Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Blood ; 135(18): 1574-1587, 2020 04 30.
Article in English | MEDLINE | ID: mdl-32016283

ABSTRACT

The Src family kinases (SFKs) Src, Lyn, and Fyn are essential for platelet activation and also involved in megakaryocyte (MK) development and platelet production. Platelet SFKs are inhibited by C-terminal Src kinase (Csk), which phosphorylates a conserved tyrosine in their C-terminal tail, and are activated by the receptor-type tyrosine phosphatase PTPRJ (CD148, DEP-1), which dephosphorylates the same residue. Deletion of Csk and PTPRJ in the MK lineage in mice results in increased SFK activity, but paradoxically hypoactive platelets resulting from negative feedback mechanisms, including upregulation of Csk homologous kinase (Chk) expression. Here, we investigate the role of Chk in platelets, functional redundancy with Csk, and the physiological consequences of ablating Chk, Csk, and PTPRJ in mice. Platelet count was normal in Chk knockout (KO) mice, reduced by 92% in Chk;Csk double KO (DKO) mice, and partially rescued in Chk;Csk;Ptprj triple KO (TKO) mice. Megakaryocyte numbers were significantly increased in both DKO and TKO mice. Phosphorylation of the inhibitory tyrosine of SFKs was almost completely abolished in DKO platelets, which was partially rescued in Src and Fyn in TKO platelets. This residual phosphorylation was abolished by Src inhibitors, revealing an unexpected mechanism in which SFKs autoinhibit their activity by phosphorylating their C-terminal tyrosine residues. We demonstrate that reduced inhibitory phosphorylation of SFKs leads to thrombocytopenia, with Csk being the dominant inhibitor in platelets and Chk having an auxiliary role. PTPRJ deletion in addition to Chk and Csk ameliorates the extent of thrombocytopenia, suggesting targeting it may have therapeutic benefits in such conditions.


Subject(s)
Blood Platelets/metabolism , CSK Tyrosine-Protein Kinase/metabolism , Proto-Oncogene Proteins pp60(c-src)/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 3/metabolism , Animals , Biomarkers , Bleeding Time , CSK Tyrosine-Protein Kinase/genetics , Immunohistochemistry , Mice , Mice, Knockout , Models, Biological , Phosphorylation , Platelet Activation , Platelet Count , Platelet Function Tests , Protein Binding , Proto-Oncogene Proteins pp60(c-src)/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 3/genetics , src-Family Kinases/genetics , src-Family Kinases/metabolism
2.
Blood ; 134(25): 2304-2317, 2019 12 19.
Article in English | MEDLINE | ID: mdl-31562133

ABSTRACT

Src homology 2 domain-containing phosphatase 2 (SHP2), encoded by the PTPN11 gene, is a ubiquitous protein tyrosine phosphatase that is a critical regulator of signal transduction. Germ line mutations in the PTPN11 gene responsible for catalytic gain or loss of function of SHP2 cause 2 disorders with multiple organ defects: Noonan syndrome (NS) and NS with multiple lentigines (NSML), respectively. Bleeding anomalies have been frequently reported in NS, but causes remain unclear. This study investigates platelet activation in patients with NS and NSML and in 2 mouse models carrying PTPN11 mutations responsible for these 2 syndromes. Platelets from NS mice and patients displayed a significant reduction in aggregation induced by low concentrations of GPVI and CLEC-2 agonists and a decrease in thrombus growth on a collagen surface under arterial shear stress. This was associated with deficiencies in GPVI and αIIbß3 integrin signaling, platelet secretion, and thromboxane A2 generation. Similarly, arterial thrombus formation was significantly reduced in response to a local carotid injury in NS mice, associated with a significant increase in tail bleeding time. In contrast, NSML mouse platelets exhibited increased platelet activation after GPVI and CLEC-2 stimulation and enhanced platelet thrombotic phenotype on collagen matrix under shear stress. Blood samples from NSML patients also showed a shear stress-dependent elevation of platelet responses on collagen matrix. This study brings new insights into the understanding of SHP2 function in platelets, points to new thrombopathies linked to platelet signaling defects, and provides important information for the medical care of patients with NS in situations involving risk of bleeding.


Subject(s)
Blood Platelets/enzymology , Germ-Line Mutation , Noonan Syndrome/enzymology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Signal Transduction , Animals , Blood Platelets/pathology , Humans , Mice , Mice, Mutant Strains , Noonan Syndrome/pathology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics
3.
Blood ; 133(4): 331-343, 2019 01 24.
Article in English | MEDLINE | ID: mdl-30429161

ABSTRACT

Conditional knockout (KO) mouse models are invaluable for elucidating the physiological roles of platelets. The Platelet factor 4-Cre recombinase (Pf4-Cre) transgenic mouse is the current model of choice for generating megakaryocyte/platelet-specific KO mice. Platelets and leukocytes work closely together in a wide range of disease settings, yet the specific contribution of platelets to these processes remains unclear. This is partially a result of the Pf4-Cre transgene being expressed in a variety of leukocyte populations. To overcome this issue, we developed a Gp1ba-Cre transgenic mouse strain in which Cre expression is driven by the endogenous Gp1ba locus. By crossing Gp1ba-Cre and Pf4-Cre mice to the mT/mG dual-fluorescence reporter mouse and performing a head-to-head comparison, we demonstrate more stringent megakaryocyte lineage-specific expression of the Gp1ba-Cre transgene. Broader tissue expression was observed with the Pf4-Cre transgene, leading to recombination in many hematopoietic lineages, including monocytes, macrophages, granulocytes, and dendritic and B and T cells. Direct comparison of phenotypes of Csk, Shp1, or CD148 conditional KO mice generated using either the Gp1ba-Cre or Pf4-Cre strains revealed similar platelet phenotypes. However, additional inflammatory and immunological anomalies were observed in Pf4-Cre-generated KO mice as a result of nonspecific deletion in other hematopoietic lineages. By excluding leukocyte contributions to phenotypes, the Gp1ba-Cre mouse will advance our understanding of the role of platelets in inflammation and other pathophysiological processes in which platelet-leukocyte interactions are involved.


Subject(s)
Blood Platelets/metabolism , Integrases/metabolism , Leukocytes/metabolism , Platelet Glycoprotein GPIb-IX Complex/metabolism , Agglutination , Animals , Bone Marrow Cells/cytology , CSK Tyrosine-Protein Kinase , Cell Lineage , Cell Size , Gene Targeting , Homeostasis , Lymphocyte Count , Megakaryocytes/cytology , Megakaryocytes/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Models, Animal , Phenotype , Platelet Aggregation , Platelet Factor 4/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 3/metabolism , Recombination, Genetic/genetics , Spleen/cytology , src-Family Kinases/metabolism
4.
Blood ; 132(13): 1413-1425, 2018 09 27.
Article in English | MEDLINE | ID: mdl-29891536

ABSTRACT

The immunoreceptor tyrosine-based inhibitory motif (ITIM)-containing receptor G6b-B has emerged as a key regulator of platelet homeostasis. However, it remains unclear how it mediates its effects. Tyrosine phosphorylation of ITIM and immunoreceptor tyrosine-based switch motif (ITSM) within the cytoplasmic tail of G6b-B provides a docking site for Src homology 2 domain-containing protein-tyrosine phosphatases Shp1 and Shp2, which are also critical regulators of platelet production and function. In this study, we investigate the physiological consequences of uncoupling G6b-B from Shp1 and Shp2. To address this, we generated a transgenic mouse model expressing a mutant form of G6b-B in which tyrosine residues 212 and 238 within ITIM and ITSM were mutated to phenylalanine. Mice homozygous for the mutation (G6b-B diY/F) were macrothrombocytopenic, as a result of the reduction in platelet production, and had large clusters of megakaryocytes and myelofibrosis at sites of hematopoiesis, similar to those observed in G6b-deficient mice and patients. Platelets from G6b-B diY/F mice were hyporesponsive to collagen, as a result of the significant reduction in the expression of the immunoreceptor tyrosine-based activation motif (ITAM)-containing collagen receptor complex GPVI-FcR γ-chain, as well as thrombin, which could be partially rescued by costimulating the platelets with adenosine diphosphate. In contrast, platelets from G6b-B diY/F, G6b KO, and megakaryocyte-specific Shp2 KO mice were hyperresponsive to antibody-mediated cross-linking of the hemi-ITAM-containing podoplanin receptor CLEC-2, suggesting that G6b-B inhibits CLEC-2-mediated platelet activation through Shp2. Findings from this study demonstrate that G6b-B must engage with Shp1 and Shp2 to mediate its regulatory effects on platelet homeostasis.


Subject(s)
Blood Platelets/pathology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Receptors, Immunologic/metabolism , Thrombocytopenia/metabolism , Animals , Binding Sites , Blood Platelets/metabolism , Homeostasis , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Molecular , Phosphorylation , Point Mutation , Protein Interaction Maps , Protein Tyrosine Phosphatase, Non-Receptor Type 11/chemistry , Protein Tyrosine Phosphatase, Non-Receptor Type 6/chemistry , Receptors, Immunologic/chemistry , Receptors, Immunologic/genetics , Signal Transduction , Thrombocytopenia/genetics , Thrombocytopenia/pathology , src Homology Domains
5.
Blood ; 131(10): 1122-1144, 2018 03 08.
Article in English | MEDLINE | ID: mdl-29301754

ABSTRACT

Src family kinases (SFKs) coordinate the initiating and propagating activation signals in platelets, but it remains unclear how they are regulated. Here, we show that ablation of C-terminal Src kinase (Csk) and receptor-like protein tyrosine-phosphatase CD148 in mice results in a dramatic increase in platelet SFK activity, demonstrating that these proteins are essential regulators of platelet reactivity. Paradoxically, Csk/CD148-deficient mice exhibit reduced in vivo and ex vivo thrombus formation and increased bleeding following injury rather than a prothrombotic phenotype. This is a consequence of multiple negative feedback mechanisms, including downregulation of the immunoreceptor tyrosine-based activation motif (ITAM)- and hemi-ITAM-containing receptors glycoprotein VI (GPVI)-Fc receptor (FcR) γ-chain and CLEC-2, respectively and upregulation of the immunoreceptor tyrosine-based inhibition motif (ITIM)-containing receptor G6b-B and its interaction with the tyrosine phosphatases Shp1 and Shp2. Results from an analog-sensitive Csk mouse model demonstrate the unconventional role of SFKs in activating ITIM signaling. This study establishes Csk and CD148 as critical molecular switches controlling the thrombotic and hemostatic capacity of platelets and reveals cell-intrinsic mechanisms that prevent pathological thrombosis from occurring.


Subject(s)
Blood Platelets/metabolism , Homeostasis , Thrombosis/metabolism , src-Family Kinases/metabolism , Amino Acid Motifs , Animals , Blood Platelets/pathology , CSK Tyrosine-Protein Kinase , Mice , Mice, Knockout , Platelet Membrane Glycoproteins/genetics , Platelet Membrane Glycoproteins/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 3/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 3/metabolism , Thrombosis/genetics , src-Family Kinases/genetics
6.
Blood ; 132(13): 1399-1412, 2018 09 27.
Article in English | MEDLINE | ID: mdl-29898956

ABSTRACT

Unlike primary myelofibrosis (PMF) in adults, myelofibrosis in children is rare. Congenital (inherited) forms of myelofibrosis (cMF) have been described, but the underlying genetic mechanisms remain elusive. Here we describe 4 families with autosomal recessive inherited macrothrombocytopenia with focal myelofibrosis due to germ line loss-of-function mutations in the megakaryocyte-specific immunoreceptor tyrosine-based inhibitory motif (ITIM)-containing receptor G6b-B (G6b, C6orf25, or MPIG6B). Patients presented with a mild-to-moderate bleeding diathesis, macrothrombocytopenia, anemia, leukocytosis and atypical megakaryocytes associated with a distinctive, focal, perimegakaryocytic pattern of bone marrow fibrosis. In addition to identifying the responsible gene, the description of G6b-B as the mutated protein potentially implicates aberrant G6b-B megakaryocytic signaling and activation in the pathogenesis of myelofibrosis. Targeted insertion of human G6b in mice rescued the knockout phenotype and a copy number effect of human G6b-B expression was observed. Homozygous knockin mice expressed 25% of human G6b-B and exhibited a marginal reduction in platelet count and mild alterations in platelet function; these phenotypes were more severe in heterozygous mice that expressed only 12% of human G6b-B. This study establishes G6b-B as a critical regulator of platelet homeostasis in humans and mice. In addition, the humanized G6b mouse will provide an invaluable tool for further investigating the physiological functions of human G6b-B as well as testing the efficacy of drugs targeting this receptor.


Subject(s)
Loss of Function Mutation , Primary Myelofibrosis/congenital , Receptors, Immunologic/genetics , Thrombocytopenia/congenital , Adolescent , Adult , Animals , Blood Platelets/metabolism , Blood Platelets/pathology , Child , Child, Preschool , Female , Gene Knock-In Techniques , Humans , Infant , Male , Megakaryocytes/metabolism , Megakaryocytes/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Pedigree , Primary Myelofibrosis/genetics , Primary Myelofibrosis/pathology , Thrombocytopenia/genetics , Thrombocytopenia/pathology , Young Adult
7.
Arterioscler Thromb Vasc Biol ; 37(5): 823-835, 2017 05.
Article in English | MEDLINE | ID: mdl-28336561

ABSTRACT

OBJECTIVE: Leukocyte-associated immunoglobulin-like receptor-1 (LAIR-1) is a collagen receptor that belongs to the inhibitory immunoreceptor tyrosine-based inhibition motif-containing receptor family. It is an inhibitor of signaling via the immunoreceptor tyrosine-based activation motif-containing collagen receptor complex, glycoprotein VI-FcRγ-chain. It is expressed on hematopoietic cells, including immature megakaryocytes, but is not detectable on platelets. Although the inhibitory function of LAIR-1 has been described in leukocytes, its physiological role in megakaryocytes and in particular in platelet formation has not been explored. In this study, we investigate the role of LAIR-1 in megakaryocyte development and platelet production by generating LAIR-1-deficient mice. APPROACH AND RESULTS: Mice lacking LAIR-1 exhibit a significant increase in platelet counts, a prolonged platelet half-life in vivo, and increased proplatelet formation in vitro. Interestingly, platelets from LAIR-1-deficient mice exhibit an enhanced reactivity to collagen and the glycoprotein VI-specific agonist collagen-related peptide despite not expressing LAIR-1, and mice showed enhanced thrombus formation in the carotid artery after ferric chloride injury. Targeted deletion of LAIR-1 in mice results in an increase in signaling downstream of the glycoprotein VI-FcRγ-chain and integrin αIIbß3 in megakaryocytes because of enhanced Src family kinase activity. CONCLUSIONS: Findings from this study demonstrate that ablation of LAIR-1 in megakaryocytes leads to increased Src family kinase activity and downstream signaling in response to collagen that is transmitted to platelets, rendering them hyper-reactive specifically to agonists that signal through Syk tyrosine kinases, but not to G-protein-coupled receptors.


Subject(s)
Blood Platelets/metabolism , Megakaryocytes/metabolism , Platelet Activation , Receptors, Immunologic/deficiency , Thrombocytosis/blood , Thrombosis/blood , Animals , Blood Platelets/drug effects , Carrier Proteins/pharmacology , Cells, Cultured , Chlorides , Disease Models, Animal , Enzyme Activation , Ferric Compounds , Genetic Predisposition to Disease , Megakaryocytes/drug effects , Mice, Knockout , Peptides/pharmacology , Phenotype , Platelet Activation/drug effects , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Platelet Membrane Glycoproteins/agonists , Platelet Membrane Glycoproteins/metabolism , Receptors, IgG/blood , Receptors, Immunologic/genetics , Signal Transduction/drug effects , Thrombocytosis/genetics , Thrombosis/chemically induced , Thrombosis/genetics , src-Family Kinases/blood
8.
Blood ; 125(5): 747-8, 2015 Jan 29.
Article in English | MEDLINE | ID: mdl-25634614

ABSTRACT

In this issue of Blood, Bender et al provide compelling evidence that the motor protein cytoplasmic dynein provides the necessary force for microtubule sliding and proplatelet elongation from megakaryocytes.


Subject(s)
Blood Platelets/metabolism , Cytoplasmic Dyneins/metabolism , Megakaryocytes/metabolism , Microtubules/metabolism , Tubulin/metabolism , Animals
9.
Blood ; 124(13): 2013-24, 2014 Sep 25.
Article in English | MEDLINE | ID: mdl-25115887

ABSTRACT

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 αIIbß3, 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.


Subject(s)
Platelet Activation/physiology , src-Family Kinases/metabolism , Animals , Gene Expression , Hemostasis , Humans , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Platelet Glycoprotein GPIb-IX Complex/metabolism , Proto-Oncogene Proteins c-fyn/metabolism , Signal Transduction , src-Family Kinases/chemistry , src-Family Kinases/genetics
10.
Chembiochem ; 16(11): 1680-8, 2015 Jul 27.
Article in English | MEDLINE | ID: mdl-26062886

ABSTRACT

Lifeact is a 17-residue peptide that can be employed in cell microscopy as a probe for F-actin when fused to fluorescent proteins, but therefore is not suitable for all cell types. We have conjugated fluorescently labelled Lifeact to three different cell-penetrating systems (a myristoylated carrier (myr), the pH low insertion peptide (pHLIP) and the cationic peptide TAT) as a strategy to deliver Lifeact into cells and developed new tools for actin staining with improved synthetic accessibility and low toxicity, focusing on their suitability in platelets and megakaryocytes. Using confocal microscopy, we characterised the cell distribution of the new hybrids in fixed cells, and found that both myr- and pHLIP-Lifeact conjugates provide efficient actin staining upon cleavage of Lifeact from the carriers, without affecting cell spreading. This new approach could facilitate the design of new tools for actin visualisation.


Subject(s)
Actins/metabolism , Blood Platelets/metabolism , Cell-Penetrating Peptides/metabolism , Fluorescent Dyes/metabolism , Megakaryocytes/metabolism , Amino Acid Sequence , Cell-Penetrating Peptides/chemical synthesis , Cell-Penetrating Peptides/chemistry , Drug Design , Fluorescent Dyes/chemical synthesis , Fluorescent Dyes/chemistry , Humans , Hydrogen-Ion Concentration , Molecular Sequence Data , Staining and Labeling
11.
Blood ; 121(20): 4205-20, 2013 May 16.
Article in English | MEDLINE | ID: mdl-23509158

ABSTRACT

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, αIIbß3, 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.


Subject(s)
Blood Platelets/physiology , Gene Deletion , Megakaryocytes/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Thrombopoiesis/genetics , Animals , Blood Platelets/metabolism , Cells, Cultured , Megakaryocyte Progenitor Cells/metabolism , Megakaryocyte Progenitor Cells/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Organ Specificity/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/physiology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/physiology , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases/physiology , Thrombopoiesis/physiology
12.
Blood ; 122(5): 791-802, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23775711

ABSTRACT

Human megakaryocytes (MKs) release trillions of platelets each day into the circulation to maintain normal homeostatic platelet levels. We have previously shown that extracellular sphingosine 1-phosphate (S1P) plays a key role in thrombopoiesis via its receptor S1pr1. In addition to its role as an extracellular mediator, S1P can also function as a second messenger in the intracellular compartment. Although signaling via intracellular S1P is involved in various cellular processes, a role in thrombopoiesis has not been examined. Sphingosine kinases are the key enzymes that produce intracellular S1P. Here we report that sphingosine kinase 2 (Sphk2) is the major messenger RNA species present in MKs. Sphk2 predominantly localizes to the nucleus and is the major source of intracellular S1P in MKs. Loss of Sphk2 significantly reduced intracellular S1P in MKs and downregulated the expression and activity of Src family kinases (SFKs). Loss of Sphk2 and inhibition of SFK activity resulted in defective intravascular proplatelet shedding, the final stage of thrombopoiesis. Correspondingly, mice lacking Sphk2 in the hematopoietic system display thrombocytopenia. Together, our data suggest that Sphk2 provides the source of intracellular S1P that controls thrombopoiesis, which is associated with SFK expression and activity in MKs.


Subject(s)
Blood Platelets/physiology , Lysophospholipids/metabolism , Phosphotransferases (Alcohol Group Acceptor)/physiology , Sphingosine/analogs & derivatives , Animals , Blood Platelets/metabolism , Cell Differentiation/genetics , Cell Survival/genetics , Homeostasis/genetics , Homeostasis/physiology , Intracellular Space/metabolism , Megakaryocytes/metabolism , Megakaryocytes/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Sphingosine/metabolism , Thrombopoiesis/genetics , Thrombopoietin/blood , src-Family Kinases/genetics , src-Family Kinases/metabolism
13.
Blood ; 122(23): 3787-97, 2013 Nov 28.
Article in English | MEDLINE | ID: mdl-24085768

ABSTRACT

The principal morbidity and mortality in patients with essential thrombocythemia (ET) and polycythemia rubra vera (PV) stems from thrombotic events. Most patients with ET/PV harbor a JAK2V617F mutation, but its role in the thrombotic diathesis remains obscure. Platelet function studies in patients are difficult to interpret because of interindividual heterogeneity, reflecting variations in the proportion of platelets derived from the malignant clone, differences in the presence of additional mutations, and the effects of medical treatments. To circumvent these issues, we have studied a JAK2V617F knock-in mouse model of ET in which all megakaryocytes and platelets express JAK2V617F at a physiological level, equivalent to that present in human ET patients. We show that, in addition to increased differentiation, JAK2V617F-positive megakaryocytes display greater migratory ability and proplatelet formation. We demonstrate in a range of assays that platelet reactivity to agonists is enhanced, with a concomitant increase in platelet aggregation in vitro and a reduced duration of bleeding in vivo. These data suggest that JAK2V617F leads to intrinsic changes in both megakaryocyte and platelet biology beyond an increase in cell number. In support of this hypothesis, we identify multiple differentially expressed genes in JAK2V617F megakaryocytes that may underlie the observed biological differences.


Subject(s)
Blood Platelets/enzymology , Janus Kinase 2/blood , Janus Kinase 2/genetics , Mutant Proteins/blood , Mutant Proteins/genetics , Mutation , Thrombocythemia, Essential/blood , Thrombocythemia, Essential/genetics , Animals , Blood Platelets/pathology , Disease Models, Animal , Gene Expression , Humans , Megakaryocytes/enzymology , Megakaryocytes/pathology , Mice , Mice, Transgenic , Platelet Aggregation/genetics , Recombinant Proteins/blood , Recombinant Proteins/genetics , Thrombocythemia, Essential/enzymology , Thrombopoiesis/genetics
14.
bioRxiv ; 2024 May 14.
Article in English | MEDLINE | ID: mdl-38798354

ABSTRACT

Platelets are highly reactive fragments of megakaryocytes that play a fundamental role in thrombosis and hemostasis. Predictably, all conventional anti-platelet therapies elicit bleeding, raising the question whether the thrombotic activity of platelets can be targeted separately. In this study, we describe a novel approach of inhibiting platelet activation through the use of bispecific single-chain variable fragments (bi-scFvs), termed cis-acting platelet receptor inhibitors (CAPRIs) that harness the immunoreceptor tyrosine-based inhibition motif (ITIM)-containing co-inhibitory receptor G6b-B (G6B) to suppress immunoreceptor tyrosine-based (ITAM)-containing receptor-mediated platelet activation. CAPRI-mediated hetero-clustering of G6B with either the ITAM-containing GPVI-FcR γ-chain complex or FcγRIIA (CD32A) inhibited collagen- or immune complex-induced platelet aggregation. G6B-GPVI CAPRIs strongly and specifically inhibited thrombus formation on collagen under arterial shear, whereas G6B-CD32A CAPRI strongly and specifically inhibited thrombus formation to heparin-induced thrombocytopenia, vaccine-induced thrombotic thrombocytopenia and antiphospholipid syndrome complexes on Von Willebrand Factor-coated surfaces and photochemical-injured endothelial cells under arterial shear. Our findings provide proof-of-concept that CAPRIs are highly effective at inhibiting ITAM receptor-mediated platelet activation, laying the foundation for a novel family of anti-thrombotic therapeutics with potentially improved efficacy and fewer bleeding outcomes compared with current anti-platelet therapies. .

15.
Blood ; 117(19): 5198-206, 2011 May 12.
Article in English | MEDLINE | ID: mdl-21385851

ABSTRACT

Dasatinib is a novel, potent, ATP-competitive inhibitor of Bcr-Abl, cKIT, and Src family kinases that exhibits efficacy in patients with imatinib-resistant chronic myelogenous leukemia. Dasatinib treatment is associated with mild thrombocytopenia and an increased risk of bleeding, but its biological effect on megakaryocytopoiesis and platelet production is unknown. In this study, we show that dasatinib causes mild thrombocytopenia in mice without altering platelet half-life, suggesting that it inhibits platelet formation. Conversely, the number of megakaryocytes (MKs) in the bone marrow of dasatinib-treated mice was increased and the ploidy of MKs derived from bone marrow progenitor cells in vitro was elevated in the presence of dasatinib. Furthermore, a significant delay in platelet recovery after immune-induced thrombocytopenia was observed in dasatinib-treated mice even though the number of MKs in the bone marrow was increased relative to controls at all time points. Interestingly, the migration of MKs toward a gradient of stromal cell-derived factor 1α (SDF1α) and the formation of proplatelets in vitro were abolished by dasatinib. We propose that dasatinib causes thrombocytopenia as a consequence of ineffective thrombopoiesis, promoting MK differentiation but also impairing MK migration and proplatelet formation.


Subject(s)
Blood Platelets/drug effects , Megakaryocytes/drug effects , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Thiazoles/pharmacology , Thrombopoiesis/drug effects , Animals , Blood Platelets/cytology , Cell Differentiation/drug effects , Cell Movement/drug effects , Cell Separation , Dasatinib , Flow Cytometry , Immunohistochemistry , Megakaryocytes/cytology , Mice , Mice, Inbred C57BL , Ploidies , Thrombocytopenia/chemically induced
16.
Res Pract Thromb Haemost ; 7(7): 102209, 2023 Oct.
Article in English | MEDLINE | ID: mdl-38077809

ABSTRACT

Background: The contribution of platelets in thrombosis within microcirculation has been extensively documented in the literature. We previously showed, in vivo, that platelet activation revealed by intracellular calcium mobilization was a crucial step in the growth of thrombi following laser-induced injury, a model of thromboinflammation. Objective: Our goal was to investigate the extent of platelet activation and the spatial distribution of platelets throughout a growing thrombus. Methods: We employed a multimodal, correlative microscopy approach and computational biology to study the state of platelets on a growing thrombus obtained after a laser injury. Results: We observed a reversible intracellular platelet calcium mobilization that correlates with the time a platelet resides during thrombus growth. Our bioinformatics analysis displayed the following 3 distinct platelet subpopulations resident within a thrombus: (1) resting, (2) partially activated, and (3) "fully" activated platelets. The spatial distribution of the platelet subpopulations in the thrombus creates a double gradient in both the transversal and longitudinal axis, with the maximal percentage of fully activated platelets close to the site of injury. However, these activated platelets did not express negative phospholipids. The injured endothelium was identified to play a vital role in activating the blood coagulation cascade in this model of thrombosis. Conclusion: Following a laser-induced injury, thrombi are formed by a gradient of activated platelets from the injury site to the periphery of the thrombus. These different activation states of platelets throughout the thrombi regulate the biomechanics of the thrombus. The injured endothelium, rather than platelets, was identified to play a key role in the activation of the blood coagulation cascade in this model of thromboinflammation.

17.
Blood Adv ; 7(1): 46-59, 2023 01 10.
Article in English | MEDLINE | ID: mdl-36269841

ABSTRACT

Mice lacking the immunoreceptor tyrosine-based inhibition motif-containing co-inhibitory receptor G6b-B (Mpig6b, G6b knockout, KO) are born with a complex megakaryocyte (MK) per platelet phenotype, characterized by severe macrothrombocytopenia, expansion of the MK population, and focal myelofibrosis in the bone marrow and spleen. Platelets are almost completely devoid of the glycoprotein VI (GPVI)-FcRγ-chain collagen receptor complex, have reduced collagen integrin α2ß1, elevated Syk tyrosine kinase activity, and a subset has increased surface immunoglobulins. A similar phenotype was recently reported in patients with null and loss-of-function mutations in MPIG6B. To better understand the cause and treatment of this pathology, we used pharmacological- and genetic-based approaches to rescue platelet counts and function in G6b KO mice. Intravenous immunoglobulin resulted in a transient partial recovery of platelet counts, whereas immune deficiency did not affect platelet counts or receptor expression in G6b KO mice. Syk loss-of-function (R41A) rescued macrothrombocytopenia, GPVI and α2ß1 expression in G6b KO mice, whereas treatment with the Syk kinase inhibitor BI1002494 partially rescued platelet count but had no effect on GPVI and α2ß1 expression or bleeding. The Src family kinase inhibitor dasatinib was not beneficial in G6b KO mice. In contrast, treatment with the thrombopoietin mimetic romiplostim rescued thrombocytopenia, GPVI expression, and platelet reactivity to collagen, suggesting that it may be a promising therapeutic option for patients lacking functional G6b-B. Intriguingly, GPVI and α2ß1 expression were significantly downregulated in romiplostim-treated wild-type mice, whereas GPVI was upregulated in romiplostim-treated G6b KO mice, suggesting a cell intrinsic feedback mechanism that autoregulates platelet reactivity depending on physiological needs.


Subject(s)
Blood Platelets , Thrombocytopenia , Mice , Animals , Blood Platelets/metabolism , Megakaryocytes/metabolism , Thrombocytopenia/genetics , src-Family Kinases/metabolism , Collagen/metabolism
18.
Blood ; 116(5): 793-800, 2010 Aug 05.
Article in English | MEDLINE | ID: mdl-20457868

ABSTRACT

Migration of megakaryocytes (MKs) from the proliferative osteoblastic niche to the capillary-rich vascular niche is essential for proplatelet formation and platelet release. In this study, we explore the role of surface glycoprotein receptors and signaling proteins in regulating MK migration and platelet recovery after immune-induced thrombocytopenia. We show that spreading and migration of mouse primary bone marrow-derived MKs on a fibronectin matrix are abolished by the Src family kinases inhibitor PP1, the Syk kinase inhibitor R406 and the integrin alphaIIbbeta3 antagonist lotrafiban. We also demonstrate that these responses are inhibited in primary phospholipase C gamma2 (PLCgamma2)-deficient MKs. Conversely, MK spreading and migration were unaltered in the absence of the collagen receptor, the glycoprotein VI-FcRgamma-chain complex. We previously reported a correlation between a defect in MK migration and platelet recovery in the absence of platelet endothelial cell adhesion molecule-1 and the tyrosine phosphatase CD148. This correlation also holds for mice deficient in PLCgamma2. This study identifies a model in which integrin signaling via Src family kinases and Syk kinase to PLCgamma2 is required for MK spreading, migration, and platelet formation.


Subject(s)
Blood Platelets/cytology , Intracellular Signaling Peptides and Proteins/physiology , Megakaryocytes/cytology , Phospholipase C gamma/physiology , Protein-Tyrosine Kinases/physiology , Proto-Oncogene Proteins pp60(c-src)/physiology , Thrombopoiesis/physiology , Animals , Benzodiazepines/pharmacology , Cell Movement/drug effects , Cell Movement/physiology , Cell Shape , Cells, Cultured/cytology , Cells, Cultured/enzymology , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Megakaryocytes/enzymology , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxazines/pharmacology , Phospholipase C gamma/antagonists & inhibitors , Phospholipase C gamma/deficiency , Phosphorylation/drug effects , Piperidines/pharmacology , Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors , Platelet Glycoprotein GPIIb-IIIa Complex/physiology , Protein Kinase Inhibitors/pharmacology , Protein Processing, Post-Translational/drug effects , Protein-Tyrosine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins pp60(c-src)/antagonists & inhibitors , Purpura, Thrombocytopenic, Idiopathic/chemically induced , Purpura, Thrombocytopenic, Idiopathic/pathology , Pyrazoles/pharmacology , Pyridines/pharmacology , Pyrimidines/pharmacology , Signal Transduction/drug effects , Signal Transduction/physiology , Syk Kinase
19.
Blood ; 113(20): 4942-54, 2009 May 14.
Article in English | MEDLINE | ID: mdl-19246339

ABSTRACT

Platelets play a fundamental role in hemostasis and thrombosis. They are also involved in pathologic conditions resulting from blocked blood vessels, including myocardial infarction and ischemic stroke. Platelet adhesion, activation, and aggregation at sites of vascular injury are regulated by a diverse repertoire of tyrosine kinase-linked and G protein-coupled receptors. Src family kinases (SFKs) play a central role in initiating and propagating signaling from several platelet surface receptors; however, the underlying mechanism of how SFK activity is regulated in platelets remains unclear. CD148 is the only receptor-like protein tyrosine phosphatase identified in platelets to date. In the present study, we show that mutant mice lacking CD148 exhibited a bleeding tendency and defective arterial thrombosis. Basal SFK activity was found to be markedly reduced in CD148-deficient platelets, resulting in a global hyporesponsiveness to agonists that signal through SFKs, including collagen and fibrinogen. G protein-coupled receptor responses to thrombin and other agonists were also marginally reduced. These results highlight CD148 as a global regulator of platelet activation and a novel antithrombotic drug target.


Subject(s)
Platelet Activation/genetics , Thrombosis/genetics , Animals , Antigens, Surface/metabolism , Blood Platelets/metabolism , Cell Adhesion/drug effects , Cells, Cultured , Fibrinogen/pharmacology , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Platelet Aggregation/drug effects , Platelet Membrane Glycoproteins/metabolism , Platelet Membrane Glycoproteins/physiology , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases/physiology , Receptor-Like Protein Tyrosine Phosphatases, Class 3/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 3/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 3/physiology , Receptors, IgG/genetics , Signal Transduction/immunology
20.
J Bone Miner Res ; 36(4): 803-813, 2021 04.
Article in English | MEDLINE | ID: mdl-33434328

ABSTRACT

Patients with chronic myelofibrosis often suffer from osteosclerosis, which is associated with bone pain and may lead to bone marrow failure. The pathogenesis of myelofibrosis is linked to aberrant megakaryocyte development and function. Null and loss-of-function mutations in MPIG6B, which codes for the inhibitory heparan sulfate receptor G6b-B, result in severe macrothrombocytopenia, large megakaryocyte clusters, and focal primary myelofibrosis in mice and humans. We investigated the development of osteosclerosis in Mpig6b null (Mpig6b-/- ) mice. Although male and female Mpig6b-/- mice presented with elevated bone marrow megakaryocyte number and macrothrombocytopenia, female Mpig6b-/- mice developed progressive splenomegaly starting at 8 weeks of age. Micro-computed tomography (µCT) of femurs showed that female Mpig6b-/- mice had increased cortical thickness and reduced bone marrow area starting at 8 weeks of age and developed occlusion of the medullary cavity by trabeculae by 16 weeks of age. In contrast, male Mpig6b-/- mice developed only a small number of trabeculae in the medullary cavity at the proximal diaphysis and demonstrated a temporary decrease in bone volume fraction and trabecular thickness at 16 weeks. Ovariectomy of 10-week-old female Mpig6b-/- mice prevented the development of medullary cavity osteosclerosis, whereas orchiectomy of male Mpig6b-/- mice did not exacerbate their disease. Importantly, ovariectomized female Mpig6b-/- mice also demonstrated improvement in spleen weight compared to sham-operated Mpig6b-/- mice, establishing estrogen as a contributing factor to the severity of the megakaryocyte-driven osteosclerosis. © 2021 American Society for Bone and Mineral Research (ASBMR).


Subject(s)
Osteosclerosis , Primary Myelofibrosis , Animals , Bone and Bones , Female , Humans , Male , Megakaryocytes , Mice , Osteosclerosis/diagnostic imaging , Osteosclerosis/genetics , Ovariectomy , Primary Myelofibrosis/diagnostic imaging , Primary Myelofibrosis/genetics , X-Ray Microtomography
SELECTION OF CITATIONS
SEARCH DETAIL