Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
2.
Sci Rep ; 12(1): 16878, 2022 10 07.
Article in English | MEDLINE | ID: mdl-36207386

ABSTRACT

Recent reports demonstrate that SARS-CoV-2 utilizes cell surface heparan sulfate as an attachment factor to facilitate the initial interaction with host cells. Heparan sulfate interacts with the receptor binding domain of SARS-CoV-2 spike glycoprotein, and blocking this interaction can decrease cell infection. We and others reported recently that the family of compounds of 2,5-dihydroxyphenylic acid interferes with the binding of the positively charged groove in growth factor molecules to negatively charged cell surface heparan sulfate. We hypothesized that Calcium Dobesilate (CaD)-calcium salt of 2,5-dihydroxyphenylic acid-may also interfere with the binding of SARS-CoV-2 spike protein to heparan sulfate. Using lentiviral SARS-CoV-2 spike protein pseudotyped particles we show that CaD could significantly reduce pseudovirus uptake into endothelial cells. On the contrary, CaD did not affect cell infection with VSVG-expressing lentivirus. CaD could also prevent retention of SARS-CoV-2 spike protein in ex vivo perfused mouse kidney. Using microfluidic culture of endothelial cells under flow, we show that CaD prevents spike protein interaction with heparan sulfate glycocalyx. Since CaD has no adverse side effects and is approved in humans for other medical indications, our findings can rapidly translate into clinical studies.


Subject(s)
COVID-19 Drug Treatment , Calcium Dobesilate , Animals , Calcium/metabolism , Endothelial Cells/metabolism , Heparitin Sulfate/metabolism , Heparitin Sulfate/pharmacology , Humans , Mice , Protein Binding , SARS-CoV-2 , Spike Glycoprotein, Coronavirus/metabolism , Virus Attachment
3.
Intensive Care Med Exp ; 9(1): 57, 2021 Nov 24.
Article in English | MEDLINE | ID: mdl-34817751

ABSTRACT

BACKGROUND: Disruption of the endothelial glycocalyx (eGC) is observed in septic patients and its injury is associated with multiple-organ failure and inferior outcomes. Besides this biomarker function, increased blood concentrations of shedded eGC constituents might play a mechanistic role in septic organ failure. We hypothesized that therapeutic plasma exchange (TPE) using fresh frozen plasma might influence eGC-related pathology by removing injurious mediators of eGC breakdown while at the time replacing eGC protective factors. METHODS: We enrolled 20 norepinephrine-dependent (NE > 0.4 µg/kg/min) patients with early septic shock (onset < 12 h). Sublingual assessment of the eGC via sublingual sidestream darkfield (SDF) imaging was performed. Plasma eGC degradation products, such as heparan sulfate (HS) and the eGC-regulating enzymes, heparanase (Hpa)-1 and Hpa-2, were obtained before and after TPE. A 3D microfluidic flow assay was performed to examine the effect of TPE on eGC ex vivo. Results were compared to healthy controls. RESULTS: SDF demonstrated a decrease in eGC thickness in septic patients compared to healthy individuals (p = 0.001). Circulating HS levels were increased more than sixfold compared to controls and decreased significantly following TPE [controls: 16.9 (8-18.6) vs. septic patients before TPE: 105.8 (30.8-143.4) µg/ml, p < 0.001; vs. after TPE: 70.7 (36.9-109.5) µg/ml, p < 0.001]. The Hpa-2 /Hpa-1 ratio was reduced in septic patients before TPE but normalized after TPE [controls: 13.6 (6.2-21.2) vs. septic patients at inclusion: 2.9 (2.1-5.7), p = 0.001; vs. septic patients after TPE: 13.2 (11.2-31.8), p < 0.001]. Ex vivo stimulation of endothelial cells with serum from a septic patient induced eGC damage that could be attenuated with serum from the same patient following TPE. CONCLUSIONS: Septic shock results in profound degradation of the eGC and an acquired deficiency of the protective regulator Hpa-2. TPE removed potentially injurious eGC degradation products and partially attenuated Hpa-2 deficiency. Trial registration clinicaltrials.gov NCT04231994, retrospectively registered 18 January 2020.

5.
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
7.
Cardiovasc Res ; 116(11): 1875-1886, 2020 09 01.
Article in English | MEDLINE | ID: mdl-31711127

ABSTRACT

AIMS: Peripartum cardiomyopathy (PPCM) is a life-threatening heart disease occurring in previously heart-healthy women. A common pathomechanism in PPCM involves the angiostatic 16 kDa-prolactin (16 kDa-PRL) fragment, which via NF-κB-mediated up-regulation of microRNA-(miR)-146a induces vascular damage and heart failure. We analyse whether the plasminogen activator inhibitor-1 (PAI-1) is involved in the pathophysiology of PPCM. METHODS AND RESULTS: In healthy age-matched postpartum women (PP-Ctrl, n = 53, left ventricular ejection fraction, LVEF > 55%), PAI-1 plasma levels were within the normal range (21 ± 10 ng/mL), but significantly elevated (64 ± 38 ng/mL, P < 0.01) in postpartum PPCM patients at baseline (BL, n = 64, mean LVEF: 23 ± 8%). At 6-month follow-up (n = 23), PAI-1 levels decreased (36 ± 14 ng/mL, P < 0.01 vs. BL) and LVEF (49 ± 11%) improved. Increased N-terminal pro-brain natriuretic peptide and Troponin T did not correlate with PAI-1. C-reactive protein, interleukin (IL)-6 and IL-1ß did not differ between PPCM patients and PP-Ctrl. MiR-146a was 3.6-fold (P < 0.001) higher in BL-PPCM plasma compared with PP-Ctrl and correlated positively with PAI-1. In BL-PPCM serum, 16 kDa-PRL coprecipitated with PAI-1, which was associated with higher (P < 0.05) uPAR-mediated NF-κB activation in endothelial cells compared with PP-Ctrl serum. Cardiac biopsies and dermal fibroblasts from PPCM patients displayed higher PAI-1 mRNA levels (P < 0.05) than healthy controls. In PPCM mice (due to a cardiomyocyte-specific-knockout for STAT3, CKO), cardiac PAI-1 expression was higher than in postpartum wild-type controls, whereas a systemic PAI-1-knockout in CKO mice accelerated peripartum cardiac fibrosis, inflammation, heart failure, and mortality. CONCLUSION: In PPCM patients, circulating and cardiac PAI-1 expression are up-regulated. While circulating PAI-1 may add 16 kDa-PRL to induce vascular impairment via the uPAR/NF-κB/miR-146a pathway, experimental data suggest that cardiac PAI-1 expression seems to protect the PPCM heart from fibrosis. Thus, measuring circulating PAI-1 and miR-146a, together with an uPAR/NF-κB-activity assay could be developed into a specific diagnostic marker assay for PPCM, but unrestricted reduction of PAI-1 for therapy may not be advised.


Subject(s)
Cardiomyopathies/blood , Peripartum Period/blood , Plasminogen Activator Inhibitor 1/blood , Puerperal Disorders/blood , Adult , Animals , Biomarkers/blood , Cardiomyopathies/diagnostic imaging , Cardiomyopathies/physiopathology , Case-Control Studies , Disease Models, Animal , Female , Humans , Mice, Knockout , Myocytes, Cardiac/metabolism , Parity , Plasminogen Activator Inhibitor 1/genetics , Pregnancy , Prognosis , Puerperal Disorders/diagnostic imaging , Puerperal Disorders/physiopathology , Recovery of Function , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Stroke Volume , Time Factors , Up-Regulation , Ventricular Function, Left
8.
Sci Rep ; 9(1): 16310, 2019 11 05.
Article in English | MEDLINE | ID: mdl-31690750

ABSTRACT

The development of effective central nervous system (CNS) drugs has been hampered by the lack of robust strategies to mimic the blood-brain barrier (BBB) and cerebrovascular impairments in vitro. Recent technological advancements in BBB modeling using induced pluripotent stem cells (iPSCs) allowed to overcome some of these obstacles, nonetheless the pertinence for their use in drug permeation study remains to be established. This mandatory information requires a cross comparison of in vitro and in vivo pharmacokinetic data in the same species to avoid failure in late clinical drug development. Here, we measured the BBB permeabilities of 8 clinical positron emission tomography (PET) radioligands with known pharmacokinetic parameters in human brain in vivo with a newly developed in vitro iPSC-based human BBB (iPSC-hBBB) model. Our findings showed a good correlation between in vitro and in vivo drug brain permeability (R2 = 0.83; P = 0.008) which contrasted with the limited correlation between in vitro apparent permeability for a set of 18 CNS/non-CNS compounds using the in vitro iPSCs-hBBB model and drug physicochemical properties. Our data suggest that the iPSC-hBBB model can be integrated in a flow scheme of CNS drug screening and potentially used to study species differences in BBB permeation.


Subject(s)
Blood-Brain Barrier/chemistry , Brain/diagnostic imaging , Induced Pluripotent Stem Cells/cytology , Neuroglia/cytology , Animals , Blood-Brain Barrier/diagnostic imaging , Brain/metabolism , Cell Differentiation , Cells, Cultured , Drug Evaluation, Preclinical , Humans , Induced Pluripotent Stem Cells/metabolism , Mice , Models, Biological , Neuroglia/metabolism , Permeability , Positron-Emission Tomography , Proof of Concept Study , Rats
9.
Sci Rep ; 9(1): 13591, 2019 09 19.
Article in English | MEDLINE | ID: mdl-31537875

ABSTRACT

The endothelial glycocalyx and its regulated shedding are important to vascular health. Endo-ß-D-glucuronidase heparanase-1 (HPSE1) is the only enzyme that can shed heparan sulfate. However, the mechanisms are not well understood. We show that HPSE1 activity aggravated Toll-like receptor 4 (TLR4)-mediated response of endothelial cells to LPS. On the contrary, overexpression of its endogenous inhibitor, heparanase-2 (HPSE2) was protective. The microfluidic chip flow model confirmed that HPSE2 prevented heparan sulfate shedding by HPSE1. Furthermore, heparan sulfate did not interfere with cluster of differentiation-14 (CD14)-dependent LPS binding, but instead reduced the presentation of the LPS to TLR4. HPSE2 reduced LPS-mediated TLR4 activation, subsequent cell signalling, and cytokine expression. HPSE2-overexpressing endothelial cells remained protected against LPS-mediated loss of cell-cell contacts. In vivo, expression of HPSE2 in plasma and kidney medullary capillaries was decreased in mouse sepsis model. We next applied purified HPSE2 in mice and observed decreases in TNFα and IL-6 plasma concentrations after intravenous LPS injections. Our data demonstrate the important role of heparan sulfate and the glycocalyx in endothelial cell activation and suggest a protective role of HPSE2 in microvascular inflammation. HPSE2 offers new options for protection against HPSE1-mediated endothelial damage and preventing microvascular disease.


Subject(s)
Endothelial Cells/cytology , Glucuronidase/genetics , Lipopolysaccharides/adverse effects , Sepsis/metabolism , Toll-Like Receptor 4/metabolism , Animals , Disease Models, Animal , Endothelial Cells/metabolism , Glucuronidase/blood , Glucuronidase/metabolism , Glycocalyx/metabolism , Heparitin Sulfate/metabolism , Humans , Male , Mice , Microfluidic Analytical Techniques , Sepsis/chemically induced , Signal Transduction
10.
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
11.
Nephrol Dial Transplant ; 34(6): 947-960, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30247663

ABSTRACT

BACKGROUND: Peritoneal membrane (PM) damage during peritoneal dialysis (PD) is mediated largely by high glucose (HG)-induced pro-inflammatory and neo-angiogenic processes, resulting in PM fibrosis and ultrafiltration failure. We recently demonstrated a crucial role for protein kinase C (PKC) isoform α in mesothelial cells. METHODS: In this study we investigate the role of PKCß in PM damage in vitro using primary mouse peritoneal macrophages (MPMΦ), human macrophages (HMΦ) and immortalized mouse peritoneal mesothelial cells (MPMCs), as well as in vivo using a chronic PD mouse model. RESULTS: We demonstrate that PKCß is the predominant classical PKC isoform expressed in primary MPMΦ and its expression is up-regulated in vitro under HG conditions. After in vitro lipopolysaccharides stimulation PKCß-/- MPMΦ demonstrates increased levels of interleukin 6 (IL-6), tumour necrosis factor α, and monocyte chemoattractant protein-1 and drastically decrease IL-10 release compared with wild-type (WT) cells. In vivo, catheter-delivered treatment with HG PD fluid for 5 weeks induces PKCß up-regulation in omentum of WT mice and results in inflammatory response and PM damage characterized by fibrosis and neo-angiogenesis. In comparison to WT mice, all pathological changes are strongly aggravated in PKCß-/- animals. Underlying molecular mechanisms involve a pro-inflammatory M1 polarization shift of MPMΦ and up-regulation of PKCα in MPMCs of PKCß-/- mice. Finally, we demonstrate PKCß involvement in HG-induced polarization processes in HMΦ. CONCLUSIONS: PKCß as the dominant PKC isoform in MPMΦ is up-regulated by HG PD fluid and exerts anti-inflammatory effects during PD through regulation of MPMΦ M1/M2 polarization and control of the dominant mesothelial PKC isoform α.


Subject(s)
Macrophages/metabolism , Peritoneal Dialysis/adverse effects , Protein Kinase C beta/deficiency , Animals , Chemokine CCL2/metabolism , Dialysis Solutions/metabolism , Disease Models, Animal , Epithelial Cells , Epithelium , Female , Glucose/metabolism , Humans , Inflammation , Lipopolysaccharides/pharmacology , Mice , Mice, Transgenic , Neovascularization, Pathologic , Omentum/metabolism , Peritoneal Fibrosis/metabolism , Peritoneum/metabolism , Protein Isoforms , Protein Kinase C-alpha/metabolism , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation
12.
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
13.
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.

14.
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
15.
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
16.
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
17.
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
18.
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
19.
Diabetes ; 62(4): 1167-74, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23434935

ABSTRACT

Activation of protein kinase C (PKC) has been implicated in the pathogenesis of diabetic nephropathy with proteinuria and peritubular extracellular matrix production. We have previously shown that the PKC isoforms α and ß mediate different cellular effects. PKC-ß contributes to hyperglycemia-induced renal matrix production, whereby PKC-α is involved in the development of albuminuria. We further tested this hypothesis by deletion of both isoforms and used a PKC inhibitor. We analyzed the phenotype of nondiabetic and streptozotocin (STZ)-induced diabetic homozygous PKC-α/ß double-knockout mice (PKC-α/ß(-/-)). After 8 weeks of diabetes mellitus, the high-glucose-induced renal and glomerular hypertrophy as well as transforming growth factor-ß1) and extracellular matrix production were diminished in the PKC-α/ß(-/-) mice compared with wild-type controls. Urinary albumin/creatinine ratio also was significantly reduced, however, it was not completely abolished in diabetic PKC-α/ß(-/-) mice. Treatment with CGP41252, which inhibits PKC-α and PKC-ß, is able to prevent the development of albuminuria and to reduce existing albuminuria in type 1 (STZ model) or type 2 (db/db model) diabetic mice. These results support our hypothesis that PKC-α and PKC-ß contribute to the pathogenesis of diabetic nephropathy, and that dual inhibition of the classical PKC isoforms is a suitable therapeutic strategy in the prevention and treatment of diabetic nephropathy.


Subject(s)
Diabetic Nephropathies/enzymology , Diabetic Nephropathies/prevention & control , Protein Kinase C-alpha/antagonists & inhibitors , Protein Kinase C/antagonists & inhibitors , Animals , Blood Glucose , Body Weight , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/enzymology , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Enzymologic , Isoenzymes/antagonists & inhibitors , Kidney/pathology , Male , Mice , Mice, Knockout , Organ Size , Protein Kinase C beta , Staurosporine/analogs & derivatives , Staurosporine/pharmacology
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL