Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
J Immunol ; 211(2): 199-208, 2023 07 15.
Article in English | MEDLINE | ID: mdl-37272840

ABSTRACT

The BAFF/APRIL-system with the two cytokines BAFF and APRIL and their three receptors, transmembrane activator and CAML interactor (TACI), BAFF receptor, and B-cell maturation Ag, is important for B cell maintenance. The BAFF/APRIL system is a therapeutic target in B cell-derived malignancies and autoimmune diseases. However, unexpected outcomes of clinical trials with atacicept (TACI-Fc) underline our incomplete understanding of this system. Shedding of the three receptors is one important regulatory element. In humans, TACI exists in two isoforms generated through alternative splicing in their extracellular portion: TACI-long (l) has two cysteine-rich domains, whereas TACI-short (s) lacks the first low-affinity one. In this study, we discriminated soluble (s) forms of TACI-l and TACI-s with newly generated mAbs and found that both were spontaneously released from activated human B cells, with a predominance of sTACI-l. Furthermore, sTACI-l was also the dominant isoform in human serum. Vaccination with the mRNA vaccine from BioNTech does not significantly affect the serum levels of sTACI-l. Both TACI-s and TACI-l were shed by a disintegrin and metalloproteinase domain-containing protein 10. TACI-l and TACI-s formed homo- and hetero-oligomers in soluble and membrane-bound forms. Both sTACI-l and sTACI-s acted as decoy receptors for BAFF, but only sTACI-l also efficiently inhibited APRIL. Dimerization of sTACI-l enhanced its decoy functions only slightly. Together, we extend our knowledge of the complexity of the BAFF/APRIL system by identifying and characterizing the two soluble isoforms of TACI.


Subject(s)
B-Lymphocytes , Transmembrane Activator and CAML Interactor Protein , Humans , Alternative Splicing , B-Cell Activating Factor/metabolism , B-Cell Activation Factor Receptor/genetics , Cytokines/genetics , Protein Isoforms/genetics , Transmembrane Activator and CAML Interactor Protein/genetics , Tumor Necrosis Factor Ligand Superfamily Member 13/genetics , Tumor Necrosis Factor Ligand Superfamily Member 13/metabolism
2.
Heart Int ; 7(2): e12, 2012 Jun 05.
Article in English | MEDLINE | ID: mdl-23185679

ABSTRACT

The ABCG1 protein is centrally involved in reverse cholesterol transport from the vessel wall. Investigation of the effects of ABCG1 overexpression or knockdown in vivo has produced controversial results and strongly depended on the gene intervention model in which it was studied. Therefore, we investigated the effect of local overexpression of human ABCG1 in a novel model of vessel wall-directed adenoviral gene transfer in atherosclerotic rabbits. We conducted local, vascular-specific gene transfer by adenoviral delivery of human ABCG1 (Ad-ABCG1-GFP) in cholesterol-fed atherosclerotic rabbits in vivo. Endothelial overexpression of ABCG1 markedly reduced atheroprogression (plaque size) and almost blunted vascular inflammation, as shown by markedly reduced macrophage and smooth muscle cell invasion into the vascular wall. Also endothelial function, as determined by vascular ultrasound in vivo, was improved in rabbits after gene transfer with Ad-ABCG1-GFP. Therefore, both earlier and later stages of atherosclerosis were improved in this model of somatic gene transfer into the vessel wall. In contrast to results in transgenic mice, over-expression of ABCG1 by somatic gene transfer to the atherosclerotic vessel wall results in a significant improvement of plaque morphology and composition, and of vascular function in vivo.

4.
Circulation ; 123(17): 1891-9, 2011 May 03.
Article in English | MEDLINE | ID: mdl-21502572

ABSTRACT

BACKGROUND: Blocking of glycoprotein VI-dependent pathways by interfering in vascular collagen sites is commonly seen as an attractive target for an antiplatelet therapy of acute atherosclerotic diseases such as myocardial infarction or stroke. Revacept (soluble dimeric glycoprotein VI-Fc fusion protein) has been shown to reduce platelet adhesion by blocking vascular collagen in plaques or erosion and to be safe in preclinical studies. A dose-escalating clinical phase I study was performed to assess the safety, tolerability, pharmacokinetics, and pharmacodynamics of Revacept in humans. METHODS AND RESULTS: In a first-in-humans study, 30 healthy men received a single intravenous administration of 10, 20, 40, 80, or 160 mg Revacept. The serum concentration-time courses of each dosage of Revacept showed a narrow variation and a concentration and time dependence. Revacept did not significantly affect the bleeding time. Collagen-induced platelet aggregation was dose-dependently inhibited up to 48 hours at lower doses and for 7 days after higher dose levels. In contrast, ADP- or thrombin receptor activating peptide-dependent platelet aggregation remained unaltered. There were no relevant drug-related adverse events or drug-related changes in laboratory parameters (biochemistry, hematology, and coagulation parameters). There were no drug-related changes in blood pressure, pulse rate, or ECG parameters (including 24-hour Holter monitoring). No anti-Revacept antibodies were detected. CONCLUSION: This phase I study demonstrated that Revacept is a safe and well-tolerated new antiplatelet compound with a clear dose-dependent pharmacokinetic profile with specific, dose-related inhibition of platelet aggregation despite completely unaltered general hemostasis. CLINICAL TRIAL REGISTRATION: URL: www.clinicaltrials.gov. Unique identifier: NCT 01042964. URL: eudract.ema.europa.eu. Identifier: 2005-004656-12.


Subject(s)
Glycoproteins/administration & dosage , Immunoglobulin Fc Fragments/administration & dosage , Platelet Aggregation Inhibitors/administration & dosage , Platelet Aggregation/drug effects , Platelet Membrane Glycoproteins/administration & dosage , Adult , Animals , CHO Cells , Collagen/administration & dosage , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Hemostasis/drug effects , Humans , Immunoglobulin Fc Fragments/adverse effects , Injections, Intravenous , Male , Platelet Aggregation Inhibitors/adverse effects , Platelet Aggregation Inhibitors/pharmacokinetics , Platelet Count , Platelet Membrane Glycoproteins/adverse effects , Platelet Membrane Glycoproteins/pharmacokinetics , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/adverse effects , Recombinant Fusion Proteins/pharmacokinetics , Young Adult
5.
Circ Res ; 108(6): 695-703, 2011 Mar 18.
Article in English | MEDLINE | ID: mdl-21293004

ABSTRACT

RATIONALE: There is strong evidence that oxidative modification of low-density lipoprotein (oxLDL) plays a critical role in atherogenesis and that oxLDL may profoundly influence the mechanical stability of atherosclerotic plaques. OBJECTIVE: To block oxLDL, we designed, expressed, and tested Fc-CD68, a soluble oxLDL binding protein consisting of human Fc and the extracellular domain of the human oxLDL-binding receptor CD68. METHODS AND RESULTS: Fc-CD68 bound with high specific affinity to oxLDL and strongly bound and colocalized with oxLDL in plaques. To study the effects of repeated administrations of Fc-CD68 on the progression of atherosclerosis and plaque vulnerability, 12- and 16-week old cholesterol-fed ApoE(-/-) mice received either Fc-CD68 (n = 6) or Fc control protein (n = 6 to 8) thrice weekly for 4 weeks. Macroscopic and histological analysis of Sudan red lipid staining showed strong and significant reduction of plaque extension in the aorta and in the aortic root, respectively. Histological analysis of pentachrome- and Sirius-stained sections of the brachiocephalic arteries of 20 week-old ApoE(-/-) mice revealed that Fc-CD68 significantly reduced the occurrence of spontaneous ruptures of established plaques by ≈20%, compared with Fc and drastically increased the collagen content of plaques. Furthermore, in immunostained sections of the brachiocephalic artery and the aortic root, Fc-CD68 reduced the infiltration of plaques with T lymphocytes, and macrophages by ≈50% and 30%, respectively. CONCLUSIONS: The oxLDL binding protein Fc-CD68 attenuates atherosclerosis and strengthens the stability of atherosclerotic plaques.


Subject(s)
Antigens, CD/genetics , Antigens, Differentiation, Myelomonocytic/genetics , Atherosclerosis/pathology , Brachiocephalic Trunk/drug effects , Brachiocephalic Trunk/pathology , Immunoglobulin Fc Fragments/genetics , Plaque, Atherosclerotic/pathology , Recombinant Fusion Proteins/pharmacology , Animals , Aorta/drug effects , Aorta/pathology , Apolipoproteins E/deficiency , Atherosclerosis/metabolism , Binding, Competitive , Carrier Proteins/metabolism , Carrier Proteins/pharmacology , Collagen/metabolism , Disease Progression , Humans , Inflammation/metabolism , Inflammation/pathology , Lipid Metabolism , Lipoproteins, LDL/metabolism , Macrophages/pathology , Male , Mice , Mice, Knockout , Plaque, Atherosclerotic/metabolism , Recombinant Fusion Proteins/metabolism , T-Lymphocytes/pathology
6.
J Mol Med (Berl) ; 88(9): 909-20, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20454888

ABSTRACT

The appearance of lipid-rich foam cells is a major feature of vulnerable atherosclerotic plaque formation. The transformation of macrophages into foam cells results from excessive uptake of cholesterol-rich particles by scavenger receptors such as CD68. We cloned a CD68-Fc immunoadhesin, a fusion protein consisting of the extracellular domain of the human CD68 and a human Fc domain, and investigated the function in vitro. Specific binding of CD68-Fc to OxLDL with an affinity of 10 nmol/L was determined by surface plasmon resonance and increased binding to lipid-rich human and ApoE(-/-) mice plaque tissue. This was confirmed both by immunohistochemical staining of CD68-Fc-treated paraffin sections from human plaques and by ELISA-based quantification of CD68-Fc binding to human atherosclerotic plaque extracts. In an in vitro model of macrophage/foam cell formation, CD68-Fc reduced foam cell formation significantly. This was caused both by interference of CD68-Fc with OxLDL uptake into macrophages and platelets and by the inhibition of platelet/OxLDL phagocytosis. Finally, expression of metalloproteinases by macrophages/foam cells was inhibited by CD68-Fc. In conclusion, CD68-Fc seems to be a promising new tool for preventing macrophage/foam cell formation. Thus, CD68-Fc might offer a novel therapeutic strategy for patients with acute coronary syndrome by modulating the generation of vulnerable plaques.


Subject(s)
Antigens, CD/chemistry , Antigens, Differentiation, Myelomonocytic/chemistry , Foam Cells/cytology , Immunoglobulin Fc Fragments/chemistry , Recombinant Fusion Proteins/chemistry , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/genetics , Antigens, Differentiation, Myelomonocytic/metabolism , Apolipoproteins E/metabolism , CHO Cells , Cricetinae , Cricetulus , Foam Cells/metabolism , Humans , Immunoglobulin Fc Fragments/genetics , Lipoproteins, LDL/metabolism , Macrophages/metabolism , Mice , Plaque, Atherosclerotic/metabolism , Receptors, Scavenger/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Surface Plasmon Resonance , Transfection
7.
J Mol Cell Cardiol ; 49(3): 532-42, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20430036

ABSTRACT

Glycoprotein VI (GPVI) mediates binding of platelets to subendothelial collagen during acute arterial thrombosis. GPVI interactions with the activated atherosclerotic vascular endothelium during early atherosclerosis, however, are not well understood. In ApoE-/- mice, platelet adhesion to atherosclerotic arteries was increased, as measured by intravital microscopy. This platelet adhesion was significantly inhibited by IV injection of GPVI-Fc (1 mg/kg body weight). Atherosclerosis in ApoE-/- mice was attenuated both after 7 and 10 weeks of treatment with the anti-GPVI antibody JAQ1 (2 mg/kg body weight i.p. twice weekly). Binding of GPVI-Fc (1 mg/kg IV) occurred to deeper layers, but also to the luminal site of plaques in atherosclerotic rabbits, but not to the vessel wall of healthy littermates. Gene transfer of GPVI-Fc to the carotid vascular wall significantly attenuated athero-progression and endothelial dysfunction in atherosclerotic rabbits in vivo. Specific binding of the soluble GPVI receptor (GPVI-Fc) to fibronectin was found in vitro to coated ELISA plates. Platelet adhesion to fibronectin was significantly inhibited both by GPVI-Fc and by the anti-GPVI antibody 5C4 ex vivo in flow chamber experiments. GPVI plays a role in platelet adhesion to atherosclerotic endothelium in the absence of plaque rupture. Inhibition of GPVI both via GPVI-Fc and anti-GPVI-antibodies results in protection against atherosclerosis in both cholesterol-fed rabbits and ApoE-/- mice. This novel mechanism of GPVI-mediated platelet adhesion-possibly via fibronectin-could relevantly contribute to platelet-triggered atheroprogression.


Subject(s)
Atherosclerosis/pathology , Endothelium, Vascular/metabolism , Fibronectins/metabolism , Platelet Adhesiveness/physiology , Platelet Membrane Glycoproteins/metabolism , Adenoviridae/genetics , Animals , Antibodies, Monoclonal/pharmacology , Apolipoproteins E/physiology , Atherosclerosis/metabolism , CHO Cells , Carotid Arteries/metabolism , Carotid Arteries/pathology , Cricetinae , Cricetulus , Endothelium, Vascular/pathology , Gene Transfer Techniques , Green Fluorescent Proteins , Mice , Mice, Inbred C57BL , Mice, Knockout , Platelet Activation , Platelet Membrane Glycoproteins/genetics , Platelet Membrane Glycoproteins/immunology , Rabbits
8.
Arterioscler Thromb Vasc Biol ; 30(6): 1127-36, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20378847

ABSTRACT

OBJECTIVE: To investigate the role of junctional adhesion molecule A (JAM-A) on adhesion and differentiation of human CD34(+) cells into endothelial progenitor cells. METHODS AND RESULTS: Tissue healing and vascular regeneration is a multistep process requiring firm adhesion of circulating progenitor cells to the vascular wall and their further differentiation into endothelial cells. The role of JAM-A in platelet-mediated adhesion of progenitor cells was investigated by adhesion assays in vitro and with the help of intravital fluorescence microscopy in mice. Preincubation of human CD34(+) progenitor cells with soluble JAM-A-Fc (sJAM-A-Fc) resulted in significantly decreased adhesion over immobilized platelets or inflammatory endothelium under high shear stress in vitro and after carotid ligation in vivo or ischemia/reperfusion injury in the microcirculation of mice. Human CD34(+) cells express JAM-A, as defined by flow cytometry and Western blot analysis. JAM-A mediates differentiation of CD34(+) cells to endothelial progenitor cells and facilitates CD34(+) cell-induced reendothelialization in vitro. Pretreatment of human CD34(+) cells with sJAM-A-Fc resulted in increased neointima formation 3 weeks after endothelial denudation in the carotid arteries of nonobese diabetic/severe combined immunodeficient mice. CONCLUSIONS: These results indicate that the expression of JAM-A on CD34(+) cells mediates adhesion to the vascular wall after injury and differentiation into endothelial progenitor cells, a mechanism potentially involved in vascular regeneration. Human CD34(+) cells express JAM-A, mediating their interaction with platelets and endothelial cells. Specifically, JAM-A expressed on human CD34(+) progenitor cells regulates their adhesion over immobilized platelets or inflammatory endothelium under high shear stress in vitro and after carotid ligation in vivo or ischemia/reperfusion injury in the microcirculation of mice. Moreover, it mediates differentiation of CD34(+) cells to endothelial progenitor cells and facilitates reendothelialization.


Subject(s)
Antigens, CD34/analysis , Carotid Artery Injuries/metabolism , Cell Adhesion Molecules/metabolism , Cell Adhesion , Cell Differentiation , Endothelial Cells/metabolism , Immunoglobulins/metabolism , Intestines/blood supply , Reperfusion Injury/metabolism , Stem Cells/metabolism , Animals , Blood Platelets/metabolism , Blotting, Western , CHO Cells , Carotid Artery Injuries/blood , Carotid Artery Injuries/immunology , Carotid Artery Injuries/pathology , Carotid Artery Injuries/physiopathology , Cell Adhesion Molecules/genetics , Cell Proliferation , Cricetinae , Cricetulus , Endothelial Cells/immunology , Endothelial Cells/transplantation , Flow Cytometry , Humans , Immunoglobulin Fc Fragments/metabolism , Immunoglobulins/genetics , Lymphocyte Function-Associated Antigen-1/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Microscopy, Fluorescence , Microscopy, Video , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Receptors, Cell Surface , Recombinant Fusion Proteins/metabolism , Reperfusion Injury/blood , Reperfusion Injury/immunology , Reperfusion Injury/pathology , Reperfusion Injury/physiopathology , Stem Cell Transplantation , Stem Cells/immunology , Time Factors , Transfection , Wound Healing
9.
Thromb Haemost ; 101(4): 682-6, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19350111

ABSTRACT

The Extracellular Matrix Metalloproteinase Inducer (EMMPRIN, CD147, basigin) is an immunoglobulin-like receptor expressed in various cell types. During cellular interactions homotypic EMMPRIN-EMMPRIN interactions are known to induce the synthesis of matrix metalloproteinases. Recently, we have identified EMMPRIN as a novel receptor on platelets. To our knowledge EMMPRIN has not been shown to serve as adhesion receptor, yet. Here we characterise platelet glycoprotein VI (GPVI) as a novel adhesion receptor for EMMPRIN. Human platelets were prestimulated with ADP and perfused over immobilised recombinant EMMPRIN-Fc or Fc-fragments under arterial shear conditions. ADP-stimulated platelets showed significantly enhanced rolling (but not enhanced firm adhesion) on immobilised EMMPRIN-Fc compared to Fc. Pretreatment of platelets with blocking mAbs anti-EMMPRIN or anti-GPVI leads to a significant reduction of rolling platelets on immobilised EMMPRIN-Fc, whereas pretreatment with blocking mAbs anti-p-selectin, anti-alpha4-integrin or anti-GPIIb/IIIa complex (20 microg/ml each) had no effect. Consistently, chinese hamster ovary (CHO) cells stably transfected with GPVI showed enhanced rolling (but not adhesion) on immobilised EMMPRIN-Fc in comparison to non-transfected CHO cells. Similarly, CHO cells stably transfected with EMMPRIN showed enhanced rolling on immobilised GPVI-Fc (or EMMPRIN-Fc) compared to non transfected CHO-cells. Finally, specific binding of EMMPRIN to GPVI was demonstrated by a modified ELISA and surface plasmon resonance technology with a dissociation constant of 88 nM. Platelet GPVI is a novel receptor for EMMPRIN and can mediate platelet rolling via GPVI-EMMPRIN interaction.


Subject(s)
Basigin/metabolism , Blood Platelets/metabolism , CD36 Antigens/metabolism , Cell Movement , Platelet Activation , Adenosine Diphosphate/metabolism , Animals , Antibodies, Monoclonal , Basigin/genetics , Basigin/immunology , CD36 Antigens/genetics , CD36 Antigens/immunology , CHO Cells , Cell Adhesion , Cricetinae , Cricetulus , Humans , Mice , P-Selectin/metabolism , Platelet Adhesiveness , Protein Binding , Recombinant Fusion Proteins/metabolism , Time Factors , Transfection
10.
Circ Res ; 102(3): 302-9, 2008 Feb 15.
Article in English | MEDLINE | ID: mdl-18048771

ABSTRACT

In atherosclerosis, circulating platelets interact with endothelial cells and monocytes, leading to cell activation and enhanced recruitment of leukocytes into the vascular wall. The invasion of monocytes is accompanied by overexpression of matrix metalloproteinases (MMPs), which are thought to promote atherosclerosis and trigger plaque rupture. Following interaction with itself, the extracellular matrix metalloproteinase inducer (EMMPRIN) induces MMP synthesis via a little-known intracellular pathway. Recently, we showed upregulation of EMMPRIN on monocytes during acute myocardial infarction. EMMPRIN also stimulates secretion of MMP-9 by monocytes and of MMP-2 by smooth muscle cells, indicating that it may be an important regulator of MMP activity. Expression of EMMPRIN on platelets has not been described until now. Here, we demonstrate that resting platelets show low surface expression of EMMPRIN, which is upregulated by various platelet stimulators (flow cytometry). EMMPRIN is located in the open canalicular system and in alpha granules of platelets (according to electron microscopy and sucrose gradient ultracentrifugation). Platelet stimulation with recombinant EMMPRIN-Fc induced surface expression of CD40L and P-selectin (according to flow cytometry), suggesting that EMMPRIN-EMMPRIN interaction activates platelets. Coincubation of platelets with monocytes induced EMMPRIN-mediated nuclear factor kappaB activation (according to Western blot) in monocytes with increased MMP-9 (zymography), interleukin-6, and tumor necrosis factor-alpha secretion (according to ELISA) by monocytes. In conclusion, EMMPRIN displays a new platelet receptor that is upregulated on activated platelets. Binding of EMMPRIN to platelets fosters platelet degranulation. Platelet-monocyte interactions via EMMPRIN stimulate nuclear factor kappaB-driven inflammatory pathways in monocytes, such as MMP and cytokine induction. Thus, EMMPRIN may represent a novel target to diminish the burden of protease activity and inflammation in atherosclerosis.


Subject(s)
Basigin/metabolism , Blood Platelets/metabolism , Gene Expression Regulation , Monocytes/metabolism , NF-kappa B/metabolism , Platelet Activation , Atherosclerosis/metabolism , Atherosclerosis/pathology , Basigin/genetics , Basigin/pharmacology , Blood Platelets/ultrastructure , CD40 Ligand/metabolism , Coculture Techniques , Endothelial Cells/metabolism , Endothelial Cells/ultrastructure , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Humans , Immunoglobulin Constant Regions/genetics , Immunoglobulin Constant Regions/metabolism , Immunoglobulin Constant Regions/pharmacology , Inflammation/metabolism , Inflammation/pathology , Interleukin-6/metabolism , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Monocytes/ultrastructure , P-Selectin/metabolism , Platelet Activation/drug effects , Platelet Activation/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , Secretory Vesicles/metabolism , Secretory Vesicles/ultrastructure , Tumor Necrosis Factor-alpha/metabolism
11.
Thromb Haemost ; 95(5): 763-6, 2006 May.
Article in English | MEDLINE | ID: mdl-16676064

ABSTRACT

Platelet-mediated thrombus formation at the site of vascular injury is a major trigger for thrombo-ischemic complications after coronary interventions. The platelet collagen receptor glycoprotein VI (GPVI) plays a critical role in the initiation of arterial thrombus formation. Endothelial denudation of the right carotid artery in rabbits was induced through balloon injury. Subsequently, local delivery of soluble, dimeric fusion protein of GPVI (GPVI-Fc) (n = 7) or control Fc (n = 7) at the site of vascular injury was performed with a modified double-balloon drug-delivery catheter. Thrombus area within the injured carotid artery was quantified using a computer-assisted image analysis and was used as index of thrombus formation. The extent of thrombus formation was significantly reduced in GPVI-Fc- compared with control Fc-treated carotid arteries (relative thrombus area, GPVI-Fc vs. Fc: 9.3 +/- 4.2 vs. 2.3 +/- 1.7, p < 0.001). Local delivery of soluble GPVI resulted in reduced thrombus formation after catheter-induced vascular injury. These data suggest a selective pharmacological modulation of GPVI-collagen interactions to be important for controlling onset and progression of pathological arterial thrombosis, predominantly or even exclusively at sites of injured carotid arteries in the absence of systemic platelet therapy.


Subject(s)
Platelet Membrane Glycoproteins/administration & dosage , Thrombosis/prevention & control , Animals , Carotid Artery Injuries , Catheterization , Drug Delivery Systems , Endothelium, Vascular/injuries , Platelet Membrane Glycoproteins/pharmacology , Rabbits , Solubility , Thrombosis/drug therapy
12.
Am J Physiol Heart Circ Physiol ; 291(2): H488-95, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16603685

ABSTRACT

The sodium-calcium exchanger (NCX) is discussed as one of the key proteins involved in heart failure. However, the causal role and the extent to which NCX contributes to contractile dysfunction during heart failure are poorly understood. NCX overexpression was induced by infection with an adenovirus coding for NCX, which coexpressed green fluorescence protein (GFP) (AdNCX) by ex vivo gene transfer to nonfailing and failing rabbit cardiomyocytes. Myocardial gene transfer in rabbits in vivo was achieved by adenoviral delivery via aortic cross-clamping. Peak cell shortening of cardiomyocytes was determined photo-optically. Hemodynamic parameters in vivo were determined by echocardiography (fractional shortening) and tip catheter [maximal first derivative of left ventricular (LV) pressure (dP/dt(max)); maximal negative derivative of LV pressure (-dP/dt(max))]. Peak cell shortening was depressed after NCX gene delivery in isolated nonfailing and in failing cardiomyocytes. In nonfailing rabbits in vivo, basal systolic contractility (fractional shortening and dP/dt(max)) and maximum rate of LV relaxation (-dP/dt(max)) in vivo were largely unaffected after NCX overexpression. However, during heart failure, long-term NCX overexpression over 2 wk significantly improved fractional shortening and dP/dt(max) compared with AdGFP-infected rabbits, both without inotropic stimulation and after beta-adrenergic stimulation with isoproterenol. -dP/dt(max) was also improved after NCX overexpression in the failing rabbits group. These results indicate that short-term effects of NCX overexpression impair contractility of isolated failing and nonfailing rabbit cardiomyocytes. NCX overexpression over 2 wk in vivo does not seem to affect myocardial contractility in nonfailing rabbits. Interestingly, in vivo overexpression of NCX decreased the progression of systolic and diastolic contractile dysfunction and improved beta-adrenoceptor-mediated contractile reserve in heart failure in rabbits in vivo.


Subject(s)
Heart Failure/metabolism , Heart Failure/physiopathology , Sodium-Calcium Exchanger/biosynthesis , Adenoviridae/genetics , Animals , Blotting, Western , Cells, Cultured , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Dogs , Electrocardiography , Gene Transfer Techniques , Genetic Vectors , Heart Function Tests , Heart Rate/physiology , Hemodynamics/physiology , Myocardial Contraction/physiology , Myocytes, Cardiac/metabolism , Rabbits , Recombinant Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Ventricular Function, Left/physiology
13.
Circulation ; 113(6): 834-41, 2006 Feb 14.
Article in English | MEDLINE | ID: mdl-16461815

ABSTRACT

BACKGROUND: Matrix metalloproteinases (MMPs) are thought to promote progression of atherosclerosis and cardiovascular complications such as plaque rupture. It has been suggested that, on tumor cells, the extracellular MMP inducer (EMMPRIN) is involved in MMP synthesis by as yet unknown mechanisms. On cardiovascular cells, regulation of EMMPRIN in vivo or any functional relevance for MMP induction in vitro has not yet been studied. Thus, we studied EMMPRIN expression on monocytes in acute myocardial infarction (MI) and its potential relevance for MMP activation. METHODS AND RESULTS: In 20 patients with acute MI, surface expression of EMMPRIN was significantly enhanced on monocytes compared with in 20 patients with chronic stable angina. EMMPRIN upregulation was associated with increased expression of the membrane type 1 MMP (MT1-MMP) on monocytes (flow cytometry) as well as MMP-9 activity (gelatin zymography) in the plasma. At 6 months after successful revascularization, EMMPRIN, MT1-MMP, and MMP-9 had normalized. The secretion of MMP-9 by monocytes was induced by monocyte adhesion to immobilized recombinant EMMPRIN or to EMMPRIN-transfected Chinese hamster ovary cells. Moreover, adherent EMMPRIN-transfected monocytic cells stimulated MMP-2 activity of human vascular smooth muscle cells. Gene silencing of EMMPRIN by small-interfering RNA hindered lipopolysaccharide-induced monocyte secretion of MMP-9, indicating a predominant role of EMMPRIN in MMP-9 induction. CONCLUSIONS: EMMPRIN and MT1-MMP are upregulated on monocytes in acute MI. During cellular interactions, EMMPRIN stimulates MMP-9 in monocytes and MMP-2 in smooth muscle cells, indicating that EMMPRIN may display a key regulatory role for MMP activity in cardiovascular pathologies.


Subject(s)
Basigin/genetics , Matrix Metalloproteinases/metabolism , Monocytes/metabolism , Myocardial Infarction/etiology , Adult , Aged , Aged, 80 and over , Atherosclerosis , Basigin/metabolism , Basigin/physiology , Cell Adhesion , Enzyme Activation , Extracellular Matrix Proteins , Female , Gene Expression Regulation , Humans , Male , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinases, Membrane-Associated , Middle Aged , Muscle, Smooth, Vascular/cytology , Myocardial Infarction/enzymology , Myocardial Infarction/pathology , Up-Regulation
14.
Thromb Haemost ; 94(3): 555-61, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16268472

ABSTRACT

Cell adhesion and proteolytic matrix degradation are central processes in atherosclerosis. Being a member of the family of ADAMs ("a disintegrin and metalloproteinase"), metargidin (ADAM15) combines a metalloproteinase domain and an RGD aminoacid sequence. We studied the potential role of ADAM15 as an adhesion receptor on endothelial cells and interactions between platelets and ADAM15 with respect to platelet adhesion, activation and thrombus formation. ADAM15 was found to be expressed on cultured endothelial cells (HUVEC). Platelet adhesion to immobilized recombinant ADAM15 was effectively enhanced under both static and high shear rate conditions reaching the maximum level of adhesion to fibrinogen. Consistently, platelet adhesion onto ADAM15 overexpressing endothelial cells was significantly increased. Adhesion to ADAM15 was reduced by blockade of GPIIb-IIIa using neutralizing anti-alpha(IIb)beta3 mAbs (7E3, 2G12), but not by anti-alpha(v)beta3 (LM609). Soluble ADAM15 binds to activated but not to resting GPIIb-IIIa. Moreover, platelets adherent to ADAM15 additionally attracted platelets under high shear rates indicating an initial role of platelet-ADAM15 interactions for thrombus formation. Furthermore, incubation of platelets with soluble ADAM15 showed a dose-dependent increase in secretion of CD62P and CD40L. ADAM15 is expressed on endothelial cells and can serve as an adhesion receptor for platelets via GPIIb-IIIa binding. Platelet adhesion to ADAM15 leads to platelet activation, secretion and promotes thrombus formation. Thus, ADAM15 may represent a novel target for antithrombotic strategies in cardiovascular pathologies.


Subject(s)
ADAM Proteins/metabolism , Blood Platelets/metabolism , Cell Communication , Endothelial Cells/metabolism , Membrane Proteins/metabolism , Platelet Adhesiveness , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Receptors, Cell Surface/metabolism , ADAM Proteins/genetics , Animals , Arteriosclerosis/etiology , Arteriosclerosis/physiopathology , Blood Platelets/immunology , CD40 Ligand/metabolism , CHO Cells , Coculture Techniques , Cricetinae , Cricetulus , Endothelial Cells/drug effects , Genes, Reporter , Green Fluorescent Proteins , Humans , Interferon-gamma/pharmacology , Membrane Proteins/genetics , P-Selectin/metabolism , Platelet Glycoprotein GPIIb-IIIa Complex/genetics , Thrombosis/etiology , Thrombosis/physiopathology , Transfection , Tumor Necrosis Factor-alpha/pharmacology
15.
Circ Res ; 97(9): 928-36, 2005 Oct 28.
Article in English | MEDLINE | ID: mdl-16166553

ABSTRACT

A hyperadrenergic state is one of the key features of human and experimental heart failure. Decreased densities and activities of the presynaptic neuronal norepinephrine (NE) transporter uptake-1 occur both in patients and animal models. It is currently unclear to what extent the reduction of uptake-1 contributes to the deterioration of heart failure. Therefore, we investigated the effects of myocardial overexpression of uptake-1 in both nonfailing rabbit hearts and in an animal model of heart failure. Heart failure was induced in rabbits by rapid ventricular pacing. Adenoviral gene transfer was used to overexpress uptake-1 in the myocardium. Uptake-1 overexpression led to increased NE uptake capacity into the myocardium. In contrast, systemic plasma NE levels in uptake-1-overexpressing failing rabbits (uptake-1-CHF) did not differ from controls. Downregulation of SERCA-2 and beta-adrenergic receptors in the failing myocardium was significantly reversed after uptake-1 overexpression. Uptake-1 overexpression significantly improved left ventricular (LV) diameters (LV end-diastolic diameter: in GCP-overexpressing failing rabbits (GFP-CHF), 17.4+/-0.4 mm; in uptake-1-CHF rabbits, 15.6+/-0.6 mm) and systolic contractility (fractional shortening: GFP-CHF, 20.7+/-0.6%; uptake-1-CHF, 27.3+/-0.7%), as assessed by echocardiography at the end of the heart failure protocol. Intraventricular tip catheter measurements revealed enhanced contractile reserve (dP/dt max with isoproterenol 1.0 microg/kg: GFP-CHF, 6964+/-230 mm Hg/sec; uptake-1-CHF, 7660+/-315 mm Hg/sec) and LV relaxation (dP/dt min with isoproterenol 1.0 microg/kg: GFP-CHF: -3960+/-260 mm Hg/sec; uptake-1-CHF, -4910+/-490 mm Hg/sec). End-diastolic filling pressures (GFP-CHF, 8.5+/-1.2 mm Hg; uptake-1-CHF, 5.6+/-0.7 mm Hg) tended to be lower in uptake-1 overexpressing animals. In summary, local overexpression of uptake-1 in the myocardium results in marked structural and functional improvement of heart failure, thus underlining the importance of uptake-1 as a key protein in heart failure.


Subject(s)
Genetic Therapy , Heart Failure/therapy , Norepinephrine Plasma Membrane Transport Proteins/genetics , Adenoviridae/genetics , Animals , HeLa Cells , Heart Failure/physiopathology , Humans , Myocardial Contraction , Myocardium/metabolism , Norepinephrine/metabolism , PC12 Cells , Rabbits , Rats , Transgenes , Ventricular Dysfunction, Left/therapy , Weight Gain
16.
Diabetes ; 53(8): 2117-21, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15277394

ABSTRACT

Diabetes is associated with an enhanced collagen-mediated platelet activation that contributes significantly to thromboischemic complications. In this study, the platelet collagen receptor glycoprotein VI (GPVI) was studied in 385 patients with type 2 diabetes. Surface expression of the platelet Fc receptor that forms a functional complex with GPVI was significantly increased in patients with diabetes compared with those without diabetes (P = 0.02). Fc receptor expression correlated with GPVI expression and was found to be independently associated with diabetes (r = 0.529, P < 0.001). Stimulation of GPVI through a specific anti-GPVI monoclonal antibody significantly enhanced surface expression of CD40L (P = 0.006). Because CD40L is a potent platelet-derived cytokine that is involved in thrombosis and atherosclerosis, we evaluated the effect of GPVI-mediated release of CD40L on activation of endothelial cells. Coincubation of GPVI-stimulated platelets resulted in substantial enhanced endothelial surface expression of CD62P, alphavbeta3, and intercellular adhesion molecule 1 (P < 0.05) and secretion of monocyte chemoattractant protein 1 of cultured human umbilical vein endothelial cells (P < 0.01). These results suggest that the function of collagen receptor GPVI is altered in type 2 diabetes and may play an important role in atherothrombotic complications. Inhibition of GPVI may be a promising pharmacological target in the treatment of high-risk diabetic patients.


Subject(s)
Blood Platelets/physiology , CD40 Ligand/blood , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/immunology , Endothelium, Vascular/physiopathology , Platelet Membrane Glycoproteins/physiology , Receptors, Collagen/blood , Receptors, IgG/physiology , Blood Platelets/immunology , Cell Adhesion Molecules/physiology , Cell Line , Chemokine CCL2/analysis , Endothelium, Vascular/immunology , Endothelium, Vascular/physiology , Flow Cytometry , Humans , Platelet Membrane Glycoproteins/genetics , Umbilical Veins
17.
FASEB J ; 18(2): 397-9, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14656994

ABSTRACT

Platelet-collagen interactions play a fundamental role in the process of arterial thrombosis. The major platelet collagen receptor is the glycoprotein VI (GPVI). Here, we determined the effects of a soluble dimeric form of GPVI on platelet adhesion in vitro and in vivo. We fused the extracellular domain of GPVI with the human immunoglobulin Fc domain. The soluble dimeric form of GPVI (GPVI-Fc) specifically bound to immobilized collagen. Binding of GPVI-Fc to collagen was inhibited competitively by soluble GPVI-Fc, but not control Fc lacking the external GPVI domain. GPVI-Fc inhibited the adhesion of CHO cells that stably express human GPVI and of platelets on collagen and attenuated thrombus formation under shear conditions in vitro. To test the effects of GPVI-Fc in vivo, arterial thrombosis was induced in the mouse carotid artery, and platelet-vessel wall interactions were visualized by intravital fluorescence microscopy. Infusion of GPVI-Fc but not of control Fc virtually abolished stable arrest and aggregation of platelets following vascular injury. Importantly, GPVI-Fc but not control Fc, was detected at areas of vascular injury. These findings further substantiate the critical role of the collagen receptor GPVI in the initiation of thrombus formation at sites of vascular injury and identify soluble GPVI as a promising antithrombotic strategy.


Subject(s)
Blood Platelets/physiology , Carotid Arteries/metabolism , Carotid Arteries/pathology , Platelet Adhesiveness , Platelet Aggregation , Platelet Membrane Glycoproteins/chemistry , Platelet Membrane Glycoproteins/metabolism , Animals , Bleeding Time , CHO Cells , Cloning, Molecular , Collagen/metabolism , Cricetinae , Dimerization , Humans , Immunoglobulin Fc Fragments/chemistry , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/metabolism , Mice , Platelet Count , Platelet Membrane Glycoproteins/genetics , Platelet Membrane Glycoproteins/isolation & purification , Protein Binding , Rheology , Solubility , Thrombosis
18.
J Gen Appl Microbiol ; 44(5): 317-326, 1998 Oct.
Article in English | MEDLINE | ID: mdl-12501411

ABSTRACT

According to sequence analysis, the spoIV-locus of Bacillus megaterium DSM 319 is 1,185 bp long; it is the second gene of a sporulation operon, which altogether contains three open reading frames. The ORF preceding spoIV encodes a putative polypeptide with 94 amino acids; the 3rd ORF of the operon has 972 bp corresponding to 324 amino acids. The operon is flanked on both sides by palindromic sequences, probably representing Rho-independent terminators. A primer extension analysis revealed that mRNA synthesis starts immediately downstream of a promoter, which is similar to the consensus sequence of Bacillus subtilis sigma(E) dependent promoters. Both the -35 and the -10 region are within the terminator region of the preceding operon. Gene knockout experiments and reporter gene assays with a newly developed system based on the heterologous Paenibacillus macerans glucanase gene (bgl) confirmed sigma(E)-dependent transcription. Two open reading frames of a further upstream operon were also identified. Northern analysis revealed that transcription of these ORFs comes about in late sporulation phases. The genetic organization of the spoIV comprising operon and adjacent loci clearly resembles that of the B. subtilis yqfa-phoH gene cluster. Thus our findings are of general significance for endospore-forming bacteria.

SELECTION OF CITATIONS
SEARCH DETAIL
...