Network


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

Hotspot


Dive into the research topics where Coen Maas is active.

Publication


Featured researches published by Coen Maas.


Journal of Clinical Investigation | 2008

Misfolded proteins activate Factor XII in humans, leading to kallikrein formation without initiating coagulation

Coen Maas; José W. P. Govers-Riemslag; Barend Bouma; Bettina Schiks; Bouke Hazenberg; Henk M. Lokhorst; Per Hammarström; Hugo ten Cate; Philip G. de Groot; Bonno N. Bouma; Martijn F. B. G. Gebbink

When blood is exposed to negatively charged surface materials such as glass, an enzymatic cascade known as the contact system becomes activated. This cascade is initiated by autoactivation of Factor XII and leads to both coagulation (via Factor XI) and an inflammatory response (via the kallikrein-kinin system). However, while Factor XII is important for coagulation in vitro, it is not important for physiological hemostasis, so the physiological role of the contact system remains elusive. Using patient blood samples and isolated proteins, we identified a novel class of Factor XII activators. Factor XII was activated by misfolded protein aggregates that formed by denaturation or by surface adsorption, which specifically led to the activation of the kallikrein-kinin system without inducing coagulation. Consistent with this, we found that Factor XII, but not Factor XI, was activated and kallikrein was formed in blood from patients with systemic amyloidosis, a disease marked by the accumulation and deposition of misfolded plasma proteins. These results show that the kallikrein-kinin system can be activated by Factor XII, in a process separate from the coagulation cascade, and point to a protective role for Factor XII following activation by misfolded protein aggregates.


Journal of Biological Chemistry | 2007

A Role for Protein Misfolding in Immunogenicity of Biopharmaceuticals

Coen Maas; Suzanne Hermeling; Barend Bouma; Wim Jiskoot; Martijn F. B. G. Gebbink

For largely unknown reasons, biopharmaceuticals evoke potentially harmful antibody formation. Such antibodies can inhibit drug efficacy and, when directed against endogenous proteins, cause life-threatening complications. Insight into the mechanisms by which biopharmaceuticals break tolerance and induce an immune response will contribute to finding solutions to prevent this adverse effect. Using a transgenic mouse model, we here demonstrate that protein misfolding, detected with the use of tissue-type plasminogen activator and thioflavin T, markers of amyloid-like properties, results in breaking of tolerance. In wild-type mice, misfolding enhances protein immunogenicity. Several commercially available biopharmaceutical products were found to contain misfolded proteins. In some cases, the level of misfolded protein was found to increase upon storage under conditions prescribed by the manufacturer. Our results indicate that misfolding of therapeutic proteins is an immunogenic signal and a risk factor for immunogenicity. These findings offer novel possibilities to detect immunogenic protein entities with tPA and reduce immunogenicity of biopharmaceuticals.


Seminars in Thrombosis and Hemostasis | 2011

The Plasma Contact System 2.0

Coen Maas; Chris Oschatz; Thomas Renné

The contact system is a protease cascade that is initiated by factor XII activation on cardiovascular cells. The system starts procoagulant and proinflammatory reactions, via the intrinsic pathway of coagulation and the kallikrein-kinin system, respectively. The biochemistry of the contact system in vitro is well understood. However, activators of the system in vivo and their contributions to disease states have remained enigmatic. Recent experimental and clinical data have identified misfolded proteins, collagens, and polyphosphates as the long-sought activators of the contact system in vivo. Here we present an overview about contact system activators and their contributions to health and pathology.


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.


Arthritis & Rheumatism | 2011

Immune responses against domain I of β2-glycoprotein I are driven by conformational changes: Domain I of β2-glycoprotein I harbors a cryptic immunogenic epitope

Bas de Laat; Miranda E.A.T. van Berkel; Rolf T. Urbanus; Berdien Siregar; Philip G. de Groot; Martijn F. B. G. Gebbink; Coen Maas

OBJECTIVE The presence of autoantibodies against a cryptic epitope in domain I of β(2)-glycoprotein I (β(2)GPI) is strongly associated with thrombotic events in patients with the antiphospholipid syndrome. We hypothesized that a conformational change could be a trigger for the formation of antibodies against domain I of β(2)GPI. Therefore, we investigated whether immune responses against β(2)GPI are related to its conformation. METHODS Conformational changes in β(2)GPI were studied using various techniques, either upon binding to cardiolipin or after disruption of the internal disulfide bonds. The immunogenicity of β(2)GPI in different conformations as well as the individual domains of β(2)GPI were studied in vivo by monitoring the generation of antibodies after intravenous administration of β(2)GPI to mice. Furthermore, plasma samples from these mice were assessed for lupus anticoagulant activity and thrombin-antithrombin complex levels. RESULTS We observed that the interaction of β(2)GPI with cardiolipin induced a conformational change in β(2)GPI: electron microscopy revealed that β(2)GPI assembled into polymeric meshworks. We next investigated the immunogenicity of both human and murine β(2)GPI in mice. Both human and murine β(2)GPI combined with cardiolipin and misfolded β(2)GPI triggered antibody formation against the native protein as well as against domain I of β(2)GPI, while native β(2)GPI was not immunogenic. In addition, we observed that anti-domain I antibodies developed in mice injected with domain I of β(2)GPI, and that antibodies did not develop in mice injected with domains II-V. The induced anti-domain I antibodies prolonged the dilute Russells viper venom plasma clotting time. The plasma of mice with anti-domain I antibodies had increased levels of circulating thrombin-antithrombin complexes. CONCLUSION The results of our studies indicate that the exposure of cryptic epitopes due to conformational changes in β(2)GPI can induce autoantibody formation.


Circulation | 2014

Plasmin Cleavage of von Willebrand Factor as an Emergency Bypass for ADAMTS13 Deficiency in Thrombotic Microangiopathy

Claudia Tersteeg; Steven de Maat; Simon F. De Meyer; Michel W.J. Smeets; Arjan D. Barendrecht; Mark Roest; Gerard Pasterkamp; Rob Fijnheer; Karen Vanhoorelbeke; Philip G. de Groot; Coen Maas

Background— Von Willebrand factor (VWF) multimer size is controlled through continuous proteolysis by ADAMTS13 (a disintegrin and metalloproteinase with a thrombospondin type I motif, member 13). This prevents spontaneous platelet agglutination and microvascular obstructions. ADAMTS13 deficiency is associated with thrombotic thrombocytopenic purpura, in which life-threatening episodes of microangiopathy damage kidneys, heart, and brain. Enigmatically, a complete ADAMTS13 deficiency does not lead to continuous microangiopathy. We hypothesized that plasmin, the key enzyme of the fibrinolytic system, serves as a physiological backup enzyme for ADAMTS13 in the degradation of pathological platelet–VWF complexes. Methods and Results— Using real-time microscopy, we determined that plasmin rapidly degrades platelet–VWF complexes on endothelial cells in absence of ADAMTS13, after activation by urokinase-type plasminogen activator or the thrombolytic agent streptokinase. Similarly, plasmin degrades platelet–VWF complexes in platelet agglutination studies. Plasminogen directly binds to VWF and its A1 domain in a lysine-dependent manner, as determined by enzyme-linked immunosorbent assay. Plasma levels of plasmin–&agr;2-antiplasmin complexes increase with the extent of thrombocytopenia in patients with acute episodes of thrombotic thrombocytopenic purpura, independent of ADAMTS13 activity. This indicates that plasminogen activation takes place during microangiopathy. Finally, we show that the thrombolytic agent streptokinase has therapeutic value for Adamts13−/− mice in a model of thrombotic thrombocytopenic purpura. Conclusions— We propose that plasminogen activation on endothelial cells acts as a natural backup for ADAMTS13 to degrade obstructive platelet–VWF complexes. Our findings indicate that thrombolytic agents may have therapeutic value in the treatment of microangiopathies and may be useful to bypass inhibitory antibodies against ADAMTS13 that cause thrombotic thrombocytopenic purpura.


FEBS Letters | 2009

Physiological responses to protein aggregates: fibrinolysis, coagulation and inflammation (new roles for old factors).

Martijn F. B. G. Gebbink; Barend Bouma; Coen Maas; Bonno N. Bouma

Misfolding is an inherent and potentially problematic propensity of proteins. Misfolded proteins tend to aggregate and the deposition of aggregated proteins is associated with a variety of highly debilitating diseases known as amyloidoses. Protein misfolding and aggregation is also increasingly recognized as the underlying cause of other health problems, including atherosclerosis and immunogenicity of biopharmaceuticals. This raises the question how nature deals with the removal of obsolete proteins in order to avoid their accumulation and disease. In recent years two proteases, tPA and factor XII, have been identified that specifically recognize aggregates of misfolded proteins. We here review these discoveries that have uncovered new roles for the fibrinolytic system and the contact activation system beyond haemostasis.


Thrombosis Research | 2012

Regulatory mechanisms of the plasma contact system

Coen Maas; Thomas Renné

The plasma contact system is a proinflammatory and procoagulant protease system. The biochemistry of the system is well established, however, its biological functions are just beginning to emerge. Here, we provide a condensed overview on mechanisms involved in activation and regulation of the contact system. These recent findings will help us to better understand the role of the enigmatic system for health and disease.


Amyloid | 2008

Identification of fibronectin type I domains as amyloid-binding modules on tissue-type plasminogen activator and three homologs

Coen Maas; Bettina Schiks; Remo D. Strangi; Tilman M. Hackeng; Bonno N. Bouma; Martijn F. B. G. Gebbink; Barend Bouma

The serine protease tissue-type plasminogen activator (tPA), a key enzyme in hemostasis, is activated by protein aggregates with amyloid-like properties. tPA is implicated in various pathologies, including amyloidoses. A major task is to further elucidate the mechanisms of amyloid pathology. We here show that the fibronectin type I domain of tPA mediates the interaction with amyloid protein aggregates. We found that in contrast to full-length tPA, a deletion-mutant of tPA, lacking the first three N-terminal domains (including the fibronectin type I domain), fails to activate in response to amyloid protein aggregates. Using recombinantly produced domains of tPA in direct binding assays, we subsequently mapped the amyloid-binding region to the fibronectin type I domain. This domain co-localized with congophylic plaques in brain sections from patients with Alzheimers disease. Fibronectin type I domains from homologous proteases factor XII, hepatocyte growth factor activator and from the extracellular matrix protein fibronectin also bound to aggregated amyloidogenic peptides. Finally, we demonstrated that the isolated fibronectin type I domain inhibits amyloid-induced aggregation of blood platelets. The identification of the fibronectin type I domain as an amyloid-binding module provides new insights into the (patho-) physiological role of tPA and the homologous proteins which may offer new targets for intervention in amyloid pathology.


Thrombosis and Haemostasis | 2013

A nanobody-based method for tracking factor XII activation in plasma

S de Maat; S. van Dooremalen; P. G. De Groot; Coen Maas

The physiological role of the plasma protein factor XII (FXII), as well as its involvement in human pathology, is poorly understood. While FXII is implicated in thrombotic pathology as a coagulation factor, it can contribute to inflammatory conditions without triggering coagulation. We recently generated nanobodies against the catalytic domain of activated FXII (FXIIa). Here, we describe two of these nanobodies, A10 and B7, both of which do not recognise FXII. Nanobody A10 recognises the catalytic domain of purified α-FXIIa (80 kDa), but not that of purified β-FXIIa (28 kDa), whereas nanobody B7 recognises both. This suggests minute differences in the catalytic domain between these isoforms of FXIIa. The detection of FXIIa by these nanobodies in plasma can become compromised through inactivation by serine protease inhibitors. This effect can be efficiently countered through the addition of the small-molecular protease inhibitor PPACK. Finally, we show that our nanobody-based assays in vitro distinguish various activation products of FXII that differ with the type of activator present: whereas procoagulant activators solely trigger the formation of a species that is captured by B7, proinflammatory activators first generate a species that is recognised by B7, which is later converted into a species that is recognised by A10. These findings suggest that a progressive proteolysis of FXIIa results in the generation a non-procoagulant form of FXIIa, whereas retention of intermediate forms triggers coagulation. Moreover, our findings indicate the development of nanobodies against activated enzymes offers improved opportunities to investigate their contribution to health and disease.

Collaboration


Dive into the Coen Maas's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Claudia Tersteeg

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge