Network


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

Hotspot


Dive into the research topics where Alexandra Dusa is active.

Publication


Featured researches published by Alexandra Dusa.


Seminars in Cell & Developmental Biology | 2008

JAKs in pathology: Role of Janus kinases in hematopoietic malignancies and immunodeficiencies

William Vainchenker; Alexandra Dusa; Stefan N. Constantinescu

The four mammalian Janus kinase (JAK) family members, JAK1, JAK2, JAK3 and TYK2, are non-receptor protein tyrosine kinases (PTKs) that are crucial for cytokine receptor signaling in blood formation and immune responses. Mutations and translocations in the JAK genes leading to constitutively active JAK proteins are associated with a variety of hematopoietic malignancies, including the myeloproliferative disorders (JAK2), acute lymphoblastic leukemia (JAK2), acute myeloid leukemia (JAK2, JAK1), acute megakaryoblastic leukemia (JAK2, JAK3) and T-cell precursor acute lymphoblastic leukemia (JAK1). In contrast, loss-of-function mutations of JAK3 and TYK2 lead to immunodeficiency. The role of JAKs as therapeutic targets is starting to expand, as more insights into their structure and activation mechanisms become available.


PLOS ONE | 2010

JAK2 V617F constitutive activation requires JH2 residue F595: a pseudokinase domain target for specific inhibitors.

Alexandra Dusa; Céline Mouton; Christian Pecquet; Murielle Herman; Stefan N. Constantinescu

The JAK2 V617F mutation present in over 95% of Polycythemia Vera patients and in 50% of Essential Thrombocythemia and Primary Myelofibrosis patients renders the kinase constitutively active. In the absence of a three-dimensional structure for the full-length protein, the mechanism of activation of JAK2 V617F has remained elusive. In this study, we used functional mutagenesis to investigate the involvement of the JH2 αC helix in the constitutive activation of JAK2 V617F. We show that residue F595, located in the middle of the αC helix of JH2, is indispensable for the constitutive activity of JAK2 V617F. Mutation of F595 to Ala, Lys, Val or Ile significantly decreases the constitutive activity of JAK2 V617F, but F595W and F595Y are able to restore it, implying an aromaticity requirement at position 595. Substitution of F595 to Ala was also able to decrease the constitutive activity of two other JAK2 mutants, T875N and R683G, as well as JAK2 K539L, albeit to a lower extent. In contrast, the F595 mutants are activated by erythropoietin-bound EpoR. We also explored the relationship between the dimeric conformation of EpoR and several JAK2 mutants. Since residue F595 is crucial to the constitutive activation of JAK2 V617F but not to initiation of JAK2 activation by cytokines, we suggest that small molecules that target the region around this residue might specifically block oncogenic JAK2 and spare JAK2 wild-type.


Journal of Biological Chemistry | 2008

Substitution of pseudokinase domain residue Val-617 by large non-polar amino acids causes activation of JAK2.

Alexandra Dusa; Judith Staerk; Joanne Elliott; Christian Pecquet; Hélène Poirel; James A. Johnston; Stefan N. Constantinescu

Explaining the uniqueness of the acquired somatic JAK2 V617F mutation, which is present in more than 95% of polycythemia vera patients, has been a challenge. The V617F mutation in the pseudokinase domain of JAK2 renders the unmutated kinase domain constitutively active. We have performed random mutagenesis at position 617 of JAK2 and tested each of the 20 possible amino acids for ability to induce constitutive signaling in Ba/F3 cells expressing the erythropoietin receptor. Four JAK2 mutants, V617W, V617M, V617I, and V617L, were able to induce cytokine independence and constitutive downstream signaling. Only V617W induced a level of constitutive activation comparable with V617F. Also, only V617W stabilized tyrosine-phosphorylated suppressor of cytokine signaling 3 (SOCS3), a mechanism by which JAK2 V617F overcomes inhibition by SOCS3. The V617W mutant induced a myeloproliferative disease in mice, mainly characterized by erythrocytosis and megakaryocytic proliferation. Although JAK2 V617W would predictably be pathogenic in humans, the substitution of the Val codon, GTC, by TTG, the codon for Trp, would require three base pair changes, and thus it is unlikely to occur. We discuss how the predicted conformations of the activated JAK2 mutants can lead to better screening assays for novel small molecule inhibitors.


Haematologica | 2011

Oncogenic JAK1 and JAK2-activating mutations resistant to ATP-competitive inhibitors

Tekla Hornakova; Lorraine Springuel; Julien Devreux; Alexandra Dusa; Stefan N. Constantinescu; Laurent Knoops; Jean-Christophe Renauld

Background Activating mutations in JAK1 and JAK2 have been described in patients with various hematologic malignancies including acute lymphoblastic leukemia and myeloproliferative neoplasms, leading to clinical trials with JAK inhibitors. While there has been a tremendous effort towards the development of specific JAK inhibitors, mutations conferring resistance to such drugs have not yet been observed. Design and Methods Taking advantage of a model of spontaneous cellular transformation, we sequenced JAK1 in selected tumorigenic BaF3 clones and identified 25 de novo JAK1 activating mutations, including 5 mutations already described in human leukemias. We further used this library of JAK1 mutation-positive cell lines to assess their sensitivity to ATP-competitive inhibitors. Results While most JAK1 mutants were sensitive to ATP-competitive JAK inhibitors, mutations targeting Phe958 and Pro960 in the hinge region of the kinase domain rendered JAK1 constitutively active but also resistant to all tested JAK inhibitors. Furthermore, mutation of the homologous Tyr931 in JAK2 wild-type or JAK2 V617F mutant found in patients with myeloproliferative neoplasms also conferred resistance to JAK inhibitors, such as INCB018424, which is currently in clinical use. Conclusions Our data indicate that some activating mutations not only promote autonomous cell proliferation but also confer resistance to ATP-competitive inhibitors. In vivo, such a mutation can potentially occur as primary JAK-activating mutations but also as secondary mutations combining oncogenicity with drug resistance.


Blood | 2012

Thrombopoietin receptor down-modulation by JAK2 V617F: restoration of receptor levels by inhibitors of pathologic JAK2 signaling and of proteasomes

Christian Pecquet; Carmen C. Diaconu; Judith Staerk; Michael Girardot; Caroline Marty; Yohan Royer; Jean-Philippe Defour; Alexandra Dusa; Rodolphe Besancenot; Stéphane Giraudier; Jean-Luc Villeval; Laurent Knoops; Pierre J. Courtoy; William Vainchenker; Stefan N. Constantinescu

The constitutively active JAK2 V617F mutant is the major determinant of human myeloproliferative neoplasms (MPNs). We show that coexpression of murine JAK2 V617F and the murine thrombopoietin (Tpo) receptor (TpoR, c-MPL) in hematopoietic cell lines or heterozygous knock-in of JAK2 V617F in mice leads to down-modulation of TpoR levels. Enhanced TpoR ubiquitinylation, proteasomal degradation, reduced recycling, and maturation are induced by the constitutive JAK2 V617F activity. These effects can be prevented in cell lines by JAK2 and proteasome inhibitors. Restoration of TpoR levels by inhibitors could be detected in platelets from JAK2 inhibitor-treated myelofibrosis patients that express the JAK2 V617F mutant, and in platelets from JAK2 V617F knock-in mice that were treated in vivo with JAK2 or proteasome inhibitors. In addition, we show that Tpo can induce both proliferative and antiproliferative effects via TpoR at low and high JAK2 activation levels, respectively, or on expression of JAK2 V617F. The antiproliferative signaling and receptor down-modulation by JAK2 V617F were dependent on signaling via TpoR cytosolic tyrosine 626. We propose that selection against TpoR antiproliferative signaling occurs by TpoR down-modulation and that restoration of down-modulated TpoR levels could become a biomarker for the treatment of MPNs.


Blood | 2013

Impact of isolated germline JAK2V617I mutation on human hematopoiesis.

Adam Mead; Onima Chowdhury; Christian Pecquet; Alexandra Dusa; Petter S. Woll; Deborah Atkinson; Adam Burns; Joannah Score; Michelle Rugless; Ruth Clifford; Simon Moule; Nicola Bienz; Paresh Vyas; Nicholas C.P. Cross; Rosemary E. Gale; Shirley Henderson; Stefan N. Constantinescu; Anna Schuh; Sten Eirik W. Jacobsen

The association between somatic JAK2 mutation and myeloproliferative neoplasms (MPNs) is now well established. However, because JAK2 mutations are associated with heterogeneous clinical phenotypes and often occur as secondary genetic events, some aspects of JAK2 mutation biology remain to be understood. We recently described a germline JAK2V617I mutation in a family with hereditary thrombocytosis and herein characterize the hematopoietic and signaling impact of JAK2V617I. Through targeted sequencing of MPN-associated mutations, exome sequencing, and clonality analysis, we demonstrate that JAK2V617I is likely to be the sole driver mutation in JAK2V617I-positive individuals with thrombocytosis. Phenotypic hematopoietic stem cells (HSCs) were increased in the blood and bone marrow of JAK2V617I-positive individuals and were sustained at higher levels than controls after xenotransplantation. In signaling and transcriptional assays, JAK2V617I demonstrated more activity than wild-type JAK2 but substantially less than JAK2V617F. After cytokine stimulation, JAK2V617I resulted in markedly increased downstream signaling compared with wild-type JAK2 and comparable with JAK2V617F. These findings demonstrate that JAK2V617I induces sufficient cytokine hyperresponsiveness in the absence of other molecular events to induce a homogeneous MPN-like phenotype. We also provide evidence that the JAK2V617I mutation may expand the HSC pool, providing insights into both JAK2 mutation biology and MPN disease pathogenesis.


Biochemical Journal | 2016

Uncoupling JAK2 V617F activation from cytokine-induced signalling by modulation of JH2 αC helix

Emilie Leroy; Alexandra Dusa; Didier Colau; Amir Motamedi; Xavier Cahu; Céline Mouton; Lily Jun Shen Huang; Andrew K. Shiau; Stefan N. Constantinescu

We show that JAK2 activation by the pseudokinase V617F mutation is mediated by a community of residues across JAK2. Charge changes on pseudokinase helix αC specifically inhibits V617F-induced activation. Thus, allosteric modulation can lead to specific JAK2 V617F inhibition.


Biochemical Society Transactions | 2013

Activating Janus kinase pseudokinase domain mutations in myeloproliferative and other blood cancers

Stefan N. Constantinescu; Emilie Leroy; Vitalina Gryshkova; Christian Pecquet; Alexandra Dusa

The discovery of the highly prevalent activating JAK (Janus kinase) 2 V617F mutation in myeloproliferative neoplasms, and of other pseudokinase domain-activating mutations in JAK2, JAK1 and JAK3 in blood cancers, prompted great interest in understanding how pseudokinase domains regulate kinase domains in JAKs. Recent functional and mutagenesis studies identified residues required for the V617F mutation to induce activation. Several X-ray crystal structures of either kinase or pseudokinase domains including the V617F mutant of JAK2 pseudokinase domains are now available, and a picture has emerged whereby the V617F mutation induces a defined conformational change around helix C of JH (JAK homology) 2. Effects of mutations on JAK2 can be extrapolated to JAK1 and TYK2 (tyrosine kinase 2), whereas JAK3 appears to be different. More structural information of the full-length JAK coupled to cytokine receptors might be required in order to define the structural basis of JH1 activation by JH2 mutants and eventually obtain mutant-specific inhibitors.


Pathologie Biologie | 2007

JAK2, the JAK2 V617F mutant and cytokine receptors

Judith Staerk; A. Kallin; Yohan Royer; Carmen-Cristina Diaconu; Alexandra Dusa; Jean-Baptiste Demoulin; William Vainchenker; Stefan N. Constantinescu


Blood | 2008

Functional Determination of the Interaction Interface Between the Kinase and Pseudokinase Domains of JAK2 V617F

Alexandra Dusa; Stefan N. Constantinescu

Collaboration


Dive into the Alexandra Dusa's collaboration.

Top Co-Authors

Avatar

Stefan N. Constantinescu

Ludwig Institute for Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Christian Pecquet

Ludwig Institute for Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Judith Staerk

Ludwig Institute for Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Yohan Royer

Ludwig Institute for Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Carmen C. Diaconu

Ludwig Institute for Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Céline Mouton

Ludwig Institute for Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Emilie Leroy

Ludwig Institute for Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Jean-Baptiste Demoulin

Université catholique de Louvain

View shared research outputs
Top Co-Authors

Avatar

Laurent Knoops

Cliniques Universitaires Saint-Luc

View shared research outputs
Top Co-Authors

Avatar

A. Kallin

Université catholique de Louvain

View shared research outputs
Researchain Logo
Decentralizing Knowledge