Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Marc W. Nolte is active.

Publication


Featured researches published by Marc W. Nolte.


Science Translational Medicine | 2014

A factor XIIa inhibitory antibody provides thromboprotection in extracorporeal circulation without increasing bleeding risk.

Magnus Larsson; Veronika Rayzman; Marc W. Nolte; Katrin F. Nickel; Jenny Björkqvist; Anne Jämsä; Matthew P. Hardy; Marion Fries; Stefan Schmidbauer; Patricia Hedenqvist; Michael Broomé; Ingo Pragst; Gerhard Dickneite; Michael Wilson; Andrew D. Nash; Con Panousis; Thomas Renné

Blocking the enzyme that initiates the intrinsic coagulation pathway protects against thrombosis in bypass systems and does not cause excess bleeding in vivo. When Life-Saving Is Life-Threatening We all need a vacation sometimes. For the heart and lungs, that time can come during surgery (such as cardiopulmonary bypass procedures), in instances of organ failure (for example, in septic patients), or while awaiting a replacement organ for transplantation. When the heart and lungs take time off, oxygenation of the blood needs to occur outside of the body by circulation through a cardiopulmonary bypass system (also called a heart-lung machine). In order to prevent blood clot formation in the extracorporeal circuit, an anticoagulant is added to the system. Anticoagulants block thrombus formation that would occlude the circulation; however, the drugs also interfere with the body’s ability to stop bleeding at the site of injury. Thus, an ideal anticoagulant would only block blood clotting in thrombosis without causing excess bleeding. Now, Larsson et al. describe a new antibody that prevents thrombosis and facilitates blood flow in a specific heart-lung machine without causing bleeding in large animal models. The anticoagulant heparin is used most often during extracorporeal oxygenation and targets multiple components of the blood coagulation cascade that are necessary formation of fibrin—a clotting protein essential for stemming injury-related blood loss. The authors used phage display to identify an antibody that binds to and inhibits the protease activity of factor XIIa (FXIIa), a protein that controls fibrin formation in vitro but does not appear to be required for cessation of bleeding from injury sites. A fully humanized version of the antibody, called 3F7, protected against pathological thrombosis in the extracorporeal bypass system without increasing bleeding from injuries in rabbits. 3F7 had the added benefits of a broad therapeutic range and easy monitoring at the point of care. And because 3F7 doesn’t cause bleeding, it should not require neutralization after surgery and can simply be cleared from the patient’s circulation naturally. Even with optimal heparin treatment, bleeding remains the most common complication of anticoagulation therapy. Additional mechanistic and clinical studies will show whether 3F7—or an optimized version—will be able to give heparin a vacation from facilitating extracorporeal circulation and possibly other scenarios that require safe anticoagulation. Currently used anticoagulants prevent thrombosis but increase bleeding. We show an anticoagulation therapy without bleeding risk based on a plasma protease factor XII function-neutralizing antibody. We screened for antibodies against activated factor XII (FXIIa) using phage display and demonstrated that recombinant fully human antibody 3F7 binds into the FXIIa enzymatic pocket. 3F7 interfered with FXIIa-mediated coagulation, abolished thrombus formation under flow, and blocked experimental thrombosis in mice and rabbits. We adapted an extracorporeal membrane oxygenation (ECMO) cardiopulmonary bypass system used for infant therapy to analyze clinical applicability of 3F7 in rabbits. 3F7 provided thromboprotection as efficiently as heparin, and both drugs prevented fibrin deposition and thrombosis within the extracorporeal circuit. Unlike heparin, 3F7 treatment did not impair the hemostatic capacity and did not increase bleeding from wounds. These data establish that targeting of FXIIa is a safe mode of thromboprotection in bypass systems, and provide a clinically relevant anticoagulation strategy that is not complicated by excess bleeding.


The Journal of Neuroscience | 2010

Generation and Therapeutic Efficacy of Highly Oligomer-Specific β-Amyloid Antibodies

Heinz Hillen; Stefan Barghorn; Andreas Striebinger; Boris Labkovsky; Reinhold Müller; Volker Nimmrich; Marc W. Nolte; Claudia Perez-Cruz; Ingrid Van der Auwera; Fred Van Leuven; Marcel Van Gaalen; Anton Bespalov; Hans Schoemaker; James P. Sullivan; Ulrich Ebert

Oligomers of the β-amyloid (Aβ) peptide have been indicated in early neuropathologic changes in Alzheimers disease. Here, we present a synthetic Aβ20-42 oligomer (named globulomer) with a different conformation to monomeric and fibrillar Aβ peptide, enabling the generation of highly Aβ oligomer-specific monoclonal antibodies. The globulomer-derived antibodies specifically detect oligomeric but not monomeric or fibrillar Aβ in various Aβ preparations. The globulomer-specific antibody A-887755 was able to prevent Aβ oligomer binding and dynamin cleavage in primary hippocampal neurons and to reverse globulomer-induced reduced synaptic transmission. In amyloid precursor protein (APP) transgenic mice, vaccination with Aβ globulomer and treatment with A-887755 improved novel object recognition. The cognitive improvement is likely attributable to reversing a deficit in hippocampal synaptic spine density in APP transgenic mice as observed after treatment with A-887755. Our findings demonstrate that selective reduction of Aβ oligomers by immunotherapy is sufficient to normalize cognitive behavior and synaptic deficits in APP transgenic mice.


Journal of Clinical Investigation | 2015

Defective glycosylation of coagulation factor XII underlies hereditary angioedema type III

Jenny Björkqvist; Steven de Maat; Urs Lewandrowski; Antonio Di Gennaro; Chris Oschatz; Kai Schönig; Markus M. Nöthen; Christian Drouet; Hal Braley; Marc W. Nolte; Albert Sickmann; Con Panousis; Coen Maas; Thomas Renné

Hereditary angioedema type III (HAEIII) is a rare inherited swelling disorder that is associated with point mutations in the gene encoding the plasma protease factor XII (FXII). Here, we demonstrate that HAEIII-associated mutant FXII, derived either from HAEIII patients or recombinantly produced, is defective in mucin-type Thr309-linked glycosylation. Loss of glycosylation led to increased contact-mediated autoactivation of zymogen FXII, resulting in excessive activation of the bradykinin-forming kallikrein-kinin pathway. In contrast, both FXII-driven coagulation and the ability of C1-esterase inhibitor to bind and inhibit activated FXII were not affected by the mutation. Intravital laser-scanning microscopy revealed that, compared with control animals, both F12-/- mice reconstituted with recombinant mutant forms of FXII and humanized HAEIII mouse models with inducible liver-specific expression of Thr309Lys-mutated FXII exhibited increased contact-driven microvascular leakage. An FXII-neutralizing antibody abolished bradykinin generation in HAEIII patient plasma and blunted edema in HAEIII mice. Together, the results of this study characterize the mechanism of HAEIII and establish FXII inhibition as a potential therapeutic strategy to interfere with excessive vascular leakage in HAEIII and potentially alleviate edema due to other causes.


Stroke | 2012

C1-Inhibitor Protects From Brain Ischemia-Reperfusion Injury by Combined Antiinflammatory and Antithrombotic Mechanisms

Nadine Heydenreich; Marc W. Nolte; Eva Göb; Friederike Langhauser; Marion Hofmeister; Peter Kraft; Christiane Albert-Weissenberger; Marc Brede; Csanad Varallyay; Kerstin Göbel; Sven G. Meuth; Bernhard Nieswandt; Gerhard Dickneite; Guido Stoll; Christoph Kleinschnitz

Background and Purpose— Inflammation and thrombosis are pathophysiological hallmarks of ischemic stroke still unamenable to therapeutic interventions. The contact-kinin system represents an interface between inflammatory and thrombotic circuits and is involved in stroke development. C1-inhibitor counteracts activation of the contact-kinin system at multiple levels. We investigated the therapeutic potential of C1-inhibitor in models of ischemic stroke. Methods— Male and female C57Bl/6 mice and rats of different ages were subjected to middle cerebral artery occlusion and treated with C1-inhibitor after 1 hour or 6 hours. Infarct volumes and functional outcomes were assessed between day 1 and day 7, and findings were validated by magnetic resonance imaging. Blood–brain barrier damage, thrombus formation, and the local inflammatory response were determined poststroke. Results— Treatment with 15.0 U C1-inhibitor, but not 7.5 U, 1 hour after stroke reduced infarct volumes by ≈60% and improved clinical scores in mice of either sex on day 1. This protective effect was preserved at later stages of infarction as well as in elderly mice and in another species, ie, rats. Delayed C1-inhibitor treatment still improved clinical outcome. Blood–brain barrier damage, edema formation, and inflammation were significantly lower compared with controls. Moreover, C1-inhibitor showed strong antithrombotic effects. Conclusions— C1-inhibitor is a multifaceted antiinflammatory and antithrombotic compound that protects from ischemic neurodegeneration in clinically meaningful settings.


Nature Communications | 2016

Blood coagulation factor XII drives adaptive immunity during neuroinflammation via CD87-mediated modulation of dendritic cells

Kerstin Göbel; Susann Pankratz; Chloi-Magdalini Asaridou; Alexander M. Herrmann; Stefan Bittner; Monika Merker; Tobias Ruck; Sarah Glumm; Friederike Langhauser; Peter Kraft; Thorsten F. Krug; Johanna Breuer; Martin Herold; Catharina C. Gross; Denise Beckmann; Adelheid Korb-Pap; Michael K. Schuhmann; Stefanie Kuerten; Ioannis Mitroulis; Clemens Ruppert; Marc W. Nolte; Con Panousis; Luisa Klotz; Beate E. Kehrel; Thomas Korn; Harald Langer; Thomas Pap; Bernhard Nieswandt; Heinz Wiendl; Triantafyllos Chavakis

Aberrant immune responses represent the underlying cause of central nervous system (CNS) autoimmunity, including multiple sclerosis (MS). Recent evidence implicated the crosstalk between coagulation and immunity in CNS autoimmunity. Here we identify coagulation factor XII (FXII), the initiator of the intrinsic coagulation cascade and the kallikrein–kinin system, as a specific immune cell modulator. High levels of FXII activity are present in the plasma of MS patients during relapse. Deficiency or pharmacologic blockade of FXII renders mice less susceptible to experimental autoimmune encephalomyelitis (a model of MS) and is accompanied by reduced numbers of interleukin-17A-producing T cells. Immune activation by FXII is mediated by dendritic cells in a CD87-dependent manner and involves alterations in intracellular cyclic AMP formation. Our study demonstrates that a member of the plasmatic coagulation cascade is a key mediator of autoimmunity. FXII inhibition may provide a strategy to combat MS and other immune-related disorders.


Journal of Thrombosis and Haemostasis | 2012

Improved kinetics of rIX‐FP, a recombinant fusion protein linking factor IX with albumin, in cynomolgus monkeys and hemophilia B dogs

Marc W. Nolte; Timothy C. Nichols; Jochen Mueller-Cohrs; E. P. Merricks; Ingo Pragst; Sabine Zollner; Gerhard Dickneite

Summary.  Background:  Prophylaxis of hemophilia B, at present, requires multiple infusions of human factor (F)IX concentrates per week. A FIX molecule with a prolonged half‐life has the potential to greatly improve the convenience of, and adherence to, prophylaxis.


Jacc-cardiovascular Imaging | 2012

Selective factor XIIa inhibition attenuates silent brain ischemia: application of molecular imaging targeting coagulation pathway.

John W. Chen; Jose-Luiz Figueiredo; Gregory R. Wojtkiewicz; Cory Siegel; Yoshiko Iwamoto; Dong-Eog Kim; Marc W. Nolte; Gerhard Dickneite; Ralph Weissleder; Matthias Nahrendorf

OBJECTIVES The purpose of this study was use molecular imaging targeting coagulation pathway and inflammation to better understand the pathophysiology of silent brain ischemia (SBI) and monitor the effects of factor XIIa inhibition. BACKGROUND SBI can be observed in patients who undergo invasive vascular procedures. Unlike acute stroke, the diffuse nature of SBI and its less tangible clinical symptoms make this disease difficult to diagnose and treat. METHODS We induced SBI in mice by intra-arterial injection of fluorescently labeled microbeads or fractionated clot into the carotid artery. After SBI induction, diffusion-weighted magnetic resonance imaging was performed to confirm the presence of microinfarcts in asymptomatic mice. Molecular imaging targeting the downstream factor XIII activity (single-photon emission computed tomography/computed tomography) at 3 h and myeloperoxidase activity (magnetic resonance imaging) on day 3 after SBI induction were performed, without and with the intravenous administration of a recombinant selective factor XIIa inhibitor derived from the hematophagous insect Triatoma infestans (rHA-Infestin-4). Statistical comparisons between 2 groups were evaluated by the Student t test or Mann-Whitney U test. RESULTS In SBI-induced mice, we found abnormal activation of the coagulation cascade (factor XIII activity) and increased inflammation (myeloperoxidase activity) close to where emboli lodge in the brain. rHA-Infestin-4 administration significantly reduced ischemic damage (53% to 85% reduction of infarct volume, p < 0.05) and pathological coagulation (35% to 39% reduction of factor XIII activity, p < 0.05) without increasing hemorrhagic frequency. Myeloperoxidase activity, when normalized to the infarct volume, did not significantly change with rHA-Infestin-4 treatment, suggesting that this treatment does not further decrease inflammation other than that resulting from the reduction in infarct volume. CONCLUSIONS Focal intracerebral clotting and inflammatory activity are part of the pathophysiology underlying SBI. Inhibiting factor XIIa with rHA-Infestin-4 may present a safe and effective treatment to decrease the morbidity of SBI.


PLOS ONE | 2013

C1 Esterase Inhibitor Reduces Lower Extremity Ischemia/Reperfusion Injury and Associated Lung Damage

Claudia Duehrkop; Yara Banz; Rolf Spirig; Sylvia Miescher; Marc W. Nolte; Martin O. Spycher; Richard Smith; Steven H. Sacks; Robert Rieben

Background Ischemia/reperfusion injury of lower extremities and associated lung damage may result from thrombotic occlusion, embolism, trauma, or surgical intervention with prolonged ischemia and subsequent restoration of blood flow. This clinical entity is characterized by high morbidity and mortality. Deprivation of blood supply leads to molecular and structural changes in the affected tissue. Upon reperfusion inflammatory cascades are activated causing tissue injury. We therefore tested preoperative treatment for prevention of reperfusion injury by using C1 esterase inhibitor (C1 INH). Methods and Findings Wistar rats systemically pretreated with C1 INH (n = 6), APT070 (a membrane-targeted myristoylated peptidyl construct derived from human complement receptor 1, n = 4), vehicle (n = 7), or NaCl (n = 8) were subjected to 3h hind limb ischemia and 24h reperfusion. The femoral artery was clamped and a tourniquet placed under maintenance of a venous return. C1 INH treated rats showed significantly less edema in muscle (P<0.001) and lung and improved muscle viability (P<0.001) compared to controls and APT070. C1 INH prevented up-regulation of bradykinin receptor b1 (P<0.05) and VE-cadherin (P<0.01), reduced apoptosis (P<0.001) and fibrin deposition (P<0.01) and decreased plasma levels of pro-inflammatory cytokines, whereas deposition of complement components was not significantly reduced in the reperfused muscle. Conclusions C1 INH reduced edema formation locally in reperfused muscle as well as in lung, and improved muscle viability. C1 INH did not primarily act via inhibition of the complement system, but via the kinin and coagulation cascade. APT070 did not show beneficial effects in this model, despite potent inhibition of complement activation. Taken together, C1 INH might be a promising therapy to reduce peripheral ischemia/reperfusion injury and distant lung damage in complex and prolonged surgical interventions requiring tourniquet application.


Haematologica | 2016

Coated platelets function in platelet-dependent fibrin formation via integrin αIIbβ3 and transglutaminase factor XIII

Nadine J.A. Mattheij; Frauke Swieringa; Tom G. Mastenbroek; Michelle A. Berny-Lang; Frauke May; Constance C. F. M. J. Baaten; Paola E. J. van der Meijden; Yvonne Henskens; Erik A.M. Beckers; Dennis Suylen; Marc W. Nolte; Tilman M. Hackeng; Owen J. T. McCarty; Johan W. M. Heemskerk; Judith M. E. M. Cosemans

Coated platelets, formed by collagen and thrombin activation, have been characterized in different ways: i) by the formation of a protein coat of α-granular proteins; ii) by exposure of procoagulant phosphatidylserine; or iii) by high fibrinogen binding. Yet, their functional role has remained unclear. Here we used a novel transglutaminase probe, Rhod-A14, to identify a subpopulation of platelets with a cross-linked protein coat, and compared this with other platelet subpopulations using a panel of functional assays. Platelet stimulation with convulxin/thrombin resulted in initial integrin αIIbβ3 activation, the appearance of a platelet population with high fibrinogen binding, (independently of active integrins, but dependent on the presence of thrombin) followed by phosphatidylserine exposure and binding of coagulation factors Va and Xa. A subpopulation of phosphatidylserine-exposing platelets bound Rhod-A14 both in suspension and in thrombi generated on a collagen surface. In suspension, high fibrinogen and Rhod-A14 binding were antagonized by combined inhibition of transglutaminase activity and integrin αIIbβ3. Markedly, in thrombi from mice deficient in transglutaminase factor XIII, platelet-driven fibrin formation and Rhod-A14 binding were abolished by blockage of integrin αIIbβ3. Vice versa, star-like fibrin formation from platelets of a patient with deficiency in αIIbβ3 (Glanzmann thrombasthenia) was abolished upon blockage of transglutaminase activity. We conclude that coated platelets, with initial αIIbβ3 activation and high fibrinogen binding, form a subpopulation of phosphatidylserine-exposing platelets, and function in platelet-dependent star-like fibrin fiber formation via transglutaminase factor XIII and integrin αIIbβ3.


Thrombosis Research | 2014

Biodistribution of the recombinant fusion protein linking coagulation factor IX with albumin (rIX-FP) in rats

Eva Herzog; Stephen Harris; Claire Henson; Andrew McEwen; Sabrina Schenk; Marc W. Nolte; Ingo Pragst; Gerhard Dickneite; Stefan Schulte; Sabine Zollner

INTRODUCTION The recombinant fusion protein linking coagulation factor IX with albumin (rIX-FP) is undergoing clinical trials for prophylaxis and on-demand treatment of haemophilia B patients. The aim of this study was to investigate the pharmacokinetics, whole-body and knee joint distribution of rIX-FP following intravenous administration to rats, compared with a marketed, non-fused rFIX and recombinant human albumin. MATERIAL AND METHODS [(3)H]-rIX-FP, [(3)H]-rFIX or [(3)H]-albumin were administered to rats followed by quantitative whole-body autoradiography over 24 or 240 hours, and the tissue distribution as well as elimination of radioactivity were measured. RESULTS Elimination of all radioactivity derived from the three proteins was shown to occur primarily via the urine. The tissue distribution of [(3)H]-rIX-FP and [(3)H]-rFIX (but not of [(3)H]-albumin) was comparable, both penetrating predominantly into bone, and well-perfused tissues, suggesting that the rIX moiety determines the distribution pattern of rIX-FP, while the albumin moity is responsible for the prolonged plasma and tissue retention. Detailed knee-joint analysis indicated rapid presence of [(3)H]-rIX-FP and [(3)H]-rFIX in synovial and mineralised bone tissue, mostly localised to the zone of calcified cartilage. Longest retention times were observed in the bone marrow and the endosteum of long bones. Intriguingly, [(3)H]-rIX-FP- and [(3)H]-albumin-derived radioactive signals were detectable up to 240 hours, while [(3)H]-rFIX-derived radioactivity rapidly declined after 1hour post-dosing correlating to the extended plasma half-life of [(3)H]-rIX-FP. CONCLUSION The prolonged plasma and tissue retention of rIX-FP achieved by albumin fusion may allow a reduction in dosing frequency leading to increased therapeutic compliance and convenience.

Collaboration


Dive into the Marc W. Nolte's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Guido Stoll

University of Würzburg

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge