Network


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

Hotspot


Dive into the research topics where Jun Mori is active.

Publication


Featured researches published by Jun Mori.


Cell | 2011

Global proteomic assessment of the classical protein-tyrosine phosphatome and "Redoxome".

Robert Karisch; Minerva Fernandez; Paul Taylor; Carl Virtanen; Jonathan R. St-Germain; Lily L. Jin; Isaac S. Harris; Jun Mori; Yotis A. Senis; Arne Östman; Michael F. Moran; Benjamin G. Neel

Protein-tyrosine phosphatases (PTPs), along with protein-tyrosine kinases, play key roles in cellular signaling. All Class I PTPs contain an essential active site cysteinyl residue, which executes a nucleophilic attack on substrate phosphotyrosyl residues. The high reactivity of the catalytic cysteine also predisposes PTPs to oxidation by reactive oxygen species, such as H(2)O(2). Reversible PTP oxidation is emerging as an important cellular regulatory mechanism and might contribute to diseases such as cancer. We exploited these unique features of PTP enzymology to develop proteomic methods, broadly applicable to cell and tissue samples, that enable the comprehensive identification and quantification of expressed classical PTPs (PTPome) and the oxidized subset of the PTPome (oxPTPome). We find that mouse and human cells and tissues, including cancer cells, display distinctive PTPomes and oxPTPomes, revealing additional levels of complexity in the regulation of protein-tyrosine phosphorylation in normal and malignant cells.


Blood | 2010

CLEC-2 activates Syk through dimerization.

Craig E. Hughes; Alice Y. Pollitt; Jun Mori; Johannes A. Eble; Michael G. Tomlinson; John H. Hartwig; Christopher A. O'Callaghan; Klaus Fütterer; Steve P. Watson

The C-type lectin receptor CLEC-2 activates platelets through Src and Syk tyrosine kinases, leading to tyrosine phosphorylation of downstream adapter proteins and effector enzymes, including phospholipase-C gamma2. Signaling is initiated through phosphorylation of a single conserved tyrosine located in a YxxL sequence in the CLEC-2 cytosolic tail. The signaling pathway used by CLEC-2 shares many similarities with that used by receptors that have 1 or more copies of an immunoreceptor tyrosine-based activation motif, defined by the sequence Yxx(L/I)x(6-12)Yxx(L/I), in their cytosolic tails or associated receptor chains. Phosphorylation of the conserved immunoreceptor tyrosine-based activation motif tyrosines promotes Syk binding and activation through binding of the Syk tandem SH2 domains. In this report, we present evidence using peptide pull-down studies, surface plasmon resonance, quantitative Western blotting, tryptophan fluorescence measurements, and competition experiments that Syk activation by CLEC-2 is mediated by the cross-linking through the tandem SH2 domains with a stoichiometry of 2:1. In support of this model, cross-linking and electron microscopy demonstrate that CLEC-2 is present as a dimer in resting platelets and converted to larger complexes on activation. This is a unique mode of activation of Syk by a single YxxL-containing receptor.


Blood | 2014

Src family kinases: at the forefront of platelet activation

Yotis A. Senis; Alexandra Mazharian; Jun Mori

Src family kinases (SFKs) play a central role in mediating the rapid response of platelets to vascular injury. They transmit activation signals from a diverse repertoire of platelet surface receptors, including the integrin αIIbβ3, the immunoreceptor tyrosine-based activation motif-containing collagen receptor complex GPVI-FcR γ-chain, and the von Willebrand factor receptor complex GPIb-IX-V, which are essential for thrombus growth and stability. Ligand-mediated clustering of these receptors triggers an increase in SFK activity and downstream tyrosine phosphorylation of enzymes, adaptors, and cytoskeletal proteins that collectively propagate the signal and coordinate platelet activation. A growing body of evidence has established that SFKs also contribute to Gq- and Gi-coupled receptor signaling that synergizes with primary activation signals to maximally activate platelets and render them prothrombotic. Interestingly, SFKs concomitantly activate inhibitory pathways that limit platelet activation and thrombus size. In this review, we discuss past discoveries that laid the foundation for this fundamental area of platelet signal transduction, recent progress in our understanding of the distinct and overlapping functions of SFKs in platelets, and new avenues of research into mechanisms of SFK regulation. We also highlight the thrombotic and hemostatic consequences of targeting platelet SFKs.


Journal of Thrombosis and Haemostasis | 2009

The novel Syk inhibitor R406 reveals mechanistic differences in the initiation of GPVI and CLEC‐2 signaling in platelets

Jennifer C. Spalton; Jun Mori; Alice Y. Pollitt; Craig E. Hughes; Johannes A. Eble; Steve P. Watson

Summary.  Background: Syk is a key mediator of signaling pathways downstream of several platelet surface receptors including GPVI/FcRγ collagen receptor, the C‐type lectin receptor CLEC‐2, and integrin αIIbβ3. A recent study identified the novel small molecule R406 as a selective inhibitor of Syk. Objectives: The present study evaluates the role of Syk in human platelets using the novel inhibitor R406. Methods: Agonist‐induced GPVI and CLEC‐2 signaling were assessed using aggregometry, immunoprecipitation and western blotting to determine the effects of R406 on platelet activation. Results: We demonstrate R406 to be a powerful inhibitor of Syk in human platelets. R406 abrogated shape change and aggregation induced by activation of GPVI and CLEC‐2, and reduced platelet spreading on fibrinogen. The inhibitory effect of R406 was associated with inhibition of tyrosine phosphorylation of signaling proteins that lay downstream of Syk for all three receptors, including PLCγ2. Strikingly, R406 markedly inhibited tyrosine phosphorylation of CLEC‐2 and Syk downstream of CLEC‐2 activation, whereas phosphorylation of Syk downstream of GPVI and integrin αIIbβ3 was unaffected. Conclusions: The inhibitory effect of R406 provides direct evidence of a role for Syk in GPVI, CLEC‐2 and integrin αIIbβ3 signaling in human platelets. Further, the results demonstrate a critical role for Syk in mediating tyrosine phosphorylation of CLEC‐2, suggesting a novel model in which both Src and Syk kinases regulate tyrosine phosphorylation of the C‐type lectin receptor leading to platelet activation.


Journal of Biological Chemistry | 2008

G6b-B Inhibits Constitutive and Agonist-induced Signaling by Glycoprotein VI and CLEC-2 *□

Jun Mori; Andrew C. Pearce; Jennifer C. Spalton; Beata Grygielska; Johannes A. Eble; Michael G. Tomlinson; Yotis A. Senis; Steve P. Watson

Platelets play an essential role in wound healing by forming thrombi that plug holes in the walls of damaged blood vessels. To achieve this, platelets express a diverse array of cell surface receptors and signaling proteins that induce rapid platelet activation. In this study we show that two platelet glycoprotein receptors that signal via an immunoreceptor tyrosine-based activation motif (ITAM) or an ITAM-like domain, namely the collagen receptor complex glycoprotein VI (GPVI)-FcR γ-chain and the C-type lectin-like receptor 2 (CLEC-2), respectively, support constitutive (i.e. agonist-independent) signaling in a cell line model using a nuclear factor of activated T-cells (NFAT) transcriptional reporter assay that can detect low level activation of phospholipase Cγ (PLCγ). Constitutive and agonist signaling by both receptors is dependent on Src and Syk family kinases, and is inhibited by G6b-B, a platelet immunoglobulin receptor that has two immunoreceptor tyrosine-based inhibitory motifs in its cytosolic tail. Mutation of the conserved tyrosines in the two immunoreceptor tyrosine-based inhibitory motifs prevents the inhibitory action of G6b-B. Interestingly, the inhibitory activity of G6b-B is independent of the Src homology 2 (SH2)-domain containing tyrosine phosphatases, SHP1 and SHP2, and the inositol 5′-phosphatase, SHIP. Constitutive signaling via Src and Syk tyrosine kinases is observed in platelets and is associated with tyrosine phosphorylation of GPVI-FcR γ-chain and CLEC-2. We speculate that inhibition of constitutive signaling through Src and Syk tyrosine kinases by G6b-B may help to prevent unwanted platelet activation.


Blood | 2014

CLEC-2 expression is maintained on activated platelets and on platelet microparticles

Eelo Gitz; Alice Y. Pollitt; Jerney J. Gitz-Francois; Osama Alshehri; Jun Mori; Samantha J. Montague; Gerard B. Nash; Michael R. Douglas; Elizabeth E. Gardiner; Robert K. Andrews; Christopher D. Buckley; Paul Harrison; Stephen P. Watson

The C-type lectin-like receptor CLEC-2 mediates platelet activation through a hem-immunoreceptor tyrosine-based activation motif (hemITAM). CLEC-2 initiates a Src- and Syk-dependent signaling cascade that is closely related to that of the 2 platelet ITAM receptors: glycoprotein (GP)VI and FcγRIIa. Activation of either of the ITAM receptors induces shedding of GPVI and proteolysis of the ITAM domain in FcγRIIa. In the present study, we generated monoclonal antibodies against human CLEC-2 and used these to measure CLEC-2 expression on resting and stimulated platelets and on other hematopoietic cells. We show that CLEC-2 is restricted to platelets with an average copy number of ∼2000 per cell and that activation of CLEC-2 induces proteolytic cleavage of GPVI and FcγRIIa but not of itself. We further show that CLEC-2 and GPVI are expressed on CD41+ microparticles in megakaryocyte cultures and in platelet-rich plasma, which are predominantly derived from megakaryocytes in healthy donors, whereas microparticles derived from activated platelets only express CLEC-2. Patients with rheumatoid arthritis, an inflammatory disease associated with increased microparticle production, had raised plasma levels of microparticles that expressed CLEC-2 but not GPVI. Thus, CLEC-2, unlike platelet ITAM receptors, is not regulated by proteolysis and can be used to monitor platelet-derived microparticles.


Journal of Biological Chemistry | 2006

Involvement of GSK-3β and DYRK1B in differentiation-inducing factor-3-induced phosphorylation of cyclin D1 in HeLa cells

Jun Mori; Etsuko Matsuzaki; Yutaka Watanabe; Masato Hirata; Yoshikazu Miwa; Sachio Morimoto; Toshiyuki Sasaguri

Differentiation-inducing factors (DIFs) are putative morphogens that induce cell differentiation in Dictyostelium discoideum. We previously reported that DIF-3 activates glycogen synthase kinase-3β (GSK-3β), resulting in the degradation of cyclin D1 in HeLa cells. In this study, we investigated the effect of DIF-3 on cyclin D1 mutants (R29Q, L32A, T286A, T288A, and T286A/T288A) to clarify the precise mechanisms by which DIF-3 degrades cyclin D1 in HeLa cells. We revealed that T286A, T288A, and T286A/T288A mutants were resistant to DIF-3-induced degradation compared with wild-type cyclin D1, indicating that the phosphorylation of Thr286 and Thr288 were critical for cyclin D1 degradation induced by DIF-3. Indeed, DIF-3 markedly elevated the phosphorylation level of cyclin D1, and mutations introduced to Thr286 and/or Thr288 prevented the phosphorylation induced by DIF-3. Depletion of endogenous GSK-3β and dual-specificity tyrosine phosphorylation regulated kinase 1B (DYRK1B) by RNA interference attenuated the DIF-3-induced cyclin D1 phosphorylation and degradation. The effect of DIF-3 on DYRK1B activity was examined and we found that DIF-3 also activated this kinase. Further, we found that not only GSK-3β but also DYRK1B modulates cyclin D1 subcellular localization by the phosphorylation of Thr288. These results suggest that DIF-3 induces degradation of cyclin D1 through the GSK-3β- and DYRK1B-mediated threonine phosphorylation in HeLa cells.


Science Signaling | 2012

Mice Lacking the ITIM-Containing Receptor G6b-B Exhibit Macrothrombocytopenia and Aberrant Platelet Function

Alexandra Mazharian; Ying Jie Wang; Jun Mori; Danai Bem; Brenda A. Finney; Silke Heising; Paul Gissen; James G. White; Michael C. Berndt; Elizabeth E. Gardiner; Bernhard Nieswandt; Michael R. Douglas; Robert A. A. Campbell; Steve P. Watson; Yotis A. Senis

An inhibitory receptor ensures that megakaryocytes produce proper numbers of functional platelets. Controlling Platelet Production Megakaryocytes reside in the bone marrow, where they produce platelets, cell fragments that form clots to prevent blood loss at sites of damage to the vasculature. Platelets and megakaryocytes share many activating receptors on their surface, but unlike platelets, megakaryocytes fail to become activated when exposed to components of the extracellular matrix. Mazharian et al. found that mice deficient in the immunoreceptor tyrosine–based inhibition motif–containing receptor G6b-B had fewer and larger platelets than did their wild-type counterparts. In addition, G6b-B–deficient mice exhibited increased bleeding in response to damage and had activated megakaryocytes, which resulted in the production of defective platelets. Together, these data suggest that G6b-B dampens activating signals in megakaryocytes to enable the generation of the appropriate number of functional platelets. Platelets are highly reactive cell fragments that adhere to exposed extracellular matrix (ECM) and prevent excessive blood loss by forming clots. Paradoxically, megakaryocytes, which produce platelets in the bone marrow, remain relatively refractory to the ECM-rich environment of the bone marrow despite having the same repertoire of receptors as platelets. These include the ITAM (immunoreceptor tyrosine–based activation motif)–containing collagen receptor complex, which consists of glycoprotein VI (GPVI) and the Fc receptor γ-chain, and the ITIM (immunoreceptor tyrosine–based inhibition motif)–containing receptor G6b-B. We showed that mice lacking G6b-B exhibited macrothrombocytopenia (reduced platelet numbers and the presence of enlarged platelets) and a susceptibility to bleeding as a result of aberrant platelet production and function. Platelet numbers were markedly reduced in G6b-B–deficient mice compared to those in wild-type mice because of increased platelet turnover. Furthermore, megakaryocytes in G6b-B–deficient mice showed enhanced metalloproteinase production, which led to increased shedding of cell-surface receptors, including GPVI and GPIbα. In addition, G6b-B–deficient megakaryocytes exhibited reduced integrin-mediated functions and defective formation of proplatelets, the long filamentous projections from which platelets bud off. Together, these findings establish G6b-B as a major inhibitory receptor regulating megakaryocyte activation, function, and platelet production.


Blood | 2013

Megakaryocyte-specific deletion of the protein-tyrosine phosphatases Shp1 and Shp2 causes abnormal megakaryocyte development, platelet production and function

Alexandra Mazharian; Jun Mori; Ying-Jie Wang; Silke Heising; Benjamin G. Neel; Stephen P. Watson; Yotis A. Senis

The SH2 domain-containing protein-tyrosine phosphatases Shp1 and Shp2 have been implicated in regulating signaling from a variety of platelet and megakaryocyte receptors. In this study, we investigate the functions of Shp1 and Shp2 in megakaryocytes and platelets. Megakaryocyte/platelet (MP)-specific deletion of Shp1 in mice resulted in platelets being less responsive to collagen-related peptide due to reduced GPVI expression and signaling via the Src family kinase (SFK)-Syk-PLCγ2 pathway, and fibrinogen due to reduced SFK activity. By contrast, deletion of Shp2 in the MP lineage resulted in macrothrombocytopenia and platelets being hyper-responsive to anti-CLEC-2 antibody and fibrinogen. Shp1- and Shp2-deficient megakaryocytes had partial blocks at 2N/4N ploidy; however, only the latter exhibited reduced proplatelet formation, thrombopoietin, and integrin signaling. Mice deficient in both Shp1 and Shp2 were severely macrothrombocytopenic and had reduced platelet surface glycoprotein expression, including GPVI, αIIbβ3, and GPIbα. Megakaryocytes from these mice were blocked at 2N/4N ploidy and did not survive ex vivo. Deletion of the immunoreceptor tyrosine-based inhibition motif-containing receptor G6b-B in the MP lineage phenocopied multiple features of Shp1/2-deficient mice, suggesting G6b-B is a critical regulator of Shp1 and Shp2. This study establishes Shp1 and Shp2 as major regulators of megakaryocyte development, platelet production, and function.


Journal of Thrombosis and Haemostasis | 2012

Distinct and overlapping functional roles of Src family kinases in mouse platelets

Sonia Severin; Ca Nash; Jun Mori; Y. Zhao; C. Abram; Clifford A. Lowell; Yotis A. Senis; Steve P. Watson

Summary.  Background and objectives:  Src family kinases (SFKs) play a critical role in initiating and propagating signals in platelets. The aims of this study were to quantitate SFK members present in platelets and to analyze their contribution to platelet regulation using glycoprotein VI (GPVI) and intregrin αIIbβ3, and in vivo.

Collaboration


Dive into the Jun Mori's collaboration.

Top Co-Authors

Avatar

Yotis A. Senis

University of Birmingham

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Silke Heising

University of Birmingham

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge