Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Pharmaceutics ; 13(9)2021 Sep 18.
Article in English | MEDLINE | ID: mdl-34575580

ABSTRACT

Medical devices directly exposed to blood are commonly used to treat cardiovascular diseases. However, these devices are associated with inflammatory reactions leading to delayed healing, rejection of foreign material or device-associated thrombus formation. We developed a novel recombinant fusion protein as a new biocompatible coating strategy for medical devices with direct blood contact. We genetically fused human serum albumin (HSA) with ectonucleoside triphosphate diphosphohydrolase-1 (CD39), a promising anti-thrombotic and anti-inflammatory drug candidate. The HSA-CD39 fusion protein is highly functional in degrading ATP and ADP, major pro-inflammatory reagents and platelet agonists. Their enzymatic properties result in the generation of AMP, which is further degraded by CD73 to adenosine, an anti-inflammatory and anti-platelet reagent. HSA-CD39 is functional after lyophilisation, coating and storage of coated materials for up to 8 weeks. HSA-CD39 coating shows promising and stable functionality even after sterilisation and does not hinder endothelialisation of primary human endothelial cells. It shows a high level of haemocompatibility and diminished blood cell adhesion when coated on nitinol stents or polyvinylchloride tubes. In conclusion, we developed a new recombinant fusion protein combining HSA and CD39, and demonstrated that it has potential to reduce thrombotic and inflammatory complications often associated with medical devices directly exposed to blood.

2.
Crit Care Med ; 47(5): e420-e427, 2019 05.
Article in English | MEDLINE | ID: mdl-30730441

ABSTRACT

OBJECTIVES: Sepsis is associated with a systemic inflammatory reaction, which can result in a life-endangering organ dysfunction. Pro-inflammatory responses during sepsis are characterized by increased activation of leukocytes and platelets, formation of platelet-neutrophil aggregates, and cytokine production. Sequestration of platelet-neutrophil aggregates in the microvasculature contributes to tissue damage during sepsis. At present no effective therapeutic strategy to ameliorate these events is available. In this preclinical pilot study, a novel anti-inflammatory approach was evaluated, which targets nucleoside triphosphate hydrolase activity toward activated platelets via a recombinant fusion protein combining a single-chain antibody against activated glycoprotein IIb/IIIa and the extracellular domain of CD39 (targ-CD39). DESIGN: Experimental animal study and cell culture study. SETTING: University-based experimental laboratory. SUBJECTS: Human dermal microvascular endothelial cells 1, human platelets and neutrophils, and C57BL/6NCrl mice. INTERVENTIONS: Platelet-leukocyte-endothelium interactions were evaluated under inflammatory conditions in vitro and in a murine lipopolysaccharide-induced sepsis model in vivo. The outcome of polymicrobial sepsis was evaluated in a murine cecal ligation and puncture model. To evaluate the anti-inflammatory potential of activated platelet targeted nucleoside triphosphate hydrolase activity, we employed a potato apyrase in vitro and in vivo, as well as targ-CD39 and as a control, nontarg-CD39 in vivo. MEASUREMENTS AND MAIN RESULTS: Under conditions of sepsis, agents with nucleoside triphosphate hydrolase activity decreased platelet-leukocyte-endothelium interaction, transcription of pro-inflammatory cytokines, microvascular platelet-neutrophil aggregate sequestration, activation marker expression on platelets and neutrophils contained in these aggregates, leukocyte extravasation, and organ damage. Targ-CD39 had the strongest effect on these variables and retained hemostasis in contrast to nontarg-CD39 and potato apyrase. Most importantly, targ-CD39 improved survival in the cecal ligation and puncture model to a stronger extent then nontarg-CD39 and potato apyrase. CONCLUSIONS: Targeting nucleoside triphosphate hydrolase activity (CD39) toward activated platelets is a promising new treatment concept to decrease systemic inflammation and mortality of sepsis. This innovative therapeutic approach warrants further development toward clinical application.


Subject(s)
Blood Platelets/metabolism , Endothelial Cells/metabolism , Sepsis/immunology , Adenosine Triphosphatases/pharmacology , Animals , Blood Platelets/drug effects , Cytokines/metabolism , Endothelial Cells/drug effects , Humans , Mice , Mice, Inbred C57BL , Neutrophils/metabolism , Pilot Projects
3.
Mol Ther ; 26(4): 1056-1065, 2018 04 04.
Article in English | MEDLINE | ID: mdl-29525742

ABSTRACT

Abdominal aortic aneurysm (AAA) is an often deadly disease without medical, non-invasive treatment options. The upregulation of vascular cell adhesion molecule-1 (VCAM-1) on aortic endothelium provides an early target epitope for a novel biotechnological theranostic approach. MicroRNA-126 was used as a therapeutic agent, based on its capability to downregulate VCAM-1 expression in endothelial cells and thereby reduces leukocyte adhesion and exerts anti-inflammatory effects. Ultrasound microbubbles were chosen as carriers, allowing both molecular imaging as well as targeted therapy of AAA. Microbubbles were coupled with a VCAM-1-targeted single-chain antibody (scFvmVCAM-1) and a microRNA-126 mimic (M126) constituting theranostic microbubbles (TargMB-M126). TargMB-M126 downregulates VCAM-1 expression in vitro and in an in vivo acute inflammatory murine model. Most importantly, using TargMB-M126 and ultrasound-guided burst delivery of M126, the development of AAA in an angiotensin-II-induced mouse model can be prevented. Overall, we describe a unique biotechnological theranostic approach with the potential for early diagnosis and long-sought-after medical therapy of AAA.


Subject(s)
Aortic Aneurysm, Abdominal/genetics , Aortic Aneurysm, Abdominal/pathology , Genetic Therapy , MicroRNAs/genetics , Animals , Aortic Aneurysm, Abdominal/metabolism , Aortic Aneurysm, Abdominal/therapy , Biomarkers , Cells, Cultured , Disease Models, Animal , Endothelial Cells , Gene Transfer Techniques , Genetic Therapy/methods , Immunohistochemistry , Male , Mice , Mice, Knockout , MicroRNAs/administration & dosage , MicroRNAs/chemistry , Molecular Imaging , Single-Chain Antibodies/pharmacology , Ultrasonography , Vascular Cell Adhesion Molecule-1/antagonists & inhibitors , Vascular Cell Adhesion Molecule-1/chemistry , Vascular Cell Adhesion Molecule-1/metabolism
4.
Nat Commun ; 9(1): 525, 2018 02 06.
Article in English | MEDLINE | ID: mdl-29410422

ABSTRACT

Integrin-based therapeutics have garnered considerable interest in the medical treatment of inflammation. Integrins mediate the fast recruitment of monocytes and neutrophils to the site of inflammation, but are also required for host defense, limiting their therapeutic use. Here, we report a novel monoclonal antibody, anti-M7, that specifically blocks the interaction of the integrin Mac-1 with its pro-inflammatory ligand CD40L, while not interfering with alternative ligands. Anti-M7 selectively reduces leukocyte recruitment in vitro and in vivo. In contrast, conventional anti-Mac-1 therapy is not specific and blocks a broad repertoire of integrin functionality, inhibits phagocytosis, promotes apoptosis, and fuels a cytokine storm in vivo. Whereas conventional anti-integrin therapy potentiates bacterial sepsis, bacteremia, and mortality, a ligand-specific intervention with anti-M7 is protective. These findings deepen our understanding of ligand-specific integrin functions and open a path for a new field of ligand-targeted anti-integrin therapy to prevent inflammatory conditions.


Subject(s)
Antibodies, Monoclonal/pharmacology , Inflammation/drug therapy , Macrophage-1 Antigen/metabolism , Molecular Targeted Therapy/methods , Animals , Binding Sites , CD40 Ligand/metabolism , Host-Pathogen Interactions/drug effects , Humans , Inflammation/pathology , Leukocytes/drug effects , Leukocytes/pathology , Male , Mice, Inbred C57BL , Neutrophils/drug effects , Sepsis/drug therapy
5.
Eur Heart J ; 39(2): 111-116, 2018 01 07.
Article in English | MEDLINE | ID: mdl-28472483

ABSTRACT

Aims: CD39 is a cell membrane NTPase with anti-inflammatory and anti-platelet effects. However, its clinical use is limited by its bleeding side effect. With the goal of harnessing its therapeutic potential while avoiding haemostatic problems, we designed a fusion protein consisting of the extracellular domain of CD39 and a single-chain antibody (Targ-CD39) that specifically binds to activated glycoprotein (GP)IIb/IIIa and thus to activated platelets. Through this enrichment at activated platelets, the required systemic dose is below the dose impairing haemostasis. Methods and results: Using an ischaemia/reperfusion mouse model (left anterior descending artery ligated for 1 h) we achieved remarkable protection of the reperfused tissue with Targ-CD39 compared with Non-targ-CD39 (mutated, non-binding version of Targ-CD39) and PBS control. Targ-CD39 restored ejection fraction and fractional shortening to a level indistinguishable from pre-injury status, while controls showed functional deterioration. Employing advanced clinically relevant methods of ultrasound analysis, we observed that both radial and longitudinal strain and strain rate showed infarct-typical changes of myocardial deformation in controls, but not in Targ-CD39 treated mice. Histological assessment confirmed strong reduction of infarct size and increase in neovascularization. Furthermore, attenuation of post-ischaemic inflammation was seen in cytokine profiling. Conclusion: Overall, we demonstrate that Targ-CD39 holds promise for treatment of myocardial infarction.


Subject(s)
Adenosine Triphosphatases/therapeutic use , Antibodies/therapeutic use , Myocardial Reperfusion Injury/prevention & control , Recombinant Fusion Proteins/therapeutic use , Adenosine Triphosphatases/pharmacology , Animals , Antibodies/pharmacology , Disease Models, Animal , Mice , Mice, Inbred C57BL , Myocardial Contraction/drug effects , Myocardial Infarction/blood , Myocardial Infarction/pathology , Myocardial Infarction/prevention & control , Myocardial Reperfusion Injury/blood , Myocardial Reperfusion Injury/pathology , Platelet Activation/drug effects , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Protein Transport , Recombinant Fusion Proteins/pharmacology
6.
Theranostics ; 7(10): 2565-2574, 2017.
Article in English | MEDLINE | ID: mdl-28819447

ABSTRACT

Rationale The early detection of primary tumours and metastatic disease is vital for successful therapy and is contingent upon highly specific molecular markers and sensitive, non-invasive imaging techniques. We hypothesized that the accumulation of activated platelets within tumours is a general phenomenon and thus represents a novel means for the molecular imaging of cancer. Here we investigate a unique single chain antibody (scFv), which specifically targets activated platelets, as a novel biotechnological tool for molecular imaging of cancer. Methods The scFvGPIIb/IIIa, which binds specifically to the activated form of the platelet integrin receptor GPIIb/IIIa present on activated platelets, was conjugated to either Cy7, 64Cu or ultrasound-enhancing microbubbles. Using the Cy7 labelled scFvGPIIb/IIIa, fluorescence imaging was performed in mice bearing four different human tumour xenograft models; SKBr3, MDA-MB-231, Ramos and HT-1080 cells. Molecular imaging via PET and ultrasound was performed using the scFvGPIIb/IIIa-64Cu and scFvGPIIb/IIIa-microbubbles, respectively, to further confirm specific targeting of scFvGPIIb/IIIa to activated platelets in the tumour stroma. Results Using scFvGPIIb/IIIa we successfully showed specific targeting of activated platelets within the microenvironment of human tumour xenografts models via three different molecular imaging modalities. The presence of platelets within the tumour microenvironment, and as such their relevance as a molecular target epitope in cancer was further confirmed via immunofluorescence of human tumour sections of various cancer types, thus validating the translational importance of our novel approach to human disease. Conclusion Our study provides proof of concept for imaging and localization of tumours by molecular targeting activated platelets. We illustrate the utility of a unique scFv as a versatile biotechnological tool which can be conjugated to various contrast agents for molecular imaging of cancer using three different imaging modalities. These findings warrant further development of this activated platelet specific scFvGPIIb/IIIa, potentially as a universal marker for cancer diagnosis and ultimately for drug delivery in an innovative theranostic approach.


Subject(s)
Blood Platelets/chemistry , Molecular Imaging/methods , Neoplasms/diagnostic imaging , Neoplasms/pathology , Optical Imaging/methods , Platelet Activation , Animals , Disease Models, Animal , Heterografts , Humans , Mice , Neoplasm Transplantation , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Single-Chain Antibodies/metabolism
7.
Theranostics ; 6(5): 726-38, 2016.
Article in English | MEDLINE | ID: mdl-27022419

ABSTRACT

RATIONALE: Myocardial infarction and stroke are leading causes of morbidity/mortality. The typical underlying pathology is the formation of thrombi/emboli and subsequent vessel occlusion. Systemically administered fibrinolytic drugs are the most effective pharmacological therapy. However, bleeding complications are relatively common and this risk as such limits their broader use. Furthermore, a rapid non-invasive imaging technology is not available. Thereby, many thrombotic events are missed or only diagnosed when ischemic damage has already occurred. OBJECTIVE: Design and preclinical testing of a novel 'theranostic' technology for the rapid non-invasive diagnosis and effective, bleeding-free treatment of thrombosis. METHODS AND RESULTS: A newly created, innovative theranostic microbubble combines a recombinant fibrinolytic drug, an echo-enhancing microbubble and a recombinant thrombus-targeting device in form of an activated-platelet-specific single-chain antibody. After initial in vitro proof of functionality, we tested this theranostic microbubble both in ultrasound imaging and thrombolytic therapy using a mouse model of ferric-chloride-induced thrombosis in the carotid artery. We demonstrate the reliable highly sensitive detection of in vivo thrombi and the ability to monitor their size changes in real time. Furthermore, these theranostic microbubbles proofed to be as effective in thrombolysis as commercial urokinase but without the prolongation of bleeding time as seen with urokinase. CONCLUSIONS: We describe a novel theranostic technology enabling simultaneous diagnosis and treatment of thrombosis, as well as monitoring of success or failure of thrombolysis. This technology holds promise for major progress in rapid diagnosis and bleeding-free thrombolysis thereby potentially preventing the often devastating consequences of thrombotic disease in many patients.


Subject(s)
Microbubbles , Theranostic Nanomedicine/methods , Thrombolytic Therapy/methods , Thrombosis/diagnostic imaging , Thrombosis/therapy , Ultrasonography/methods , Animals , Bleeding Time , Blood Platelets/immunology , Disease Models, Animal , Fibrinolytic Agents/administration & dosage , Fibrinolytic Agents/pharmacokinetics , Mice , Single-Chain Antibodies/immunology , Single-Chain Antibodies/pharmacokinetics , Treatment Outcome
8.
PLoS One ; 10(9): e0138375, 2015.
Article in English | MEDLINE | ID: mdl-26381750

ABSTRACT

BACKGROUND: Stent angioplasty provides a minimally invasive treatment for atherosclerotic vessels. However, no treatment option for atherosclerosis-associated endothelial dysfunction, which is accompanied by a loss of CD39, is available, and hence, adverse effects like thromboembolism and restenosis may occur. Messenger RNA (mRNA)-based therapy represents a novel strategy, whereby de novo synthesis of a desired protein is achieved after delivery of a modified mRNA to the target cells. METHODS AND FINDINGS: Our study aimed to develop an innovative bioactive stent coating that induces overexpression of CD39 in the atherosclerotic vessel. Therefore, a modified CD39-encoding mRNA was produced by in vitro transcription. Different endothelial cells (ECs) were transfected with the mRNA, and CD39 expression and functionality were analyzed using various assays. Furthermore, CD39 mRNA was immobilized using poly(lactic-co-glycolic-acid) (PLGA), and the transfection efficiency in ECs was analyzed. Our data show that ECs successfully translate in vitro-generated CD39 mRNA after transfection. The overexpressed CD39 protein is highly functional in hydrolyzing ADP and in preventing platelet activation. Furthermore, PLGA-immobilized CD39 mRNA can be delivered to ECs without losing its functionality. SUMMARY: In summary, we present a novel and promising concept for a stent coating for the treatment of atherosclerotic blood vessels, whereby patients could be protected against angioplasty-associated complications.


Subject(s)
Angioplasty/methods , Antigens, CD/genetics , Apyrase/genetics , Atherosclerosis/surgery , RNA, Messenger/therapeutic use , Stents/adverse effects , Angioplasty/adverse effects , Animals , Antigens, CD/metabolism , Apyrase/metabolism , Cell Line , Endothelial Cells/metabolism , HEK293 Cells , Humans , Swine
9.
Circ Res ; 114(7): 1083-93, 2014 Mar 28.
Article in English | MEDLINE | ID: mdl-24508759

ABSTRACT

RATIONALE: Fibrinolysis is a valuable alternative for the treatment of myocardial infarction when percutaneous coronary intervention is not available in a timely fashion. For acute ischemic stroke, fibrinolysis is the only treatment option with a very narrow therapeutic window. Clinically approved thrombolytics have significant drawbacks, including bleeding complications. Thus their use is highly restricted, leaving many patients untreated. OBJECTIVE: We developed a novel targeted fibrinolytic drug that is directed against activated platelets. METHODS AND RESULTS: We fused single-chain urokinase plasminogen activator (scuPA) to a small recombinant antibody (scFvSCE5), which targets the activated form of the platelet-integrin glycoprotein IIb/IIIa. Antibody binding and scuPA activity of this recombinant fusion protein were on par with the parent molecules. Prophylactic in vivo administration of scFvSCE5-scuPA (75 U/g body weight) prevented carotid artery occlusion after ferric chloride injury in a plasminogen-dependent process compared with saline (P<0.001), and blood flow recovery was similar to high-dose nontargeted urokinase (500 U/g body weight). Tail bleeding time was significantly prolonged with this high dose of nontargeted urokinase, but not with equally effective targeted scFvSCE5-scuPA at 75 U/g body weight. Real-time in vivo molecular ultrasound imaging demonstrates significant therapeutic reduction of thrombus size after administration of 75 U/g body weight scFvSCE5-scuPA as compared with the same dose of a mutated, nontargeting scFv-scuPA or vehicle. The ability of scFvSCE5-scuPA to lyse thrombi was lost in plasminogen-deficient mice, but could be restored by intravenous injection of plasminogen. CONCLUSIONS: Targeting of scuPA to activated glycoprotein IIb/IIIa allows effective thrombolysis and the potential novel use as a fibrinolytic agent for thromboprophylaxis without bleeding complications.


Subject(s)
Blood Platelets/drug effects , Carotid Arteries/diagnostic imaging , Fibrinolytic Agents/therapeutic use , Single-Chain Antibodies/therapeutic use , Thromboembolism/drug therapy , Urokinase-Type Plasminogen Activator/therapeutic use , Animals , Blood Platelets/immunology , CHO Cells , Cricetinae , Cricetulus , Drug Evaluation, Preclinical , Fibrinolytic Agents/adverse effects , Integrin alpha2/immunology , Mice , Mice, Inbred C57BL , Plasminogen/metabolism , Platelet Activation , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/therapeutic use , Single-Chain Antibodies/genetics , Single-Chain Antibodies/immunology , Thromboembolism/prevention & control , Thrombolytic Therapy , Ultrasonography , Urokinase-Type Plasminogen Activator/genetics
10.
Blood ; 121(16): 3067-75, 2013 Apr 18.
Article in English | MEDLINE | ID: mdl-23380744

ABSTRACT

The ecto-nucleoside triphosphate diphosphohydrolase CD39 represents a promising antithrombotic therapeutic. It degrades adenosine 5'-diphosphate (ADP), a main platelet activating/recruiting agent. We hypothesized that delayed enrichment of CD39 on developing thrombi will allow for a low and safe systemic concentration and thus avoid bleeding. We use a single-chain antibody (scFv, specific for activated GPIIb/IIIa) for targeting CD39. This should allow delayed enrichment on growing thrombi but not on the initial sealing layer of platelets, which do not yet express activated GPIIb/IIIa. CD39 was recombinantly fused to an activated GPIIb/IIIa-specific scFv (targ-CD39) and a nonfunctional scFv (non-targ-CD39). Targ-CD39 was more effective at preventing ADP-induced platelet activation than non-targ-CD39. In a mouse carotid artery thrombosis model, non-targ-CD39, although protective against vessel occlusion, was associated with significant bleeding on tail transection. In contrast, targ-CD39 concentrated at the thrombus site; hence, a dose ∼10 times less of CD39 prevented vessel occlusion to a similar extent as high-dose non-targ-CD39, without prolonged bleeding time. An equimolar dose of non-targ-CD39 at this low concentration was ineffective at preventing vessel occlusion. Thus, delayed targeting of CD39 via scFv to activated platelets provides strong antithrombotic potency and yet prevents bleeding and thereby promotes CD39 toward clinical use.


Subject(s)
Antigens, CD/therapeutic use , Apyrase/therapeutic use , Fibrinolytic Agents/therapeutic use , Platelet Activation/drug effects , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Recombinant Fusion Proteins/therapeutic use , Single-Chain Antibodies/therapeutic use , Thrombosis/drug therapy , Adenosine Diphosphate/metabolism , Animals , Antigens, CD/genetics , Apyrase/genetics , Blood Platelets/drug effects , Blood Platelets/pathology , Drug Delivery Systems , Fibrinolytic Agents/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Recombinant Fusion Proteins/genetics , Single-Chain Antibodies/genetics , Thrombosis/metabolism , Thrombosis/pathology
11.
Circulation ; 125(25): 3117-26, 2012 Jun 26.
Article in English | MEDLINE | ID: mdl-22647975

ABSTRACT

BACKGROUND: Molecular imaging is a fast emerging technology allowing noninvasive detection of vascular pathologies. However, imaging modalities offering high resolution currently do not allow real-time imaging. We hypothesized that contrast-enhanced ultrasound with microbubbles (MBs) selectively targeted to activated platelets would offer high-resolution, real-time molecular imaging of evolving and dissolving arterial thrombi. METHODS AND RESULTS: Lipid-shell based gas-filled MBs were conjugated to either a single-chain antibody specific for activated glycoprotein IIb/IIIa via binding to a Ligand-Induced Binding Site (LIBS-MBs) or a nonspecific single-chain antibody (control MBs). Successful conjugation was assessed in flow cytometry and immunofluorescence double staining. LIBS-MBs but not control MBs strongly adhered to both immobilized activated platelets and microthrombi under flow. Thrombi induced in carotid arteries of C57Bl6 mice in vivo by ferric chloride injury were then assessed with ultrasound before and 20 minutes after MB injection through the use of gray-scale area intensity measurement. Gray-scale units converted to decibels demonstrated a significant increase after LIBS-MB but not after control MB injection (9.55±1.7 versus 1.46±1.3 dB; P<0.01). Furthermore, after thrombolysis with urokinase, LIBS-MB ultrasound imaging allows monitoring of the reduction of thrombus size (P<0.001). CONCLUSION: We demonstrate that glycoprotein IIb/IIIa-targeted MBs specifically bind to activated platelets in vitro and allow real-time molecular imaging of acute arterial thrombosis and monitoring of the success or failure of pharmacological thrombolysis in vivo.


Subject(s)
Binding Sites, Antibody , Drug Delivery Systems/methods , Microbubbles , Platelet Activation , Single-Chain Antibodies , Thrombolytic Therapy , Thrombosis/diagnostic imaging , Thrombosis/diagnosis , Animals , Binding Sites, Antibody/immunology , Disease Models, Animal , Integrin beta3/immunology , Integrin beta3/metabolism , Mice , Mice, Inbred C57BL , Molecular Imaging/methods , Molecular Imaging/trends , Platelet Activation/immunology , Platelet Membrane Glycoprotein IIb/immunology , Platelet Membrane Glycoprotein IIb/metabolism , Single-Chain Antibodies/metabolism , Thrombolytic Therapy/methods , Thrombolytic Therapy/trends , Thrombosis/metabolism , Time Factors , Treatment Failure , Treatment Outcome , Ultrasonography
12.
Circ Res ; 109(11): 1269-79, 2011 Nov 11.
Article in English | MEDLINE | ID: mdl-21998326

ABSTRACT

RATIONALE: CD40L figures prominently in chronic inflammatory diseases such as atherosclerosis. However, since CD40L potently regulates immune function and hemostasis by interaction with CD40 receptor and the platelet integrin GPIIb/IIIa, its global inhibition compromises host defense and generated thromboembolic complications in clinical trials. We recently reported that CD40L mediates atherogenesis independently of CD40 and proposed Mac-1 as an alternate receptor. OBJECTIVE: Here, we molecularly characterized the CD40L-Mac-1 interaction and tested whether its selective inhibition by a small peptide modulates inflammation and atherogenesis in vivo. METHODS AND RESULTS: CD40L concentration-dependently bound to Mac-1 I-domain in solid phase binding assays, and a high-affinity interaction was revealed by surface-plasmon-resonance analysis. We identified the motif EQLKKSKTL, an exposed loop between the α1 helix and the ß-sheet B, on Mac-1 as binding site for CD40L. A linear peptide mimicking this sequence, M7, specifically inhibited the interaction of CD40L and Mac-1. A cyclisized version optimized for in vivo use, cM7, decreased peritoneal inflammation and inflammatory cell recruitment in vivo. Finally, LDLr(-/-) mice treated with intraperitoneal injections of cM7 developed smaller, less inflamed atherosclerotic lesions featuring characteristics of stability. However, cM7 did not interfere with CD40L-CD40 binding in vitro and CD40L-GPIIb/IIIa-mediated thrombus formation in vivo. CONCLUSIONS: We present the novel finding that CD40L binds to the EQLKKSKTL motif on Mac-1 mediating leukocyte recruitment and atherogenesis. Specific inhibition of CD40L-Mac-1 binding may represent an attractive anti-inflammatory treatment strategy for atherosclerosis and other inflammatory conditions, potentially avoiding the unwanted immunologic and thrombotic effects of global inhibition of CD40L.


Subject(s)
Atherosclerosis/metabolism , CD40 Ligand/metabolism , Chemotaxis, Leukocyte/physiology , Macrophage-1 Antigen/metabolism , Thrombosis/etiology , Amino Acid Motifs , Animals , Atherosclerosis/genetics , Atherosclerosis/prevention & control , Bleeding Time , Blood Coagulation/drug effects , Blood Coagulation/physiology , CHO Cells , Cells, Cultured , Cricetinae , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Molecular , Peptide Fragments/pharmacology , Peptide Fragments/therapeutic use , Peptides, Cyclic/pharmacology , Peritonitis/blood , Peritonitis/prevention & control , Protein Conformation , Protein Interaction Mapping , Protein Structure, Tertiary , Receptors, LDL/deficiency , Recombinant Fusion Proteins/physiology , Surface Plasmon Resonance
13.
Arterioscler Thromb Vasc Biol ; 31(9): 2015-23, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21659646

ABSTRACT

OBJECTIVE: Therapeutic hypothermia is successfully used, for example, in cardiac surgery to protect organs from ischemia. Cardiosurgical procedures, especially in combination with extracorporeal circulation, and hypothermia itself are potentially prothrombotic. Despite the obvious need, the long half-life of antiplatelet drugs and thus the risk of postoperative bleedings have restricted their use in cardiac surgery. We describe here the design and testing of a unique recombinant hypothermia-controlled antiplatelet fusion protein with the aim of providing increased safety of hypothermia, as well as cardiac surgery. METHODS AND RESULTS: An elastin-mimetic polypeptide was fused to an activation-specific glycoprotein (GP) IIb/IIIa-blocking single-chain antibody. In silico modeling illustrated the sterical hindrance of a ß-spiral conformation of elastin-mimetic polypeptide preventing the single-chain antibody from inhibiting GPIIb/IIIa at 37°C. Circular dichroism spectra demonstrated reverse temperature transition, and flow cytometry showed binding to and blocking of GPIIb/IIIa at hypothermic body temperature (≤32°C) but not at normal body temperature. In vivo thrombosis in mice was selectively inhibited at hypothermia but not at 37°C. CONCLUSIONS: This is the first description of a broadly applicable pharmacological strategy by which the activity of a potential drug can be controlled by temperature. In particular, this drug steerability may provide substantial benefits for antiplatelet therapy.


Subject(s)
Hypothermia, Induced , Platelet Aggregation Inhibitors/administration & dosage , Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors , Recombinant Fusion Proteins/administration & dosage , Tropoelastin/administration & dosage , Animals , Circular Dichroism , Coronary Artery Bypass , Fibrinogen/metabolism , Humans , Mice , Models, Molecular , Platelet Aggregation , Protein Conformation , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Single-Chain Antibodies/administration & dosage , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/metabolism
14.
Arterioscler Thromb Vasc Biol ; 31(7): 1607-16, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21512161

ABSTRACT

OBJECTIVE: Hypothermia is used in various clinical settings to inhibit ischemia-related organ damage. However, prothrombotic effects have been described as potential side effects. This study aimed to elucidate the mechanism of hypothermia-induced platelet activation and subsequent prothrombotic events and to develop preventative pharmacological strategies applicable during clinically used hypothermia. METHODS AND RESULTS: Platelet function was investigated ex vivo and in vivo at clinically used hypothermia (28°C/18°C). Hypothermic mice demonstrated increased expression of platelet activation marker P-selectin, platelet-leukocyte aggregate formation, and thrombocytopenia. Intravital microscopy of FeCl(3)-injured murine mesenteric arteries revealed increased platelet thrombus formation with hypothermia. Ex vivo flow chamber experiments indicated increased platelet-fibrinogen adhesion under hypothermia. We show that hypothermia results in reduced ADP hydrolysis via reduction of CD39 (E-NTPDase1) activity, resulting in increased levels of ADP and subsequent augmented primary and secondary platelet activation. In vivo administration of ADP receptor P(2)Y(12) antagonists and recombinant soluble CD39 prevented hypothermia-induced thrombus formation and thrombocytopenia, respectively. CONCLUSIONS: The platelet agonist ADP plays a key role in hypothermia-induced platelet activation. Inhibition of receptor binding or hydrolysis of ADP has the potential to protect platelets against hypothermia-induced activation. Our findings provide a rational basis for further evaluation of novel antithrombotic strategies in clinically applied hypothermia.


Subject(s)
Adenosine Diphosphate/blood , Blood Platelets/drug effects , Fibrinolytic Agents/pharmacology , Hypothermia, Induced , Platelet Activation/drug effects , Platelet Aggregation Inhibitors/pharmacology , Thrombosis/prevention & control , Analysis of Variance , Animals , Antigens, CD/blood , Antigens, CD/pharmacology , Apyrase/blood , Apyrase/pharmacology , Blood Platelets/metabolism , Fibrinogen/metabolism , Humans , Hydrolysis , Hypothermia, Induced/adverse effects , Leukopenia/blood , Leukopenia/etiology , Membrane Glycoproteins/blood , Mice , Mice, Inbred C57BL , P-Selectin/blood , Platelet Adhesiveness/drug effects , Platelet Glycoprotein GPIb-IX Complex , Purinergic P2Y Receptor Antagonists/pharmacology , Receptors, Purinergic P2Y1/blood , Receptors, Purinergic P2Y1/drug effects , Receptors, Purinergic P2Y12/blood , Receptors, Purinergic P2Y12/drug effects , Recombinant Proteins/pharmacology , Thrombocytopenia/blood , Thrombocytopenia/etiology , Thrombosis/blood , Thrombosis/etiology , von Willebrand Factor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...