Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Front Immunol ; 11: 573550, 2020.
Article in English | MEDLINE | ID: mdl-33362762

ABSTRACT

GPI-anchored uPAR is the receptor for the extracellular serine protease urokinase-type plasminogen activator (uPA). Though uPAR role in inflammatory processes is documented, underlying mechanisms are not fully understood. In this study we demonstrate that uPAR is a part of Toll-like receptor 4 (TLR4) interactome. Downregulation of uPAR expression resulted in diminished LPS-induced TLR4 signaling, less activation of NFκB, and decreased secretion of inflammatory mediators in myeloid and non-myeloid cells in vitro. In vivo uPAR-/- mice demonstrated better survival, strongly diminished inflammatory response and better organ functions in cecal ligation and puncture mouse polymicrobial sepsis model. Mechanistically, GPI-uPAR and soluble uPAR colocalized with TLR4 on the cell membrane and interacted with scavenger receptor CD36. Our data show that uPAR can interfere with innate immunity response via TLR4 and this mechanism represents a potentially important target in inflammation and sepsis therapy.


Subject(s)
Epithelial Cells/drug effects , Inflammation/metabolism , Lipopolysaccharides/pharmacology , Macrophages, Peritoneal/drug effects , Receptors, Urokinase Plasminogen Activator/metabolism , Sepsis/metabolism , Toll-Like Receptor 4/metabolism , Animals , CD36 Antigens/metabolism , Cytokines/metabolism , Disease Models, Animal , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Humans , Inflammation/genetics , Inflammation/microbiology , Inflammation/prevention & control , Inflammation Mediators/metabolism , Macrophages, Peritoneal/metabolism , Macrophages, Peritoneal/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , RAW 264.7 Cells , Receptors, Urokinase Plasminogen Activator/genetics , Sepsis/genetics , Sepsis/microbiology , Sepsis/prevention & control , Signal Transduction , Toll-Like Receptor 4/genetics
2.
J Cardiovasc Transl Res ; 12(5): 447-458, 2019 10.
Article in English | MEDLINE | ID: mdl-30840186

ABSTRACT

Calcification of aortic valves leads to aortic stenosis mainly in elderly individuals, but the underlying molecular mechanisms are still not understood. Here, we studied microRNA (miR, miRNA) expression and function in healthy and stenotic human aortic valves. We identified miR-21, miR-24, and miR-143 to be highly upregulated in stenotic aortic valves. Using luciferase reporter systems, we found direct binding of miR-143 to the 3'UTR region of the matrix gla protein (MGP), which in turn is a key factor to sustain homeostasis in aortic valves. In subsequent experiments, we demonstrated a therapeutic potential of miRNA regulation during calcification in cardiac valvular interstitial cells. Collectively, our data provide evidence that deregulated miR expression contributes to the development of stenotic valve disease and thus form novel therapeutic opportunities of this severe cardiovascular disease.


Subject(s)
Aortic Valve Stenosis/metabolism , Aortic Valve/metabolism , Aortic Valve/pathology , Calcinosis/metabolism , MicroRNAs/metabolism , 3' Untranslated Regions , Aged , Aged, 80 and over , Animals , Aortic Valve Stenosis/genetics , Aortic Valve Stenosis/pathology , Binding Sites , Calcinosis/genetics , Calcinosis/pathology , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Case-Control Studies , Cells, Cultured , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Female , Humans , Male , MicroRNAs/genetics , Osteogenesis , Signal Transduction , Sus scrofa , Up-Regulation , Matrix Gla Protein
3.
Mol Biol Rep ; 46(3): 3487-3496, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30847850

ABSTRACT

Bone abnormalities as a consequence of osteoblast deregulation are associated with several diseases such as diabetes and chronic kidney disease. Important role for oxidized low density lipoproteins (oxLDL) in the pathophysiology of bone disorders has been reported. However, little is known about the effects and mechanisms of oxLDL on the process of osteoblastogenesis in human mesenchymal stem cells (MSCs). We show that oxLDL concentrations of ~ 10-25 µg protein (0.43-1.0 µM MDA/mg protein) inhibited the differentiation of MSCs to osteoblasts. We demonstrate that the underlying mechanism entails the suppression of the Wnt signaling through the down-regulation of ß-catenin. Further, we show the association of scavenger receptor CD36 with the receptors LRP5/6 and Frizzled in mediating the oxLDL effects on the differentiation of MSCs to pre-osteoblasts. Inhibiting CD36 restored osteoblasts differentiation in the presence of oxLDL. Our findings suggest that oxLDL interferes with the canonical Wnt signaling pathway in a CD36 dependent manner leading to an inhibition of osteoblastogenesis.


Subject(s)
CD36 Antigens/metabolism , Lipoproteins, LDL/pharmacology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Osteoblasts/metabolism , Wnt Signaling Pathway/drug effects , Cell Differentiation/drug effects , Cell Line , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Lipoproteins, LDL/metabolism , Mesenchymal Stem Cells/metabolism , Osteogenesis/drug effects , Wnt Proteins/metabolism
4.
Sci Rep ; 8(1): 11604, 2018 08 02.
Article in English | MEDLINE | ID: mdl-30072716

ABSTRACT

Resorptive activity of osteoclasts is important for maintaining bone homeostasis. Endogenous compounds such as oxidized low density lipoprotein (oxLDL) have been shown to disturb this activity. While some studies have investigated the effects of oxLDL on the process of osteoclastogenesis, the underlying mechanism are not fully understood. We show here that oxLDL concentrations of ~10-25 µg protein (0.43-1.0 µM MDA/mg protein) completely blocked the formation of functional osteoclasts. The underlying mechanism implies an inhibition of autophagy that in turn leads to a decreased fusion of cathepsin K (CatK)-loaded lysosomal vesicles with the ruffled border membrane. As result, a lower secretion of CatK and impaired protonation of the resorption lacunae by vacuolar-ATPase (v-ATPase) is observed in the presence of oxLDL. We demonstrate that scavenger receptor A (SR-A) mediates oxLDL effects on osteoclastogenesis and repressing this receptor partially rescued oxLDL effects. Collectively, our data provides an insight into the possible mechanism of oxLDL on osteoclastogenesis suggesting that it does not perturb the packaging of CatK and v-ATPase (V-a3) in the secretory lysosome, but inhibits the fusion of these lysosomes to the ruffled border. The relevance of our findings suggests a distinct link between oxLDL, autophagy and osteoclastogenesis.


Subject(s)
Autophagy , Cathepsin K/metabolism , Cell Differentiation , Lipoproteins, LDL/metabolism , Osteoclasts/metabolism , Scavenger Receptors, Class A/metabolism , Humans , Osteoclasts/pathology
5.
Oncotarget ; 8(60): 101572-101590, 2017 Nov 24.
Article in English | MEDLINE | ID: mdl-29254187

ABSTRACT

Urokinase plasminogen activator receptor (PLAUR) has been implicated in a variety of physiological and pathological conditions. The multi-functionality of PLAUR is due to its capacity to interact with many co-receptors to regulate extracellular proteolysis and intracellular signaling. Recent reports are identifying novel functions of PLAUR which were not evident in the past; however, the molecular mechanisms of PLAUR signaling are not completely understood. Here, we have compared the transcriptomes of silencing control (sicon) and PLAUR silenced (PLAURsi) MDA-MB-231 breast cancer cells on treatment with radiation. We isolated RNA from the cells, synthesized cDNA and measured the gene expression changes by microarray. We identified 24 downregulated and 53 upregulated genes, which were significantly (P-value < 0.005) affected by PLAUR silencing. Our analysis revealed 415 canonical pathways and 743 causal disease networks affected on silencing PLAUR. Transcriptomic changes and predicted pathways supported and consolidated some of the earlier understanding in the context of PLAUR signaling; including our recent observations in DNA damage and repair process. In addition, we have identified several novel pathways where PLAUR is implicated.

6.
Cell Death Dis ; 7(9): e2383, 2016 09 29.
Article in English | MEDLINE | ID: mdl-27685627

ABSTRACT

Mechanisms of DNA damage and repair signaling are not completely understood that hinder the efficiency of cancer therapy. Urokinase-type plasminogen activator receptor (PLAUR) is highly expressed in most solid cancers and serves as a marker of poor prognosis. We show that PLAUR actively promotes DNA repair in cancer cells. On the contrary, downregulation of PLAUR expression results in delayed DNA repair. We found PLAUR to be essential for activation of Checkpoint kinase 1 (CHK1); maintenance of cell cycle arrest after DNA damage in a TP53-dependent manner; expression, nuclear import and recruitment to DNA-damage foci of RAD51 recombinase, the principal protein involved in the homologous recombination repair pathway. Underlying mechanism implies auto-/paracrine signaling of PLAUR/TLR4 receptor complex leading to activation of CHK1 and DNA repair. The signaling is induced by a danger molecule released by DNA-damaged cells and mediates, at least partially, activation of DNA-damage response. This study describes a new mechanism of DNA repair activation initiated by auto-/paracrine signaling of membrane receptors PLAUR/TLR4. It adds to the understanding of role of PLAUR in cancer and provides a rationale for therapeutic targeting of PLAUR/TLR4 interaction in TP53-positive cancers.


Subject(s)
Checkpoint Kinase 1/metabolism , DNA Damage , Rad51 Recombinase/metabolism , Receptors, Urokinase Plasminogen Activator/metabolism , Signal Transduction , Toll-Like Receptor 4/metabolism , Cell Cycle Checkpoints , Cell Nucleus/metabolism , DNA Repair , HEK293 Cells , HeLa Cells , Humans , Models, Biological , Phosphorylation , Protein Transport , RNA, Small Interfering/metabolism , Tumor Suppressor Protein p53/metabolism
7.
J Bone Miner Res ; 30(2): 379-88, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25196912

ABSTRACT

Bone remodeling is a dynamic process based on a fine-tuned balance between formation and degradation of bone. Osteoblasts (OBLs) are responsible for bone formation and bone resorption is mediated by osteoclasts (OCLs). The mechanisms regulating the OBL-OCL balance are critical in health and disease; however, they are still far from being understood. We reported recently that the multifunctional urokinase receptor (uPAR) mediates osteogenic differentiation of mesenchymal stem cells (MSCs) to OBLs and vascular calcification in atherosclerosis. Here, we address the question of whether uPAR may also be engaged in regulation of osteoclastogenesis. We show that uPAR mediates this process in a dual fashion. Thus, uPAR affected OBL-OCL interplay. We observed that osteoclastogenesis was significantly impaired in co-culture of monocyte-derived OCLs and in OBLs derived from MSCs lacking uPAR. We show that expression and release, from OBLs, of macrophage colony-stimulating factor (M-CSF), which is indispensable for OCL differentiation, was inhibited by uPAR loss. We further found that uPAR, on the other hand, controlled formation, differentiation, and functional properties of macrophage-derived OCLs. Expression of osteoclastogenic markers, such as tartrate-resistant acid phosphatase (TRAP) and cathepsin K, was impaired in OCLs derived from uPAR-deficient macrophages. The requirement of uPAR for osteoclastogenesis was further confirmed by immunocytochemistry and in bone resorption assay. We provide evidence that the underlying signaling mechanisms involve uPAR association with the M-CSF binding receptor c-Fms followed by c-Fms phosphorylation and activation of the PI3K/Akt/NF-κB pathway in OCLs. We further show that uPAR uses this pathway to regulate a balance between OCL differentiation, apoptosis, and cell proliferation. Our study identified uPAR as an important and multifaceted regulator of OBL-OCL molecular interplay that may serve as an attractive target in bone disease and ectopic calcification.


Subject(s)
Macrophage Colony-Stimulating Factor/metabolism , Osteoblasts/metabolism , Osteoclasts/metabolism , Osteogenesis , Signal Transduction , Urokinase-Type Plasminogen Activator/metabolism , Apoptosis , Cell Differentiation , Coculture Techniques , HEK293 Cells , Humans , Macrophages/metabolism , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Macrophage Colony-Stimulating Factor/metabolism
8.
PLoS One ; 9(7): e101529, 2014.
Article in English | MEDLINE | ID: mdl-24987841

ABSTRACT

DNA damage induced by numerous exogenous or endogenous factors may have irreversible consequences on the cell leading to cell cycle arrest, senescence and cell death. The DNA damage response (DDR) is powerful signaling machinery triggered in response to DNA damage, to provide DNA damage recognition, signaling and repair. Most anticancer drugs induce DNA damage, and DNA repair in turn attenuates therapeutic efficiency of those drugs. Approaches delaying DNA repair are often used to increase efficiency of treatment. Recent data show that ubiquitin-proteasome system is essential for signaling and repair of DNA damage. However, mechanisms providing regulation of proteasome intracellular localization, activity, and recruitment to DNA damage sites are elusive. Even less investigated are the roles of extranuclear signaling proteins in these processes. In this study, we report the involvement of the serine protease urokinase-type plasminogen activator receptor (uPAR) in DDR-associated regulation of proteasome. We show that in vascular smooth muscle cells (VSMC) uPAR activates DNA single strand break repair signaling pathway. We provide evidence that uPAR is essential for functional assembly of the 26S proteasome. We further demonstrate that uPAR mediates DNA damage-induced phosphorylation, nuclear import, and recruitment of the regulatory subunit PSMD6 to proteasome. We found that deficiency of uPAR and PSMD6 delays DNA repair and leads to decreased cell survival. These data may offer new therapeutic approaches for diseases such as cancer, cardiovascular and neurodegenerative disorders.


Subject(s)
DNA Breaks, Single-Stranded , DNA Repair , Muscle, Smooth, Vascular/metabolism , Receptors, Urokinase Plasminogen Activator/genetics , Active Transport, Cell Nucleus , Animals , Cell Line , Cell Survival , Cells, Cultured , Gene Deletion , Humans , Mice, Inbred C57BL , Muscle, Smooth, Vascular/cytology , Proteasome Endopeptidase Complex/metabolism , Receptors, Urokinase Plasminogen Activator/metabolism
9.
Stem Cells Dev ; 23(4): 352-62, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24192237

ABSTRACT

Vascular calcification is a severe consequence of several pathological processes with a lack of effective therapy. Recent studies suggest that circulating and resident mesenchymal stem cells (MSC) contribute to the osteogenic program of vascular calcification. Molecular mechanisms underlying MSC osteogenic potential and differentiation remain, however, sparsely explored. We investigated a role for the complement receptor C5aR in these processes. We found that expression of C5aR was upregulated upon differentiation of human MSC to osteoblasts. C5aR inhibition by silencing and specific antagonist impaired osteogenic differentiation. We demonstrate that C5aR expression upon MSC differentiation was regulated by the multifunctional urokinase receptor (uPAR). uPAR targeting by siRNA resulted in complete abrogation of C5aR expression and consequently in the inhibition of MSC-osteoblast differentiation. We elucidated the NFκB pathway as the mechanism utilized by the uPAR-C5aR axis. MSC treatment with the NFκB inhibitor completely blocked the differentiation process. Nuclear translocation of the p65 RelA component of the NFκB complex was induced under osteogenic conditions and impaired by the inhibition of uPAR or C5aR. Dual-luciferase reporter assays demonstrated enhanced NFκB signaling upon MSC differentiation, whereas uPAR and C5aR downregulation lead to inhibition of the NFκB activity. We show involvement of the Erk1/2 kinase in this cascade. In vivo studies in a uPAR/LDLR double knockout mouse model of diet-induced atherosclerosis revealed impaired C5aR expression and calcification in aortic sinus plaques in uPAR(-/-)/LDLR(-/-) versus uPAR(+/+)/LDLR(-/-) control animals. These results suggest that uPAR-C5aR axis via the underlying NFκB transcriptional program controls osteogenic differentiation with functional impact on vascular calcification in vivo.


Subject(s)
Cell Differentiation , Mesenchymal Stem Cells/physiology , Receptor, Anaphylatoxin C5a/metabolism , Receptors, Urokinase Plasminogen Activator/metabolism , Vascular Calcification/metabolism , Animals , Cells, Cultured , Humans , MAP Kinase Signaling System , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Osteoblasts/metabolism , Osteogenesis
10.
J Mol Cell Cardiol ; 66: 72-82, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24239845

ABSTRACT

The pathogenesis of atherosclerosis involves an imbalanced lipid metabolism and a deregulated immune response culminating in chronic inflammation of the arterial wall. Recent studies show that endogenous ligands, such as modified plasma lipoproteins, can trigger pattern recognition receptors (PRR) of innate immunity for cellular and humoral reactions. The underlying molecular pathways remain less explored. In this study, we investigated the mechanisms of inflammatory effects of oxidized low-density lipoproteins (oxLDL) on human primary coronary artery smooth muscle cells (VSMC). We show that already low concentration of oxLDL initiated atherogenic signals triggering VSMC transition to proinflammatory phenotype. oxLDL impaired the expression of contractile proteins and myocardin in VSMC and initiated changes in cell functional responses, including expression of proinflammatory molecules. The effects of oxLDL were abolished by downregulation of the multifunctional urokinase receptor (uPAR). In response to oxLDL uPAR associated with CD36 and TLR4, the two main PRR for both pathogen and endogenous ligands. We demonstrate that uPAR association with CD36 and TLR4 mediated oxLDL-induced and NF-κB-dependent G-CSF and GM-CSF expression and changes in VSMC contractile proteins. uPAR-mediated release of G-CSF and GM-CSF by VSMC affected macrophage behavior and production of MCP-1. We provide evidence for functional relevance of our in vitro findings to in vivo human atherosclerotic tissues. Our data imply uPAR as a part of a PRR cluster interfering structurally and functionally with CD36 and TLR4 and responding to endogenous atherogenic ligands. They further point to specific function of each component of this cluster in mediating the ultimate signaling pattern.


Subject(s)
Atherosclerosis/metabolism , CD36 Antigens/metabolism , Lipoproteins, LDL/pharmacology , Myocytes, Smooth Muscle/drug effects , Receptors, Urokinase Plasminogen Activator/metabolism , Toll-Like Receptor 4/metabolism , Atherosclerosis/genetics , Atherosclerosis/pathology , CD36 Antigens/genetics , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Chemotaxis , Contractile Proteins/genetics , Contractile Proteins/metabolism , Coronary Vessels/metabolism , Coronary Vessels/pathology , Granulocyte Colony-Stimulating Factor/biosynthesis , Granulocyte Colony-Stimulating Factor/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/pathology , Macrophages/drug effects , Macrophages/metabolism , Macrophages/pathology , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , NF-kappa B/genetics , NF-kappa B/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Primary Cell Culture , Receptors, Urokinase Plasminogen Activator/genetics , Toll-Like Receptor 4/genetics , Trans-Activators/genetics , Trans-Activators/metabolism
11.
Theranostics ; 3(7): 516-26, 2013.
Article in English | MEDLINE | ID: mdl-23843899

ABSTRACT

Current treatments for human coronary artery disease necessitate the development of the next generations of vascular bioimplants. Recent reports provide evidence that controlling cell orientation and morphology through topographical patterning might be beneficial for bioimplants and tissue engineering scaffolds. However, a concise understanding of cellular events underlying cell-biomaterial interaction remains missing. In this study, applying methods of laser material processing, we aimed to obtain useful markers to guide in the choice of better vascular biomaterials. Our data show that topographically treated human primary vascular smooth muscle cells (VSMC) have a distinct differentiation profile. In particular, cultivation of VSMC on the microgrooved biocompatible polymer E-shell induces VSMC modulation from synthetic to contractile phenotype and directs formation and maintaining of cell-cell communication and adhesion structures. We show that the urokinase receptor (uPAR) interferes with VSMC behavior on microstructured surfaces and serves as a critical regulator of VSMC functional fate. Our findings suggest that microtopography of the E-shell polymer could be important in determining VSMC phenotype and cytoskeleton organization. They further suggest uPAR as a useful target in the development of predictive models for clinical VSMC phenotyping on functional advanced biomaterials.


Subject(s)
Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/cytology , Urokinase-Type Plasminogen Activator/metabolism , Cell Communication , Cells, Cultured , Focal Adhesions/genetics , Focal Adhesions/metabolism , Humans , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Urokinase-Type Plasminogen Activator/genetics
12.
J Vasc Res ; 50(2): 109-23, 2013.
Article in English | MEDLINE | ID: mdl-23172421

ABSTRACT

The anthracycline doxorubicin is a widely used effective anti-cancer drug. However, its application and dosage are severely limited due to its cardiotoxicity. The exact mechanisms of doxorubicin-induced cardiotoxic side effects remain poorly understood. Even less is known about the impact of doxorubicin treatment on vascular damage. We found that low doses of doxorubicin induced a senescent response in human primary vascular smooth muscle cells (VSMC). We observed that expression of urokinase receptor (uPAR) was upregulated in response to doxorubicin. Furthermore, the level of uPAR expression played a decisive role in developing doxorubicin-induced senescence. uPAR silencing in human VSMC by means of RNA interference as well as uPAR knockout in mouse VSMC resulted in abrogation of doxorubicin-induced cellular senescence. On the contrary, uPAR overexpression promoted VSMC senescence. We further found that proteasomal degradation of telomeric repeat binding factor 2 (TRF2) mediates doxorubicin-induced VSMC senescence. Our results demonstrate that uPAR controls the ubiquitin-proteasome system in VSMC and regulates doxorubicin-induced TRF2 ubiquitination and proteasomal degradation via this mechanism. Therefore, VSMC senescence induced by low doses of doxorubicin may contribute to vascular damage upon doxorubicin treatment. uPAR-mediated TRF2 ubiquitination and proteasomal degradation are further identified as a molecular mechanism underlying this process.


Subject(s)
Cellular Senescence/drug effects , Doxorubicin/pharmacology , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Proteasome Endopeptidase Complex/metabolism , Receptors, Urokinase Plasminogen Activator/physiology , Telomeric Repeat Binding Protein 2/metabolism , Animals , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Doxorubicin/toxicity , Gene Expression Regulation/drug effects , Humans , Mice , Mice, Inbred C57BL , Myocytes, Smooth Muscle/metabolism , Primary Cell Culture , Proteasome Endopeptidase Complex/drug effects , Protein Processing, Post-Translational/drug effects , RNA Interference , RNA, Small Interfering/pharmacology , Receptors, Urokinase Plasminogen Activator/deficiency , Receptors, Urokinase Plasminogen Activator/genetics , Recombinant Fusion Proteins/physiology , Telomere/metabolism , Telomeric Repeat Binding Protein 2/antagonists & inhibitors , Telomeric Repeat Binding Protein 2/genetics , Transfection , Ubiquitination/drug effects , Umbilical Arteries/cytology
13.
Stem Cells Dev ; 21(17): 3104-13, 2012 Nov 20.
Article in English | MEDLINE | ID: mdl-22676452

ABSTRACT

Multipotent mesenchymal stem cells (MSCs) are promising candidates for regenerative cell-based therapy. The mechanisms underlying MSC differentiation and other functions relevant to therapeutic avenues remain however a matter of debate. Recent reports imply a critical role for intercellular contacts in MSC differentiation. We studied MSC differentiation to vascular smooth muscle cells (VSMCs) in a coculture model using human primary MSCs and VSMCs. We observed that under these conditions, MSCs did not undergo the expected differentiation process. Instead, they revealed an increased proliferation rate. The upregulated MSC proliferation was initiated by direct contacts of MSCs with VSMCs; indirect coculture of both cell types in transwells was ineffective. Intercellular contacts affected cell growth in a unidirectional fashion, since VSMC proliferation was not changed. We observed formation of so-called tunneling nanotubes (TNTs) between MSCs and VSMCs that revealed an intercellular exchange of a fluorescent cell tracker dye. Disruption of TNTs using cytochalasin D or latrunculin B abolished increased proliferation of MSCs initiated by contacts with VSMCs. Using specific fluorescent markers, we identified exchange of mitochondria via TNTs. By generation of VSMCs with mitochondrial dysfunction, we show that mitochondrial transfer from VSMCs to MSCs was required to regulate MSC proliferation in coculture. Our data suggest that MSC interaction with other cell types does not necessarily result in the differentiation process, but rather may initiate a proliferative response. They further point to complex machinery of intercellular communications at the place of vascular injury and to an unrecognized role of mitochondria in these processes.


Subject(s)
Cell Communication/drug effects , Cell Proliferation , Mesenchymal Stem Cells/cytology , Mitochondria/metabolism , Myocytes, Smooth Muscle/metabolism , Nanotubes , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cell Differentiation , Cells, Cultured , Coculture Techniques , Cytochalasin D/pharmacology , Endocytosis , Flow Cytometry , Fluorescent Dyes , Humans , Intercellular Junctions/drug effects , Intercellular Junctions/metabolism , Mesenchymal Stem Cells/metabolism , Microscopy, Confocal/methods , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/cytology , Thiazolidines/pharmacology
14.
Arterioscler Thromb Vasc Biol ; 32(1): 110-22, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22075245

ABSTRACT

OBJECTIVE: The urokinase-type plasminogen activator (uPA) and its specific receptor (uPAR) are a potent multifunctional system involved in vascular remodeling. The goal of the study was to unravel the mechanisms of uPA/uPAR-directed vascular smooth muscle cell (VSMC) differentiation. METHODS AND RESULTS: Using cultured human primary VSMCs, we identified a new molecular mechanism controlling phenotypic modulation in vitro and in vivo. We found that the urokinase-type plasminogen activator receptor (uPAR) acts together with the transcriptional coactivator myocardin to regulate the VSMC phenotype. uPAR, a glycosylphosphatidylinositol-anchored cell-surface receptor family member, undergoes ligand-induced internalization and nuclear transport in VSMCs. Platelet-derived growth factor receptor ß and SUMOylated RanGAP1 mediate this trafficking. Nuclear uPAR associates with myocardin, which is then recruited from the promoters of serum response factor target genes and undergoes proteasomal degradation. This chain of events initiates the synthetic VSMC phenotype. Using mouse carotid artery ligation model, we show that this mechanism contributes to adverse vascular remodeling after injury in vivo. We then cultured cells on a microstructured biomaterial and found that substrate topography induced uPAR-mediated VSMC differentiation. CONCLUSIONS: These findings reveal the transcriptional activity of uPAR, controlling the differentiation of VSMCs in a vascular disease model. They also suggest a new role for uPAR as a therapeutic target and as a marker for VSMC phenotyping on prosthetic biomaterials.


Subject(s)
Myocytes, Smooth Muscle/metabolism , Nuclear Proteins/metabolism , Receptors, Urokinase Plasminogen Activator/metabolism , Trans-Activators/metabolism , Vascular Diseases/metabolism , Active Transport, Cell Nucleus , Animals , Carotid Artery Injuries/metabolism , Carotid Artery Injuries/pathology , Cells, Cultured , Endocytosis , GTPase-Activating Proteins/antagonists & inhibitors , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , Humans , Mice , Mice, Knockout , Myocytes, Smooth Muscle/pathology , Phenotype , Proteasome Endopeptidase Complex/metabolism , RNA, Small Interfering/genetics , Receptors, Urokinase Plasminogen Activator/deficiency , Receptors, Urokinase Plasminogen Activator/genetics , Sumoylation , Urokinase-Type Plasminogen Activator/deficiency , Urokinase-Type Plasminogen Activator/genetics , Urokinase-Type Plasminogen Activator/metabolism , Vascular Diseases/pathology
15.
Arterioscler Thromb Vasc Biol ; 32(2): 449-58, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22155455

ABSTRACT

OBJECTIVE: The atherosclerotic lesion is characterized by lipid peroxide accumulation. Paraoxonase 1 (PON1) reduces atherosclerotic lesion oxidative stress, whereas urokinase-type plasminogen activator (uPA) increases oxidative stress in atherosclerotic lesions and contributes to the progression and complications of atherosclerosis. We hypothesized that uPA may promote oxidative stress in the arterial wall via modulation of PON1 activity. Because the liver is the main site for PON1 production, in the present study, we tested whether uPA influences PON1 expression in hepatocytes. METHODS AND RESULTS: HuH7 hepatocytes were incubated in culture with increasing concentrations of uPA. uPA decreased PON1 gene expression and activity in a dose-dependent manner and accordingly suppressed PON1 secretion from hepatocytes. This effect required uPA/uPA receptor interaction. uPA downregulated PON1 gene expression via inactivation of peroxisome proliferator-activated receptor-γ (PPARγ) activity, and this effect was dependent on uPA-mediated mitogen-activated protein kinase kinase activation. Mechanistic studies showed that uPA enhanced mitogen-activated protein kinase kinase-PPARγ interaction, resulting in PPARγ nuclear export to the cytosol. CONCLUSIONS: This study provides the first evidence that uPA interferes with PPARγ transcriptional activity in hepatocytes, resulting in downregulation of PON1 expression and its secretion to the medium. This may explain, at least in part, the prooxidative effect of uPA in the vascular wall.


Subject(s)
Aryldialkylphosphatase/metabolism , Cell Nucleus/metabolism , Down-Regulation/drug effects , Hepatocytes/drug effects , Hepatocytes/metabolism , PPAR gamma/metabolism , Urokinase-Type Plasminogen Activator/pharmacology , Active Transport, Cell Nucleus/drug effects , Active Transport, Cell Nucleus/physiology , Animals , Aryldialkylphosphatase/genetics , Cell Line, Tumor , Cell Nucleus/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Hepatocytes/cytology , Humans , MAP Kinase Kinase Kinases/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Oxidative Stress/drug effects , Rats , Rats, Inbred Lew , Receptors, Urokinase Plasminogen Activator/deficiency , Receptors, Urokinase Plasminogen Activator/genetics , Receptors, Urokinase Plasminogen Activator/metabolism , Signal Transduction/physiology
16.
Biochem Biophys Res Commun ; 410(3): 531-6, 2011 Jul 08.
Article in English | MEDLINE | ID: mdl-21679692

ABSTRACT

Recent evidence points to a multifunctional role of ZO-2, the tight junction protein of the MAGUK (membrane-associated guanylate kinase-like) family. Though ZO-2 has been found in cell types lacking tight junction structures, such as vascular smooth muscle cells (VSMC), little is known about ZO-2 function in these cells. We provide evidence that ZO-2 mediates specific homotypic cell-to-cell contacts between VSMC. Using mass spectrometry we found that ZO-2 is associated with the non-receptor tyrosine kinase Jak1. By generating specific ZO-2 constructs we further found that the N-terminal fragment of ZO-2 molecule is responsible for this interaction. Adenovirus-based expression of Jak1 inactive mutant demonstrated that Jak1 mediates ZO-2 tyrosine phosphorylation. By means of RNA silencing, expression of Jak1 mutant form and fluorescently labeled ZO-2 fusion protein we further specified that active Jak1, but not Jak1 inactive mutant, mediates ZO-2 localization to the sites of intercellular contacts. We identified the urokinase receptor uPAR as a pre-requisite for these cellular events. Functional requirement of the revealed signaling complex for VSMC network formation was confirmed in experiments using Matrigel and in contraction assay. Our findings imply involvement of the ZO-2 tight junction independent signaling complex containing Jak1 and uPAR in VSMC intercellular communications. This mechanism may contribute to vascular remodeling in occlusive cardiovascular diseases and in arteriogenesis.


Subject(s)
Cell Communication , Janus Kinase 1/metabolism , Membrane Proteins/metabolism , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/physiology , Tight Junctions/physiology , Cells, Cultured , Humans , Phosphorylation , Receptors, Urokinase Plasminogen Activator/metabolism , Tight Junctions/metabolism , Zonula Occludens-2 Protein
17.
Cardiovasc Res ; 90(1): 113-21, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21088115

ABSTRACT

AIMS: Multipotent mesenchymal stem cells (MSCs) have regenerative properties and are recognized as putative players in the pathogenesis of cardiovascular diseases. The underlying molecular mechanisms remain, however, sparsely explored. Our study was designed to elucidate a probable role for the multifunctional urokinase (uPA)/urokinase receptor (uPAR) system in MSC regulation. Though uPAR has been implicated in a broad spectrum of pathophysiological processes, nothing is known about uPAR in MSCs. METHODS AND RESULTS: uPAR was required to mobilize MSCs from the bone marrow (BM) of mice stimulated with granulocyte colony-stimulating factor (G-CSF) in vivo. An insignificant amount of MSCs was mobilized in uPAR(-/-) C57BL/6J mice, whereas in wild-type animals G-CSF induced an eight-fold increase of mobilized MSCs. uPAR(-/-) mice revealed up-regulated expression of G-CSF and stromal cell-derived factor 1 (CXCR4) receptors in BM. uPAR down-regulation leads to inhibition of human MSC migration, as shown in different migration assays. uPAR down- or up-regulation resulted in inhibition or stimulation of MSC differentiation into vascular smooth muscle cells (VSMCs) correspondingly, as monitored by changes in cell morphology and expression of specific marker proteins. Injection of fluorescently labelled MSCs in non-obese diabetic/severe combined immunodeficiency (NOD/SCID) mice after femoral artery wire injury demonstrated impaired engraftment of uPAR-deficient MSCs at the place of injury. CONCLUSIONS: These data suggest a multifaceted function of uPAR in MSC biology contributing to vascular repair. uPAR might guide and control the trafficking of MSCs to the vascular wall in response to injury or ischaemia and their differentiation towards functional VSMCs at the site of arterial injury.


Subject(s)
Cell Differentiation , Cell Movement , Mesenchymal Stem Cells/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Receptors, Urokinase Plasminogen Activator/metabolism , Vascular System Injuries/metabolism , Animals , Biomarkers/metabolism , Cell Shape , Disease Models, Animal , Femoral Artery/metabolism , Femoral Artery/pathology , Granulocyte Colony-Stimulating Factor/administration & dosage , HEK293 Cells , Hematopoietic Stem Cell Mobilization/methods , Humans , Male , Mesenchymal Stem Cell Transplantation , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Muscle, Smooth, Vascular/injuries , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Myocytes, Smooth Muscle/transplantation , RNA Interference , Receptors, CXCR4/metabolism , Receptors, Urokinase Plasminogen Activator/deficiency , Receptors, Urokinase Plasminogen Activator/genetics , Time Factors , Transfection , Vascular System Injuries/genetics , Vascular System Injuries/pathology , Vascular System Injuries/surgery
18.
J Vasc Res ; 47(5): 432-40, 2010.
Article in English | MEDLINE | ID: mdl-20145416

ABSTRACT

BACKGROUND: The growth-promoting effect of mechanical stress on vascular smooth muscle cells (VSMC) has been implicated in the progress of cardiovascular diseases related to elevated blood pressure. The underlying molecular mechanisms are, however, not completely defined. METHODS: We have studied primary human aortic VSMC using a model for multilateral stretch. Expression of the suppressor of cytokine signaling (SOCS) family member SOCS-1 and related molecular mechanisms were studied using TaqMan analysis, immunoblotting, protein silencing, specific cell treatment, immunoprecipitation and immunocytochemistry. RESULTS: Mechanical stretch inhibits SOCS-1 mRNA and protein expression. This effect was abolished by cell treatment with methyl-beta-cyclodextrin disrupting lipid rafts and with RGD peptide affecting integrins. Inhibition of integrin interaction with another cellular receptor, urokinase receptor (uPAR), as well as uPAR silencing also abolished stretch-induced SOCS-1 downregulation. Mechanical stretch resulted in uPAR redistribution to lipid rafts and in its colocalization with focal adhesion kinase (FAK). Stretch impairs polyubiquitination and proteosomal degradation of FAK leading to FAK upregulation in stretched VSMC. SOCS-1 silencing and inhibition of proteosomal degradation simulate this effect. CONCLUSION: Our study identifies SOCS-1 as a novel participant involved in the propagation of mechanical stimuli in human VSMC, which might be relevant for the development of cardiovascular diseases.


Subject(s)
Muscle, Smooth, Vascular/metabolism , Stress, Mechanical , Suppressor of Cytokine Signaling Proteins/biosynthesis , Cells, Cultured , Down-Regulation , Focal Adhesion Kinase 1/metabolism , Humans , Membrane Microdomains/metabolism , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/metabolism , Receptors, Urokinase Plasminogen Activator/genetics , Suppressor of Cytokine Signaling 1 Protein , Up-Regulation
19.
PLoS One ; 4(12): e8302, 2009 Dec 14.
Article in English | MEDLINE | ID: mdl-20011528

ABSTRACT

BACKGROUND: Members of the signal transducer and activator of transcription (Stat) family of transcription factors traverse the nuclear membrane through a specialized structure, called the nuclear pore complex (NPC), which represents a selective filter for the import of proteins. Karyophilic molecules can bind directly to a subset of proteins of the NPC, collectively called nucleoporins. Alternatively, the transport is mediated via a carrier molecule belonging to the importin/karyopherin superfamily, which transmits the import into the nucleus through the NPC. METHODOLOGY/PRINCIPAL FINDINGS: In this study, we provide evidence for an alternative Stat1 nuclear import mechanism, which is mediated by the shuttle protein nucleolin. We observed Stat1-nucleolin association, nuclear translocation and specific binding to the regulatory DNA element GAS. Using expression of nucleolin transgenes, we found that the nuclear localization signal (NLS) of nucleolin is responsible for Stat1 nuclear translocation. We show that this mechanism is utilized upon differentiation of myeloid cells and is specific for the differentiation step from monocytes to macrophages. CONCLUSIONS/SIGNIFICANCE: Our data add the nucleolin-Stat1 complex as a novel functional partner for the cell differentiation program, which is uniquely poised to regulate the transcription machinery via Stat1 and nuclear metabolism via nucleolin.


Subject(s)
Cell Differentiation , Cell Nucleus/metabolism , Monocytes/cytology , Monocytes/metabolism , Phosphoproteins/metabolism , RNA-Binding Proteins/metabolism , STAT1 Transcription Factor/metabolism , Active Transport, Cell Nucleus , Animals , Base Sequence , CD36 Antigens/metabolism , Cell Line , Gene Silencing , Humans , Macrophages/cytology , Macrophages/metabolism , Mice , Nuclear Localization Signals/metabolism , Phosphoproteins/chemistry , Protein Binding , RNA-Binding Proteins/chemistry , Structure-Activity Relationship , Time Factors , Nucleolin
20.
Biochem J ; 423(3): 343-51, 2009 Oct 12.
Article in English | MEDLINE | ID: mdl-19691446

ABSTRACT

The cholesterol-enriched membrane microdomains lipid rafts play a key role in cell activation by recruiting and excluding specific signalling components of cell-surface receptors upon receptor engagement. Our previous studies have demonstrated that the GPI (glycosylphosphatidylinositol)-linked uPAR [uPA (urokinase-type plasminogen activator) receptor], which can be found in lipid rafts and in non-raft fractions, can mediate the differentiation of VSMCs (vascular smooth muscle cells) towards a pathophysiological de-differentiated phenotype. However, the mechanism by which uPAR and its ligand uPA regulate VSMC phenotypic changes is not known. In the present study, we provide evidence that the molecular machinery of uPAR-mediated VSMC differentiation employs lipid rafts. We show that the disruption of rafts in VSMCs by membrane cholesterol depletion using MCD (methyl-beta-cyclodextrin) or filipin leads to the up-regulation of uPAR and cell de-differentiation. uPAR silencing by means of interfering RNA resulted in an increased expression of contractile proteins. Consequently, disruption of lipid rafts impaired the expression of these proteins and transcriptional activity of related genes. We provide evidence that this effect was mediated by uPAR. Similar effects were observed in VSMCs isolated from Cav1Z(-/-) (caveolin-1-deficient) mice. Despite the level of uPAR being significantly higher after the disruption of the rafts, uPA/uPAR-dependent cell migration was impaired. However, caveolin-1 deficiency impaired only uPAR-dependent cell proliferation, whereas cell migration was strongly up-regulated in these cells. Our results provide evidence that rafts are required in the regulation of uPAR-mediated VSMC phenotypic modulations. These findings suggest further that, in the context of uPA/uPAR-dependent processes, caveolae-associated and non-associated rafts represent different signalling membrane domains.


Subject(s)
Cell Differentiation/physiology , Membrane Microdomains/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Receptors, Urokinase Plasminogen Activator/metabolism , Animals , Anti-Bacterial Agents/pharmacology , Caveolin 1/genetics , Caveolin 1/metabolism , Cell Dedifferentiation/drug effects , Cell Dedifferentiation/physiology , Cell Differentiation/drug effects , Cell Movement/drug effects , Cell Movement/physiology , Cells, Cultured , Filipin/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Gene Silencing , Humans , Membrane Microdomains/genetics , Mice , Mice, Knockout , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/cytology , Receptors, Urokinase Plasminogen Activator/genetics , Transcription, Genetic/drug effects , Transcription, Genetic/physiology , Urokinase-Type Plasminogen Activator/genetics , Urokinase-Type Plasminogen Activator/metabolism , beta-Cyclodextrins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...