Network


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

Hotspot


Dive into the research topics where Frédéric Adam is active.

Publication


Featured researches published by Frédéric Adam.


Blood | 2011

Thrombocytopenia resulting from mutations in filamin A can be expressed as an isolated syndrome

Paquita Nurden; Najet Debili; Isabelle Coupry; Marijke Bryckaert; Ibtissam Youlyouz-Marfak; Guilhem Solé; Anne-Cécile Pons; Eliane Berrou; Frédéric Adam; Alexandre Kauskot; Jean-Marie Daniel Lamazière; Philippe Rameau; Patricia Fergelot; Caroline Rooryck; Dorothée Cailley; Benoit Arveiler; Didier Lacombe; William Vainchenker; Alan T. Nurden; Cyril Goizet

Filaminopathies A caused by mutations in the X-linked FLNA gene are responsible for a wide spectrum of rare diseases including 2 main phenotypes, the X-linked dominant form of periventricular nodular heterotopia (FLNA-PVNH) and the otopalatodigital syndrome spectrum of disorders. In platelets, filamin A (FLNa) tethers the principal receptors ensuring the platelet-vessel wall interaction, glycoprotein Ibα and integrin αIIbβ3, to the underlying cytoskeleton. Hemorrhage, coagulopathy, and thrombocytopenia are mentioned in several reports on patients with FLNA-PVNH. Abnormal platelet morphology in 2 patients with FLNA-PVNH prompted us to examine a third patient with similar platelet morphology previously diagnosed with immunologic thrombocytopenic purpura. Her enlarged platelets showed signs of FLNa degradation in Western blotting, and a heterozygous missense mutation in FLNA was detected. An irregular distribution of FLNa within the total platelet population was shown by confocal microscopy for all 3 patients. In vitro megakaryocyte cultures showed an abnormal differentiation, including an irregular distribution of FLNa with a frayed aspect, the presence of enlarged α-granules, and an abnormal fragmentation of the cytoplasm. Mutations in FLNA may represent an unrecognized cause of macrothrombocytopenia with an altered platelet production and a modified platelet-vessel wall interaction.


Journal of Biological Chemistry | 2007

Protease-activating Receptor-4 Induces Full Platelet Spreading on a Fibrinogen Matrix INVOLVEMENT OF ERK2 AND p38 AND Ca2+ MOBILIZATION

Alexandra Mazharian; Séverine Roger; Eliane Berrou; Frédéric Adam; Alexandre Kauskot; Paquita Nurden; Martine Jandrot-Perrus; Marijke Bryckaert

Although the involvement of protease-activating receptor PAR1 and PAR4 is well established in platelet aggregation, their role in platelet adhesion and spreading has yet to be characterized. We investigated platelet adhesion and spreading on a fibrinogen matrix after PAR1 and PAR4 stimulation in correlation with the activation of two MAPKs, ERK2 and p38. Of the two PAR-activating peptides (PAR-APs), PAR1-AP and PAR4-AP, which both induce adhesion, only PAR4-AP induced full platelet spreading. Although both PAR1-AP and PAR4-AP induced ADP secretion, which is required for platelet spreading, only PAR4-AP induced sustained Ca2+ mobilization. In these conditions of PAR4 induction, ERK2 and p38 activation were involved in platelet spreading but not in platelet adhesion. p38 phosphorylation was dependent on ADP signaling through P2Y12, its receptor. ERK2 phosphorylation was triggered through integrin αIIbβ3 outside-in signaling and was dependent on the Rho pathway. ERK2 and p38 activation induced phosphorylation of the myosin light chain and actin polymerization, respectively, necessary for cytoskeleton reorganization. These findings provide the first evidence that thrombin requires PAR4 for the full spreading response. ERK2 and p38 and sustained Ca2+ mobilization, involved in PAR4-induced platelet spreading, contribute to the stabilization of platelet thrombi at sites of high thrombin production.


Journal of Biological Chemistry | 2007

Involvement of the Mitogen-activated Protein Kinase c-Jun NH2-terminal Kinase 1 in Thrombus Formation

Alexandre Kauskot; Frédéric Adam; Alexandra Mazharian; Nadine Ajzenberg; Eliane Berrou; Arnaud Bonnefoy; Jean-Philippe Rosa; Marc Hoylaerts; Marijke Bryckaert

The involvement of the mitogen-activated protein kinase c-Jun NH2-terminal kinase-1 (JNK1) has never been investigated in hemostasis and thrombosis. Using two JNK inhibitors (SP600125 and 6o), we have demonstrated that JNK1 is involved in collagen-induced platelet aggregation dependent on ADP. In these conditions, JNK1 activation requires the coordinated signaling pathways of collagen receptors (α2β1 and glycoprotein (GP)VI) and ADP. In contrast, JNK1 is not required for platelet adhesion on a collagen matrix in static or blood flow conditions (300–1500 s–1) involving collagen receptors (α2β1 and GPVI). Importantly, at 1500 s–1, JNK1 acts on thrombus formation on a collagen matrix dependent on GPIb-von Willebrand factor (vWF) interaction but not ADP receptor activation. This is confirmed by the involvement of JNK1 in shear-induced platelet aggregation at 4000 s–1. We also provide evidence during rolling and adhesion of platelets to vWF that platelet GPIb-vWF interaction triggers αIIbβ3 activation in a JNK1-dependent manner. This was confirmed with a Glanzmann thrombastenic patient lacking αIIbβ3. Finally, in vivo, JNK1 is involved in arterial but not in venular thrombosis in mice. Overall, our in vitro studies define a new role of JNK1 in thrombus formation in flowing blood that is relevant to thrombus development in vivo.


Journal of Thrombosis and Haemostasis | 2006

Insights into abnormal hemostasis in the Quebec platelet disorder from analyses of clot lysis

M. Diamandis; Frédéric Adam; W. H. A. Kahr; P. Wang; K. A. Chorneyko; A. L. Arsenault; Georges E. Rivard; Catherine P. M. Hayward

Summary.  Background: The Quebec platelet disorder (QPD) is inherited and characterized by delayed‐onset bleeding following hemostatic challenge. Other characteristics include increased expression and storage of active urokinase‐type plasminogen activator (u‐PA) in platelets in the setting of normal to increased u‐PA in plasma. There is also consumption of platelet plasminogen activator inhibitor‐1 and increased generation of plasmin in platelets accompanied by proteolysis of stored α‐granule proteins, including Factor V. Aims and Methods: Although fibrinolysis has been proposed to contribute to QPD bleeding, the effects of QPD blood and platelets on clot lysis have not been evaluated. We used thromboelastography® (TEG®), biochemical evaluations of whole blood clot lysis, assessments of clot ultrastructure, and perfusion of blood over preformed fibrin to gain insights into the disturbed hemostasis in the QPD. Results: Thromboelastography was not sensitive to the increased u‐PA in QPD blood. However, there was abnormal plasmin generation in QPD whole blood clots, generated at low shear, with biochemical evidence of increased fibrinolysis. The incorporation of QPD platelets into a forming clot led to progressive disruption of fibrin and platelet aggregates unless drugs were added to inhibit plasmin. In whole blood perfusion studies, QPD platelets showed normal adherence to fibrin, but their adhesion was followed by accelerated fibrinolysis. Conclusions: The QPD is associated with ‘gain‐of‐function’ abnormalities that increase the lysis of forming or preformed clots. These findings suggest accelerated fibrinolysis is an important contributor to QPD bleeding.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2013

Heterogeneity of Platelet Functional Alterations in Patients With Filamin A Mutations

Eliane Berrou; Frédéric Adam; Marilyne Lebret; Patricia Fergelot; Alexandre Kauskot; Isabelle Coupry; Martine Jandrot-Perrus; Alan T. Nurden; Rémi Favier; Jean-Philippe Rosa; Cyril Goizet; Paquita Nurden; Marijke Bryckaert

Objective—We examined platelet functions in 4 unrelated patients with filaminopathy A caused by dominant mutations of the X-linked filamin A (FLNA) gene. Methods and Results—Patients P1, P2, and P4 exhibited periventricular nodular heterotopia, heterozygozity for truncating FLNA mutations, and thrombocytopenia (except P2). P3 exhibited isolated thrombocytopenia and heterozygozity for a p.Glu1803Lys FLNA mutation. Truncated FLNa was undetectable by Western blotting of P1, P2, and P4 platelets, but full-length FLNa was detected at 37%, 82%, and 57% of control, respectively. P3 FLNa (p.Glu1803Lys and full-length) was assessed at 79%. All patients exhibited a platelet subpopulation negative for FLNa. Platelet aggregation, secretion, glycoprotein VI signaling, and thrombus growth on collagen were decreased for P1, P3, and P4, but normal for P2. For the 2 patients analyzed (P1 and P4), spreading was enhanced and, more markedly, in FLNa-negative platelets, suggesting that FLNa negatively regulates cytoskeleton reorganization. Platelet adhesion to von Willebrand factor under flow correlated with platelet full-length FLNa content: markedly reduced for P1 and P4 and unchanged for P2. Interestingly, von Willebrand factor flow adhesion was increased for P3, consistent with a gain-of-function effect enhancing glycoprotein Ib-IX-V/von Willebrand factor interaction. These results are consistent with a positive role for FLNa in platelet adhesion under high shear. Conclusion—FLNA mutation heterogeneity correlates with different platelet functional impacts and points to opposite regulatory roles of FLNa in spreading and flow adhesion under shear.


Journal of Clinical Investigation | 2013

von Willebrand factor mutation promotes thrombocytopathy by inhibiting integrin αIIbβ3

Caterina Casari; Eliane Berrou; Marilyne Lebret; Frédéric Adam; Alexandre Kauskot; Regis Bobe; Céline Desconclois; Edith Fressinaud; Olivier D. Christophe; Peter J. Lenting; Jean-Philippe Rosa; Cécile V. Denis; Marijke Bryckaert

von Willebrand disease type 2B (vWD-type 2B) is characterized by gain-of-function mutations in von Willebrand factor (vWF) that enhance its binding to the glycoprotein Ib-IX-V complex on platelets. Patients with vWD-type 2B have a bleeding tendency that is linked to loss of vWF multimers and/or thrombocytopenia. In this study, we uncovered evidence that platelet dysfunction is a third possible mechanism for bleeding tendency. We found that platelet aggregation, secretion, and spreading were diminished due to inhibition of integrin αIIbβ3 in platelets from mice expressing a vWD-type 2B-associated vWF (vWF/p.V1316M), platelets from a patient with the same mutation, and control platelets pretreated with recombinant vWF/p.V1316M. Impaired platelet function coincided with reduced thrombus growth. Further, αIIbβ3 activation and activation of the small GTPase Rap1 were impaired by vWF/p.V1316M following exposure to platelet agonists (thrombin, ADP, or convulxin). Conversely, thrombin- or ADP-induced Ca2+ store release, which is required for αIIbβ3 activation, was normal, indicating that vWF/p.V1316M acts downstream of Ca2+ release and upstream of Rap1. We found normal Syk phosphorylation and PLCγ2 activation following collagen receptor signaling, further implying that vWF/p.V1316M acts directly on or downstream of Ca2+ release. These data indicate that the vWD-type 2B mutation p.V1316M is associated with severe thrombocytopathy, which likely contributes to the bleeding tendency in vWD-type 2B.


Thrombosis and Haemostasis | 2005

Analyses of cellular multimerin 1 receptors: in vitro evidence of binding mediated by αIibβ3 and αvβ3

Frédéric Adam; Shilun Zheng; Nilesh Joshi; David S. Kelton; Amin Sandhu; Youko Suehiro; Samira B. Jeimy; Aurelio Santos; Jean-Marc Massé; John G. Kelton; Elisabeth M. Cramer; Catherine P. M. Hayward

Multimerin 1 (MMRN1) is a large, soluble, polymeric, factor V binding protein and member of the EMILIN protein family. In vivo, MMRN1 is found in platelets, megakaryocytes, endothelium and extracellular matrix fibers, but not in plasma. To address the mechanism of MMRN1 binding to activated platelets and endothelial cells, we investigated the identity of the major MMRN1 receptors on these cells using wild-type and RGE-forms of recombinant MMRN1.Ligand capture,cell adhesion,ELISA and flow cytometry analyses of platelet-MMRN1 binding, indicated that MMRN1 binds to integrins αIibβ 3 and αvβ3. Endothelial cell binding to MMRN1 was predominantly mediated by αvβ3 and did not require the MMRN1 RGD site or cellular activation.Like many other αvβ3 ligands, MMRN1 had the ability to support adhesion of additional cell types, including stimulated neutrophils. Expression studies, using a cell line capable of endothelial-like MMRN1 processing, indicated that MMRN1 adhesion to cellular receptors enhanced its extracellular matrix fiber assembly. These studies implicate integrin-mediated binding in MMRN1 attachment to cells and indicate that MMRN1 is a ligand for aIibβ 3 and αvβ3.


Journal of Thrombosis and Haemostasis | 2009

Platelet adhesion to multimerin 1 in vitro: influences of platelet membrane receptors, von Willebrand factor and shear

Subia Tasneem; Frédéric Adam; I. Minullina; M. Pawlikowska; S. K. Hui; S. Zheng; J. L. Miller; Catherine P. M. Hayward

Summary.  Background: Multimerin 1 (MMRN1) is a large, homopolymeric adhesive protein, stored in platelets and endothelium, that when released, binds to activated platelets, endothelial cells and the extracellular matrix. Objectives: The goals of our study were to determine if (i) MMRN1 supports adhesion of resting and/or activated platelets under conditions of blood flow, and (ii) if MMRN1 enhances platelet adhesion to types I and III collagen. Patients/methods: Platelet adhesion was evaluated using protein‐coated microcapillaries, with or without added adhesive proteins and receptor antibodies. Platelets from healthy controls, Glanzmann thrombasthenia (GT) and severe von Willebrand factor (VWF)‐deficient donors were tested. Results: MMRN1 supported the adhesion of activated, but not resting, washed platelets over a wide range of shear rates. At low shear (150 s−1), this adhesion was supported by integrins αvβ3 and glycoprotein (GP) Ibα but it did not require integrins αIIbβ3 or VWF. At high shear (1500 s−1), adhesion to MMRN1 was supported by β3 integrin‐independent mechanisms, involving GPIbα and VWF, that did not require platelet activation when VWF was perfused over MMRN1 prior to platelets. MMRN1 bound to types I and III collagen, independent of VWF, however, its enhancing effects on platelet adhesion to collagen at high shear were VWF dependent. Conclusions: MMRN1 supports platelet adhesion by VWF‐dependent and ‐independent mechanisms that vary by flow rate. Additionally, MMRN1 binds to, and enhances, platelet adhesion to collagen. These findings suggest that MMRN1 could function as an adhesive ligand that promotes platelet adhesion at sites of vascular injury.


Thrombosis and Haemostasis | 2004

Human platelets contain forms of Factor V in disulfide-linkage with multimerin

Catherine P. M. Hayward; Nola Fuller; Shilun Zheng; Frédéric Adam; Samira B. Jeimy; Ian Horsewood; Mary Ann Quinn-Allen; William H. Kane

Factor V is an essential cofactor for blood coagulation that circulates in platelets and plasma. Unlike plasma factor V, platelet factor V is stored complexed with the polymeric alpha-granule protein multimerin. In analyses of human platelet factor V on nonreduced denaturing multimer gels, we identified that approximately 25% was variable in size and migrated larger than single chain factor V, the largest form in plasma. Upon reduction, the unusually large, variably-sized forms of platelet factor V liberated components that comigrated with other forms of platelet factor V, indicating that they contained factor V in interchain disulfide-linkages. With thrombin cleavage, factor Va heavy and light chain domains, but not B-domains,were liberated from the components linked by interchain disulfide bonds, indicating that the single cysteine in the B-domain at position 1085 was the site of disulfide linkage. Since unusually large factor V had a variable size and included forms larger than factor V dimers, the data suggested disulfide-linkage with another platelet protein, possibly multimerin. Immunoprecipitation experiments confirmed that unusually large factor V was associated with multimerin and it remained associated in 0.5 M salt. Moreover, platelets contained a subpopulation of multimerin polymers that resisted dissociation from factor V by denaturing detergent and comigrated with unusually large platelet factor V, before and after thrombin cleavage. The disulfide-linked complexes of multimerin and factor V in platelets, which are cleaved by thrombin to liberate factor Va, could be important for modulating the function of platelet factor V and its delivery onto activated platelets. Factor Va generation and function from unusually large platelet factor V is only speculative at this time.


Scientific Reports | 2016

A genetically-engineered von Willebrand disease type 2B mouse model displays defects in hemostasis and inflammation

Frédéric Adam; Caterina Casari; Nicolas Prévost; Alexandre Kauskot; Cécile Loubière; Paulette Legendre; Christelle Repérant; Dominique Baruch; Jean-Philippe Rosa; Marijke Bryckaert; Philip G. de Groot; Olivier D. Christophe; Peter J. Lenting; Cécile V. Denis

von Willebrand disease (VWD)-type 2B is characterized by gain-of-function mutations in the von Willebrand factor (VWF) A1-domain, leading to increased affinity for its platelet-receptor, glycoprotein Ibα. We engineered the first knock-in (KI) murine model for VWD-type 2B by introducing the p.V1316M mutation in murine VWF. Homozygous KI-mice replicated human VWD-type 2B with macrothrombocytopenia (platelet counts reduced by 55%, platelet volume increased by 44%), circulating platelet-aggregates and a severe bleeding tendency. Also, vessel occlusion was deficient in the FeCl3-induced thrombosis model. Platelet aggregation induced by thrombin or collagen was defective for KI-mice at all doses. KI-mice manifested a loss of high molecular weight multimers and increased multimer degradation. In a model of VWF-string formation, the number of platelets/string and string-lifetime were surprisingly enhanced in KI-mice, suggesting that proteolysis of VWF/p.V1316M is differentially regulated in the circulation versus the endothelial surface. Furthermore, we observed increased leukocyte recruitment during an inflammatory response induced by the reverse passive Arthus reaction. This points to an active role of VWF/p.V1316M in the exfiltration of leukocytes under inflammatory conditions. In conclusion, our genetically-engineered VWD-type 2B mice represent an original model to study the consequences of spontaneous VWF-platelet interactions and the physiopathology of this human disease.

Collaboration


Dive into the Frédéric Adam's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Paquita Nurden

Centre national de la recherche scientifique

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Georges E. Rivard

Centre Hospitalier Universitaire Sainte-Justine

View shared research outputs
Top Co-Authors

Avatar

Caterina Casari

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Jean-Claude Bordet

French Institute of Health and Medical Research

View shared research outputs
Researchain Logo
Decentralizing Knowledge