is a marker of drug resistance expressed in a variety of solid tumors. Originally, it was described as an important element of chemoresistance to taxanes. Recent studies have revealed that TUBB3 is also involved in an adaptive response to a microenvironmental stressor, e.g. low oxygen levels and poor nutrient supply in some solid tumors, independently of the microtubule targeting agent. Furthermore, it has been demonstrated that TUBB3 is a marker of biological aggressiveness associated with modulation of metastatic abilities in colon cancer. The epithelial-to-mesenchymal transition (EMT) is a basic cellular process by which epithelial cells lose their epithelial behavior and become invasive cells involved in cancer metastasis. Snail is a zinc-finger transcription factor which is able to induce EMT through the repression of E-cadherin expression. In the presented studies we focused on the analysis of the TUBB3 role in EMT-induced colon adenocarcinoma cell lines HT-29 and LS180. We observed a positive correlation between Snail presence and TUBB3 upregulation in tested adenocarcinoma cell lines. The cellular and behavioral analysis revealed for the first time that elevated TUBB3 level is functionally linked to increased cell migration and invasive capability of EMT induced cells. Additionally, the post-transcriptional modifications (phosphorylation, glycosylation) appear to regulate the cellular localization of TUBB3 and its phosphorylation, observed in cytoskeleton, is probably involved in cell motility modulation.


Subject(s)
Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Epithelial-Mesenchymal Transition , Snail Family Transcription Factors/metabolism , Tubulin/metabolism , Adenocarcinoma/pathology , Cell Compartmentation/drug effects , Cell Movement/drug effects , Cytoskeleton/drug effects , Cytoskeleton/metabolism , HT29 Cells , Humans , Microtubules/drug effects , Microtubules/metabolism , Neoplasm Invasiveness , Phosphorylation/drug effects , RNA, Small Interfering/metabolism , Transforming Growth Factor beta1/pharmacology , Up-Regulation/drug effects
2.
Exp Cell Res ; 324(2): 124-36, 2014 Jun 10.
Article in English | MEDLINE | ID: mdl-24657343

ABSTRACT

The mechanisms controlling the switch between the pro-angiogenic and pro-inflammatory states of endothelial cells are still poorly understood. In this paper, we show that: (a) COX-2 expression induced by VEGF-A is NFAT2-dependent; and (b) the integrin profile in endothelial cells induced by the pro-angiogenic VEGF-A is distinct from that brought on by the inflammatory cytokine TNF-α. Two groups of integrin subunits specifically upregulated over time by both cytokines were identified using RT-PCR and Western Immunoblotting. The first group included α4, α5, α6, and ß5 subunits that were upregulated by VEGF-A; the second group consisted of αV and ß3 induced by TNF-α. Both cytokines significantly enhanced the expression of ß1 and modulated α2 mRNA. In contrast to TNF-α, VEGF-A induction of integrin subunits depended on the activation of the calcineurin/NFAT pathway. Both calcineurin inhibitors (cyclosporineA and 11R-VIVIT) and downregulation of NFAT2 with specific siRNA decreased induction of integrin subunits. This process of induction could be increased by upregulation of NFAT2 by pBJ5-NFAT2 transfection. This suggests that NFAT2 mediates VEGF-induced upregulation of integrin subunit synthesis by providing a constant supply of newly synthesized "refreshed" mature integrin receptors, particularly α2ß1, α5ß1, α4ß1, α6ß1 and αVß5, which are involved at different stages of angiogenesis.


Subject(s)
Cyclooxygenase 2/genetics , Human Umbilical Vein Endothelial Cells/metabolism , Inflammation , Integrins/metabolism , NFATC Transcription Factors/physiology , Neovascularization, Physiologic , Cells, Cultured , Cyclooxygenase 2/metabolism , Gene Expression Regulation, Enzymologic , Humans , Inflammation/genetics , Inflammation/metabolism , Neovascularization, Physiologic/genetics , Transcriptome/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Vascular Endothelial Growth Factor A/pharmacology
3.
J Biol Chem ; 289(9): 5758-73, 2014 Feb 28.
Article in English | MEDLINE | ID: mdl-24415753

ABSTRACT

Recent studies support the role of cysteine oxidation in actin cytoskeleton reorganization during cell adhesion. The aim of this study was to explain whether protein disulfide isomerase (PDI) is responsible for the thiol-disulfide rearrangement in the ß-actin molecule of adhering cells. First, we showed that PDI forms a disulfide-bonded complex with ß-actin with a molecular mass of 110 kDa. Specific interaction of both proteins was demonstrated by a solid phase binding assay, surface plasmon resonance analysis, and immunoprecipitation experiments. Second, using confocal microscopy, we found that both proteins colocalized when spreading MEG-01 cells on fibronectin. Colocalization of PDI and ß-actin could be abolished by the membrane-permeable sulfhydryl blocker, N-ethylmaleimide, by the RGD peptide, and by anti-αIIbß3 antibodies. Consequently, down-regulation of PDI expression by antisense oligonucleotides impaired the spreading of cells and initiated reorganization of the cytoskeleton. Third, because of transfection experiments followed by immunoprecipitation and confocal analysis, we provided evidence that PDI binds to the ß-actin Cys(374) thiol. Formation of the ß-actin-PDI complex was mediated by integrin-dependent signaling in response to the adhesion of cells to the extracellular matrix. Our data suggest that PDI is released from subcellular compartments to the cytosol and translocated toward the periphery of the cell, where it forms a disulfide bond with ß-actin when MEG-01 cells adhere via the αIIbß3 integrin to fibronectin. Thus, PDI appears to regulate cytoskeletal reorganization by the thiol-disulfide exchange in ß-actin via a redox-dependent mechanism.


Subject(s)
Actins/metabolism , Cytoskeleton/metabolism , Protein Disulfide-Isomerases/metabolism , Signal Transduction/physiology , Actins/genetics , Antineoplastic Agents/pharmacology , Cell Adhesion/drug effects , Cell Adhesion/genetics , Cell Line , Cytoskeleton/genetics , Enzyme Inhibitors/pharmacology , Ethylmaleimide/pharmacology , Fibronectins/genetics , Fibronectins/metabolism , Humans , Oligopeptides/pharmacology , Oxidation-Reduction/drug effects , Platelet Glycoprotein GPIIb-IIIa Complex/genetics , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Protein Disulfide-Isomerases/genetics , Signal Transduction/drug effects
4.
Exp Cell Res ; 319(8): 1213-9, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23474086

ABSTRACT

Due to the lack of an N-terminal signal peptide, SerpinB2 (plasminogen activator inhibitor type 2) accumulates in cells and only a small percentage of it is secreted. The extracellular concentration of SerpinB2 significantly increases during inflammation. In the present study we investigated the mechanism with which SerpinB2 can be secreted from endothelial cells activated with LPS. We evaluated the intracellular distribution of SerpinB2 by double immunogold labeling followed by a high resolution electron microscopy analysis. We found that SerpinB2 gathers in the vesicular structures and in the endothelial cell periphery. These vesicles stained positive for the trans-Golgi network marker TGN46, which is consistent with their formation by the endoplasmatic reticulum (ER) and Golgi-dependent pathways. SerpinB2 was delivered to the plasma membrane, apparently together with TGN46 in the same vesicles, which after fusion with the membranes released cargo. Secretion of SerpinB2 was partially inhibited by brefeldin A. The secreted SerpinB2 was predominantly in its nonglycosylated 43kDa form as evaluated by Western immunoblotting. Our data suggest that increased expression of SerpinB2 by an inflammatory stimulus is sufficient to generate structures that resemble secretory vesicles. These vesicles may represent the mechanism by which high local concentrations of SerpinB2 are released at inflammation sites from endothelial cells.


Subject(s)
Endothelial Cells/drug effects , Endothelial Cells/metabolism , Inflammation/metabolism , Lipopolysaccharides/pharmacology , Plasminogen Activator Inhibitor 2/metabolism , Cells, Cultured , Endothelial Cells/pathology , Golgi Apparatus/metabolism , Humans , Inflammation/pathology , Inflammation Mediators/pharmacology , Membrane Glycoproteins/metabolism , Microscopy, Electron , Secretory Vesicles/metabolism , Stimulation, Chemical , trans-Golgi Network/metabolism
5.
Ann N Y Acad Sci ; 1269: 44-52, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23045969

ABSTRACT

Thymosin ß4 (Tß4) is a multifunctional protein that has pleiotropic activities both intracellularly and extracellularly. The mechanisms by which it influences cellular processes such as adhesion, migration, differentiation, or apoptosis are not yet understood. Calcium is a ubiquitous signal molecule that is involved in the regulation of almost all cellular functions. Our data indicate that the release of Ca(2+) from intracellular stores following stimulation of cells with Tß4 does not occur. Interestingly, Tß4 becomes rapidly internalized, supporting the concept that it may express its activities via intracellular receptors.


Subject(s)
Calcium/metabolism , Thymosin/metabolism , Animals , Apoptosis/physiology , Biological Transport/physiology , Cell Differentiation/physiology , Humans
6.
J Biol Chem ; 287(43): 36556-66, 2012 Oct 19.
Article in English | MEDLINE | ID: mdl-22898815

ABSTRACT

Cancer cell invasion is a key element in metastasis that requires integrins for adhesion/de-adhesion, as well as matrix metalloproteinases (MMPs) for focalized proteolysis. Herein we show that MMP-2 is up-regulated in resected colorectal tumors and degrades ß1 integrins with the release of fragments containing the ß1 I-domain. The ß1 cleavage pattern is similar to that produced by digestion of α5ß1 and α2ß1 with MMP-2. Two such fragments, at 25 and 75 kDa, were identified after immunoprecipitation, with monoclonal antibody BD610468 reacting with the NH(2)-terminal I-like ectodomain followed by SDS-PAGE and microsequencing using electrospray (ISI-Q-TOF-Micromass) spectrometry. Cleavage of the ß1 integrin can be abolished by inhibition of MMP-2 activity; it can be induced by up-regulation of MMP-2 expression, as exemplified by HT29 colon cancer cells transfected with pCMV6-XL5-MMP-2. Co-immunoprecipitation studies of colon cancer cells showed that the ß1 integrin subunit is associated with MMP-2. The MMP-2-mediated shedding of the I-like domain from ß1 integrins resulted in decreased adhesion of colon cancer cells to collagen and fibronectin, thus abolishing their receptivity. Furthermore, such cells showed enhanced motility as evaluated by a "wound healing-like" assay and time-lapse microscopy, indicating their increased invasiveness. Altogether, our data demonstrate that MMP-2 amplifies the motility of colon cancer cells, not only by digesting the extracellular matrix components in the vicinity of cancer cells but also by inactivating their major ß1 integrin receptors.


Subject(s)
Cell Movement , Colonic Neoplasms/metabolism , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Integrin beta1/metabolism , Matrix Metalloproteinase 2/metabolism , Neoplasm Proteins/metabolism , Proteolysis , Cell Adhesion/genetics , Cell Line, Tumor , Collagen/genetics , Collagen/metabolism , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Fibronectins/genetics , Fibronectins/metabolism , Humans , Integrin alpha2beta1/genetics , Integrin alpha2beta1/metabolism , Integrin alpha5beta1/genetics , Integrin alpha5beta1/metabolism , Integrin beta1/genetics , Matrix Metalloproteinase 2/genetics , Neoplasm Proteins/genetics , Protein Structure, Tertiary , Up-Regulation/genetics
7.
Arch Med Sci ; 8(2): 347-56, 2012 May 09.
Article in English | MEDLINE | ID: mdl-22662010

ABSTRACT

INTRODUCTION: There are scarce data on immunochemical properties of pemphigus antibodies detected in clinical remission in pemphigus vulgaris (PV) patients. The aim of the study was to compare biological activity of anti-Dsg3 autoantibodies purified from the sera of PV patients in active stage and in clinical remission. MATERIAL AND METHODS: The effect of purified antibodies on expression of procaspase-3, Bax, Bcl-2, uPAR, IL-1ß, IL-6, and TNF-α mRNAs in the HaCaT keratinocytes was evaluated by Western blot and RT-PCR method. RESULTS: Incubation of HaCaT cells with anti-Dsg-3 autoantibodies caused their binding to cell membranes surfaces. Anti-Dsg3 autoantibodies isolated from the patients in active stage and clinical remission showed proapoptotic effect, caused enhanced expression of analyzed proinflammatory cytokines' mRNAs and uPAR mRNA. CONCLUSIONS: Our data revealed similar pathogenic activity of anti Dsg-3 autoantibodies isolated from active and clinical remission PV patients.

8.
Exp Cell Res ; 318(14): 1659-66, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22652458

ABSTRACT

Numerous studies have demonstrated the effects of Tß4 on cell migration, proliferation, apoptosis and inflammation after exogenous treatment, but the mechanism by which Tß4 functions is still unclear. Previously, we demonstrated that incubation of endothelial cells with Tß4 induced synthesis and secretion of various proteins, including plasminogen activator inhibitor type 1 and matrix metaloproteinases. We also showed that Tß4 interacts with Ku80, which may operate as a novel receptor for Tß4 and mediates its intracellular activity. In this paper, we provide evidence that Tß4 induces cellular processes without changes in the intracellular Ca(2+) concentration. External treatment of HUVECs with Tß4 and its mutants deprived of the N-terminal tetrapeptide AcSDKP (Tß4(AcSDKPT/4A)) or the actin-binding sequence KLKKTET (Tß4(KLKKTET/7A)) resulted in enhanced cell migration and formation of tubular structures in Matrigel. Surprisingly, the increased cell motility caused by Tß4 was not associated with the intracellular Ca(2+) elevation monitored with Fluo-4 NW or Fura-2 AM. Therefore, it is unlikely that externally added Tß4 induces HUVEC migration via the surface membrane receptors known to generate Ca(2+) influx. Our data confirm the concept that externally added Tß4 must be internalized to induce intracellular mechanisms supporting endothelial cell migration.


Subject(s)
Calcium/metabolism , Endothelial Cells/metabolism , Thymosin/metabolism , Cell Movement , Cells, Cultured , Endothelial Cells/cytology , Humans
9.
J Biol Chem ; 286(50): 43164-71, 2011 Dec 16.
Article in English | MEDLINE | ID: mdl-21976669

ABSTRACT

Quiescent endothelial cells contain low concentrations of plasminogen activator inhibitor type 2 (PAI-2). However, its synthesis can be rapidly stimulated by a variety of inflammatory mediators. In this study, we provide evidence that PAI-2 interacts with proteasome and affects its activity in endothelial cells. To ensure that the PAI-2·proteasome complex is formed in vivo, both proteins were coimmunoprecipitated from endothelial cells and identified with specific antibodies. The specificity of this interaction was evidenced after (a) transfection of HeLa cells with pCMV-PAI-2 and coimmunoprecipitation of both proteins with anti-PAI-2 antibodies and (b) silencing of the PAI-2 gene using specific small interfering RNA (siRNA). Subsequently, cellular distribution of the PAI-2·proteasome complexes was established by immunogold staining and electron microscopy analyses. As judged by confocal microscopy, both proteins appeared in a diffuse cytosolic pattern, but they also could be found in a dense perinuclear and nuclear location. PAI-2 was not polyubiquitinated, suggesting that it bound to proteasome not as the substrate but rather as its inhibitor. Consistently, increased PAI-2 expression (a) abrogated degradation of degron analyzed after cotransfection of HeLa cells with pCMV-PAI-2 and pd2EGFP-N1, (b) prevented degradation of p53, as evidenced both by confocal microscopy and Western immunoblotting, and (c) inhibited proteasome cleavage of specific fluorogenic substrate. This suggests that PAI-2, in endothelial cells induced with inflammatory stimuli, can inhibit proteasome and thus tilt the balance favoring proapoptotic signaling.


Subject(s)
Endothelial Cells/metabolism , Plasminogen Activator Inhibitor 2/metabolism , Proteasome Endopeptidase Complex/metabolism , Cell Line , Endothelial Cells/drug effects , Endothelial Cells/ultrastructure , HeLa Cells , Humans , Immunoprecipitation , Lipopolysaccharides/pharmacology , Microscopy, Confocal , Microscopy, Electron, Transmission , Plasminogen Activator Inhibitor 2/genetics , Proteasome Endopeptidase Complex/ultrastructure , Protein Binding/drug effects , RNA Interference , Tumor Necrosis Factor-alpha/pharmacology , Ubiquitination/drug effects
10.
Thromb Res ; 128(5): 452-7, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21752432

ABSTRACT

INTRODUCTION: Previous studies showed that lumican, a small leucine-rich proteoglycan that binds to α2 integrin I domain, is an efficient inhibitor of cell adhesion and migration. In this report, we tested its effect on angiogenesis in vitro and in vivo. MATERIALS AND METHODS: Effect of lumican on angiogenesis was evaluated by in vitro capillary tube formation test performed between Fibrin II Gels or in Matrigel™ and in vivo by Matrigel(™) plug assay in BALB/c mice. Changes in matrix metalloproteinases expression caused by lumican were analyzed in endothelial cells by real-time PCR, Western immunoblotting and gelatin zymography. RESULTS: In unchallenged endothelial cells, Matrigel™ induced robust capillary morphogenesis. In contrast, tube formation was dramatically reduced by lumican, and by siRNA to ß1 integrin subunit mRNA but not by control siRNA. Similarly, lumican effectively inhibited neovascularization in vivo in assays using Matrigel™ plugs formed in BALB/c mice. Interestingly, lumican significantly reduced expression of matrix metalloproteinases, particularly MMP-14 that is known to activate other MMPs in close vicinity of endothelial cell membranes. CONCLUSIONS: Our results provide strong evidence that lumican affects angiogenesis both by interfering with α2ß1 receptor activity and downregulating proteolytic activity associated with surface membranes of endothelial cells.


Subject(s)
Chondroitin Sulfate Proteoglycans/pharmacology , Integrin alpha2beta1/antagonists & inhibitors , Keratan Sulfate/pharmacology , Matrix Metalloproteinase Inhibitors , Neovascularization, Physiologic/drug effects , Animals , Down-Regulation/drug effects , Down-Regulation/genetics , Endothelial Cells , Integrin alpha2beta1/metabolism , Integrin beta1/genetics , Lumican , Matrix Metalloproteinase 14/genetics , Mice , RNA, Small Interfering/pharmacology
11.
J Biol Chem ; 286(8): 6820-31, 2011 Feb 25.
Article in English | MEDLINE | ID: mdl-21135093

ABSTRACT

Plasminogen activator inhibitor type-1 (PAI-1), a multifunctional protein, is an important physiological regulator of fibrinolysis, extracellular matrix homeostasis, and cell motility. Recent observations show that PAI-1 may also be implicated in maintaining integrity of cells, especially with respect to cellular proliferation or apoptosis. In the present study we provide evidence that PAI-1 interacts with proteasome and affects its activity. First, by using the yeast two-hybrid system, we found that the α3 subunit of proteasome directly interacts with PAI-1. Then, to ensure that the PAI-1-proteasome complex is formed in vivo, both proteins were coimmunoprecipitated from endothelial cells and identified with specific antibodies. The specificity of this interaction was evidenced after transfection of HeLa cells with pCMV-PAI-1 and coimmunoprecipitation of both proteins with anti-PAI-1 antibodies. Subsequently, cellular distribution of the PAI-1-proteasome complexes was established by immunogold staining and electron microscopy analyses. Both proteins appeared in a diffuse cytosolic pattern but also could be found in a dense perinuclear and nuclear location. Furthermore, PAI-1 induced formation of aggresomes freely located in endothelial cytoplasm. Increased PAI-1 expression abrogated degradation of degron analyzed after cotransfection of HeLa cells with pCMV-PAI-1 and pd2EGFP-N1 and prevented degradation of p53 as well as IκBα, as evidenced both by confocal microscopy and Western immunoblotting.


Subject(s)
Cell Nucleus/metabolism , Cytoplasm/metabolism , Plasminogen Activator Inhibitor 1/metabolism , Cell Nucleus/genetics , Cytoplasm/genetics , HeLa Cells , Humans , I-kappa B Proteins/genetics , I-kappa B Proteins/metabolism , NF-KappaB Inhibitor alpha , Plasminogen Activator Inhibitor 1/genetics , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
12.
Exp Cell Res ; 317(6): 802-11, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21182838

ABSTRACT

Matrin 3 is an integral component of nuclear matrix architecture that has been implicated in interacting with other nuclear proteins and thus modulating the activity of proximal promoters. In this study, we evaluated the contribution of this protein to proliferation of endothelial cells. To selectively modulate matrin 3 expression, we used siRNA oligonucleotides and transfection of cells with a pEGFP-N1-Mtr3. Our data indicate that downregulation of matrin 3 is responsible for reduced proliferation and leads to necrosis of endothelial cells. This conclusion is supported by observations that reducing matrin 3 expression results in (a) producing signs of necrosis detected by PI staining, LDH release, and scatter parameters in flow cytometry, (b) affecting cell cycle progression. It does not cause (c) membrane asymmetry of cells as indicated by lack of Annexin V binding as well as (d) activation of caspase 3 and cleavage of PARP. We conclude that matrin 3 plays a significant role in controlling cell growth and proliferation, probably via formation of complexes with nuclear proteins that modulate pro- and antiapoptotic signaling pathways. Thus, degradation of matrin 3 may be a switching event that induces a shift from apoptotic to necrotic death of cells.


Subject(s)
Endothelial Cells/physiology , Nuclear Matrix-Associated Proteins/metabolism , RNA-Binding Proteins/metabolism , Apoptosis , Blotting, Western , Cell Cycle/physiology , Cell Line , Cell Line, Tumor , Cell Survival , Endothelial Cells/cytology , Endothelial Cells/metabolism , Gene Expression Regulation , HEK293 Cells , Humans , Microscopy, Confocal , RNA, Small Interfering/metabolism
13.
Ann N Y Acad Sci ; 1194: 60-71, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20536451

ABSTRACT

Aberrant expression of thymosin beta4 (Tbeta4) has recently been found to be associated with colorectal carcinoma (CRC) progression evidently due to an increase of the motility and invasion of tumor cells and the induction of a proangiogenic phenotype of endothelial cells. Both mechanisms depend upon matrix-degrading proteases, particularly plasmin and matrix metalloproteinases (MMPs) that are responsible for extensive tissue remodeling. Cleavage of ECM macromolecules weakens the structural integrity of tissues and exposes cryptic domains of extracellular components, which elicit biological responses distinct from intact molecules. Interestingly, signaling via integrins (alphaVbeta3, alpha5beta1) in CRC cells (HT29, CX1.1) is induced by Tbeta4 and VEGF-A only when they grow in 3D fibrin gels but not in 2D ones. The cells growing in 3D fibrin gels release upon Tbeta4 significant amounts of active MMPs (MMP-2, MMP-9, and MMP-7) that cause extensive proteolysis in their close vicinity. As evidenced by a variety of approaches (transfection experiments, coimmunoprecipitation, gene silencing with siRNA), we found that this involves interaction of Tbeta4 with Ku80, which has recently been described by us to mediate Tbeta4 intracellular activity.


Subject(s)
Colonic Neoplasms/metabolism , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Thymosin/metabolism , Cell Movement/genetics , Cells/metabolism , Colonic Neoplasms/genetics , Endothelial Cells/metabolism , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Humans , Integrins/genetics , Integrins/metabolism , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinases/genetics , Matrix Metalloproteinases/metabolism , Matrix Metalloproteinases/physiology , Neoplasms/genetics , Neoplasms/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction/genetics , Thymosin/genetics , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
14.
J Biol Chem ; 285(39): 29874-83, 2010 Sep 24.
Article in English | MEDLINE | ID: mdl-20562109

ABSTRACT

Recent evidence supports a role of protein-disulfide isomerase (PDI) in redox-controlled remodeling of the exofacial domains of α(IIb)ß(3) in blood platelets. The aim of this study was to explain whether Ero1α can be responsible for extracellular reoxidation of the PDI active site. We showed that Ero1α can be found on platelets and is rapidly recruited to the cell surface in response to platelet agonists. It is physically associated with PDI and α(IIb)ß(3), as suggested by colocalization analysis in confocal microscopy and confirmed by immunoprecipitation experiments. Apart from monomeric oxidized Ero1α, anti-α(IIb)ß(3) immunoprecipitates showed the presence of several Ero1α-positive bands that corresponded to the complexes α(IIb)ß(3)-PDI-Ero1α, PDI-Ero1α, and Ero1α-Ero1α dimers. It binds more efficiently to the activated α(IIb)ß(3) conformer, and its interaction is inhibited by RGD peptides. Ero1α appears to be involved in the regulation of α(IIb)ß(3) receptor activity because of the following: (a) blocking the cell surface Ero1α by antibodies leads to a decrease in platelet aggregation in response to agonists and a decrease in fibrinogen and PAC-1 binding, and (b) transfection of MEG01 with Ero1α increases α(IIb)ß(3) receptor activity, as indicated by increased binding of fibrinogen.


Subject(s)
Blood Platelets/metabolism , Membrane Glycoproteins/metabolism , Multiprotein Complexes/metabolism , Oxidoreductases/metabolism , Platelet Aggregation/physiology , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Protein Disulfide-Isomerases/metabolism , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Catalytic Domain , Fibrinogen/genetics , Fibrinogen/metabolism , Humans , Membrane Glycoproteins/genetics , Multiprotein Complexes/genetics , Oligopeptides/pharmacology , Oxidation-Reduction/drug effects , Oxidoreductases/genetics , Platelet Aggregation/drug effects , Platelet Glycoprotein GPIIb-IIIa Complex/genetics , Protein Binding/drug effects , Protein Binding/physiology , Protein Disulfide-Isomerases/genetics
15.
Exp Cell Res ; 316(6): 907-14, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-20096685

ABSTRACT

Endothelial cells in tumor vessels display unusual characteristics in terms of survival and angiogenic properties which result from the increased expression of VEGF-D and its autocrine effect. To evaluate mechanisms by which VEGF-D leads to such abnormal phenotype, we searched for proteins with modified expression in HUVECs enriched in the recombinant mature VEGF-D (VEGFD(DeltaNDeltaC)) delivered by adenovirus. Expression of membrane proteins in endothelial cells was characterized by FACS using anti-human IT-Box-135 antibodies. HUVECs transduced with Ad-VEGF-D(DeltaNDeltaC) revealed markedly increased expression of proteins involved in adhesion and migration such as (a) integrins (alphaVbeta5, alpha2beta1, alpha5beta1, alphaMbeta2, alphaLbeta2), (b) matrix metalloproteinases (MMP-2, MMP-9, and MMP-14), (c) components of fibrinolytic system (PAI-1, u-PAR), and (d) CD45, CD98, CD147. Interestingly, there also were numerous proteins with significantly reduced expression, particularly among surface exposed membrane proteins. Thus, it can be concluded that to induce proangiogenic phenotype and facilitate migration of HUVECs, VEGF-D(DeltaNDeltaC) not only upregulates expression of proteins known to participate in the cell-matrix interactions but also silences some membrane proteins which could interfere with this process.


Subject(s)
Autocrine Communication/physiology , Endothelial Cells/metabolism , Vascular Endothelial Growth Factor D/metabolism , Adenoviridae/genetics , Adenoviridae/metabolism , Animals , Cell Line , Cell Movement , Cell Proliferation , Endothelial Cells/cytology , Humans , Matrix Metalloproteinase 14/genetics , Matrix Metalloproteinase 14/metabolism , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mice , Plasminogen Activator Inhibitor 1/genetics , Plasminogen Activator Inhibitor 1/metabolism , Receptors, Urokinase Plasminogen Activator/genetics , Receptors, Urokinase Plasminogen Activator/metabolism , Transduction, Genetic , Vascular Endothelial Growth Factor D/genetics
16.
FEBS J ; 275(8): 1813-23, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18331351

ABSTRACT

Adhesive properties of endothelial cells are influenced by the thioldisulfide balance. However, the molecular mechanism of this effect is unclear, although recent observations indicate that integrin receptors may be direct targets for redox modulation. The purpose of this study was to examine whether protein disulfide isomerase (PDI) is directly involved in this process. As manganese ions are known to affect the thioldisulfide balance and activate integrins to maximal affinity, we searched for PDI interactions with integrins, particularly with alpha(V)beta(3), in Mn(2+)-treated endothelial cells. By employing confocal microscopy, flow cytometry and coimmunoprecipitation experiments, we showed that exposure of endothelial cells to Mn(2+) resulted in: (a) the appearance of surface protein thiol groups, which can be found in PDI and alpha(V)beta(3), and both proteins colocalizing on the cellular surface; and (b) the formation of the PDI-alpha(V)beta(3) complex, which dissociates upon reduction. In addition, PDI in a complex with alpha(V)beta(3) induces conversion of the integrin to the ligand-competent high-affinity state, as evidenced by increased binding of vitronectin. The membrane-impermeable sulfhydryl blockers 3-N-maleimidylpropionyl biocytin 3-N-maleimidylpropionyl biocytin and p-chloromercuriphenyl sulfonate, as well as the PDI inhibitors bacitracin, MA3 018, and MA3 019, abolished the binding of vitronectin and LM609 to endothelial cells that is activated by Mn(2+). Consistently, LM609 almost completely blocked binding of vitronectin to such cells. The formation of the PDI-alpha(V)beta(3) stoichiometric complex was further demonstrated by surface plasmon resonance analysis, which showed that the initial reversible binding of PDI becomes irreversible in the presence of Mn(2+), probably mediated by disulfide bonds. Thus, we show that Mn(2+) simultaneously modulates the thiol isomerase activity of PDI that is bound to alpha(V)beta(3) and induces its transition to the ligand-competent state, suggesting an alternative mechanism of integrin regulation.


Subject(s)
Endothelial Cells/metabolism , Integrin alphaVbeta3/metabolism , Protein Disulfide-Isomerases/metabolism , Cell Line , Endothelial Cells/drug effects , Humans , Ligands , Protein Binding , Vitronectin/metabolism , Zinc/pharmacology
17.
Acta Biochim Pol ; 55(1): 43-9, 2008.
Article in English | MEDLINE | ID: mdl-18246205

ABSTRACT

In this report, we describe proteomic analysis of corpora amylacea collected by postmortem laser microdissection from multiple sclerosis (MS) brain lesions. Using low level protein loads (about 30 microg), a combination of two-dimensional electrophoresis with matrix-assisted laser desorption/ionization-time of flight mass spectrometry and database interrogations we identified 24 proteins of suspected neuronal origin. In addition to major cytoskeletal proteins like actin, tubulin, and vimentin, we identified a variety of proteins implicated specifically in cellular motility and plasticity (F-actin capping protein), regulation of apoptosis and senescence (tumor rejection antigen-1, heat shock proteins, valosin-containing protein, and ubiquitin-activating enzyme E1), and enzymatic pathways (glyceraldehyde-3-dehydrogenase, protein disulfide isomerase, protein disulfide isomerase related protein 5, lactate dehydrogenase). Samples taken from regions in the vicinity of corpora amylacea showed only traces of cellular proteins suggesting that these bodies may represent remnants of neuronal aggregates with highly polymerized cytoskeletal material. Our data provide evidence supporting the concept that biogenesis of corpora amylacea involves degeneration and aggregation of cells of neuronal origin.


Subject(s)
Amyloid/chemistry , Brain/metabolism , Multiple Sclerosis/metabolism , Neurons/metabolism , Actins/metabolism , Apoptosis , Axons/metabolism , Axons/pathology , Brain/pathology , Cytoskeleton/metabolism , Electrophoresis, Gel, Two-Dimensional , Humans , Mass Spectrometry/methods , Proteomics/methods , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tubulin/metabolism , Vimentin/metabolism
18.
J Biol Chem ; 283(3): 1534-1544, 2008 Jan 18.
Article in English | MEDLINE | ID: mdl-17984099

ABSTRACT

Our data demonstrate that increased intracellular expression of thymosin beta4(Tbeta4) is necessary and sufficient to induce plasminogen activator inhibitor type 1 (PAI-1) gene expression in endothelial cells. To describe the mechanism of this effect, we produced Tbeta4 mutants with impaired functional motifs and tested their intracellular location and activity. Cytoplasmic distributions of Tbeta4((AcSDKPT/4A)), Tbeta4((KLKKTET/7A)), and Tbeta4((K16A)) mutants fused with green fluorescent protein did not differ significantly from those of wild-type Tbeta4. Overexpression of Tbeta4, Tbeta4((AcSDKPT/4A)), and Tbeta4((K16A)) affected intracellular formation of actin filaments. As expected, Tbeta4((K16A)) uptake by nuclei was impaired. On the other hand, overexpression of Tbeta4((KLKKTET/7A)) resulted in developing the actin filament network typical of adhering cells, indicating that the mutant lacked the actin binding site. The mechanism by which intracellular Tbeta4 induced the PAI-1 gene did not depend upon the N-terminal tetrapeptide AcSDKP and depended only partially on its ability to bind G-actin or enter the nucleus. Both Tbeta4 and Tbeta4((AcSDKPT/4A)) induced the PAI-1 gene to the same extent, whereas mutants Tbeta4((KLKKTET/7A)) and Tbeta4((K16A)) retained about 60% of the original activity. By proteomic analysis, the Ku80 subunit of ATP-dependent DNA helicase II was found to be associated with Tbeta4. Ku80 and Tbeta4 consistently co-immunoprecipitated in a complex from endothelial cells. Co-transfection of endothelial cells with the Ku80 deletion mutants and Tbeta4 showed that the C-terminal arm domain of Ku80 is directly involved in this interaction. Furthermore, down-regulation of Ku80 by specific short interference RNA resulted in dramatic reduction in PAI-1 expression at the level of both mRNA and protein synthesis. These data suggest that Ku80 functions as a novel receptor for Tbeta4 and mediates its intracellular activity.


Subject(s)
Actins/metabolism , Antigens, Nuclear/metabolism , DNA-Binding Proteins/metabolism , Thymosin/metabolism , Antigens, Nuclear/genetics , Cell Nucleus/metabolism , Cells, Cultured , DNA-Binding Proteins/genetics , Down-Regulation , Endothelial Cells/metabolism , Gene Expression Regulation , Green Fluorescent Proteins/metabolism , Humans , Ku Autoantigen , Mutant Proteins/metabolism , Nuclear Localization Signals , Plasminogen Activator Inhibitor 1/genetics , Plasminogen Activator Inhibitor 1/metabolism , Protein Binding , Protein Transport , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Recombinant Fusion Proteins/metabolism , Sequence Deletion , Subcellular Fractions/metabolism , Thymosin/genetics
19.
Ann N Y Acad Sci ; 1112: 123-39, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17495245

ABSTRACT

The early stages of angiogenesis are usually accompanied by the occurrence of vascular leakage, and the deposition of fibrin in extravascular spaces. Initially, the fibrin network acts as a sealing matrix, but later on also as a scaffolding for invading endothelial cells. This process is induced by angiogenic growth factors, particularly by vascular endothelial growth factor (VEGF). Angiogenesis involves proteolytic activities, in particular cell-bound urokinase/plasmin and matrix metalloproteinase (MMPs) activities that modulate the fibrin structure and affect adhesion and migration of endothelial cells. Recent data show that formation of new vessels may be stimulated by thymosin beta-4 (Tbeta-4), but it is still not clear whether Tbeta-4 alone is angiogenic or the angiogenic potential of Tbeta-4 is mediated by VEGF. In this report to further characterize Tbeta-4 angiogenic activity, we produced its mutants that were deprived of the N-terminal tetrapeptide AcSDKP (Tbeta-4((AcSDKPT/4A))), the actin-binding sequence KLKKTET (Tbeta-4((KLKKTET/7A))) and with the nuclear localization sequence damaged by a point mutation Lys16Ala (Tbeta-4((K16A))). Then we tested their activity to induce expression and release of MMPs as well as plasminogen activators inhibitor type-1 (PAI-1). We also analyzed their effect on migration and proliferation of endothelial cells in three-dimensional (3D) fibrin matrix as well as on their ability to stimulate the outgrowth of human endothelial cells in capillary-like tubular structures. Our data demonstrate that increased intracellular expression of Tbeta-4 and its mutants is necessary and sufficient to induce PAI-1 gene expression in endothelial cells. Similarly, they stimulate expression and release of MMP-1, -2, and -3. As evaluated by using specific inhibitors to these MMPs, they modified specifically the structure of fibrin and thus facilitated migration of endothelial cells. To sum up, our data show that the mechanism by which Tbeta-4 induced transition of endothelial cells from quiescent to proangiogenic phenotype is characterized by increased expression of PAI-1 and MMPs did not require the presence of the N-terminal sequence AcSDKP, and depended only partially on its ability to bind G-actin or to enter the nucleus.


Subject(s)
Cell Adhesion/drug effects , Endothelial Cells/physiology , Thymosin/pharmacology , Amino Acid Sequence , Animals , Cell Movement/drug effects , Endothelial Cells/drug effects , Humans , Mice , Molecular Sequence Data , Neoplasms/blood supply , Neovascularization, Pathologic/prevention & control , Neovascularization, Physiologic/drug effects , Thymosin/chemistry , Thymosin/genetics
20.
J Biol Chem ; 281(32): 23034-41, 2006 Aug 11.
Article in English | MEDLINE | ID: mdl-16723352

ABSTRACT

The A-domains within integrin beta subunits contain three metal sites termed the metal ion-dependent adhesion site (MIDAS), site adjacent to the metal ion-dependent adhesion site (ADMIDAS), and ligand-induced metal-binding site (LIMBS), and these sites are involved in ligand engagement. The selectivity of these metal sites and their role in ligand binding have been investigated by expressing a fragment corresponding to the beta3 A-domain, beta3-(109-352), and single point mutants in which each of the cation-binding sites has been disabled. Equilibrium dialysis experiments identified three Mn2+- and two Ca2+-binding sites with the LIMBS being the site that did not bind Ca2+. Although the ADMIDAS could bind Ca2+, it did not bind Mg2+. These results indicate that the Ca2+-specific site that inhibits ligand binding is the ADMIDAS. Two different assay systems, surface plasmon resonance and a microtiter plate assay, demonstrated that the beta3 A-domain fragment bound fibrinogen in the presence of 0.1 mm Ca2+ but not in 3 mm Ca2+. This behavior recapitulated the effects of Ca2+ on fibrinogen binding to alphavbeta3 but not alphaIIbbeta3. Disabling any of the three cation-binding sites abrogated fibrinogen binding. These results indicate that the specificities of the three metal-binding sites for divalent cations are distinct and that each site can regulate the ligand binding potential of the beta3 A-domain.


Subject(s)
Integrin beta3/chemistry , Integrin beta3/physiology , Binding Sites , DNA, Complementary/metabolism , Fibrinogen/chemistry , Humans , Kinetics , Ligands , Magnesium/chemistry , Manganese/chemistry , Metals/chemistry , Protein Binding , Protein Structure, Tertiary , Surface Plasmon Resonance
SELECTION OF CITATIONS
SEARCH DETAIL
...