Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
Blood ; 140(14): 1592-1606, 2022 10 06.
Article in English | MEDLINE | ID: mdl-35767701

ABSTRACT

Adult hematopoietic stem cells (HSCs) are predominantly quiescent and can be activated in response to acute stress such as infection or cytotoxic insults. STAT1 is a pivotal downstream mediator of interferon (IFN) signaling and is required for IFN-induced HSC proliferation, but little is known about the role of STAT1 in regulating homeostatic hematopoietic stem/progenitor cells (HSPCs). Here, we show that loss of STAT1 altered the steady state HSPC landscape, impaired HSC function in transplantation assays, delayed blood cell regeneration following myeloablation, and disrupted molecular programs that protect HSCs, including control of quiescence. Our results also reveal STAT1-dependent functional HSC heterogeneity. A previously unrecognized subset of homeostatic HSCs with elevated major histocompatibility complex class II (MHCII) expression (MHCIIhi) displayed molecular features of reduced cycling and apoptosis and was refractory to 5-fluorouracil-induced myeloablation. Conversely, MHCIIlo HSCs displayed increased megakaryocytic potential and were preferentially expanded in CALR mutant mice with thrombocytosis. Similar to mice, high MHCII expression is a feature of human HSCs residing in a deeper quiescent state. Our results therefore position STAT1 at the interface of stem cell heterogeneity and the interplay between stem cells and the adaptive immune system, areas of broad interest in the wider stem cell field.


Subject(s)
Hematopoietic Stem Cells , Megakaryocytes , STAT1 Transcription Factor , Animals , Cell Proliferation , Fluorouracil/pharmacology , Hematopoietic Stem Cells/metabolism , Humans , Interferons , Megakaryocytes/metabolism , Mice , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism
2.
Blood ; 139(14): 2227-2239, 2022 04 07.
Article in English | MEDLINE | ID: mdl-35051265

ABSTRACT

The process of platelet production has so far been understood to be a 2-stage process: megakaryocyte maturation from hematopoietic stem cells followed by proplatelet formation, with each phase regulating the peripheral blood platelet count. Proplatelet formation releases into the bloodstream beads-on-a-string preplatelets, which undergo fission into mature platelets. For the first time, we show that preplatelet maturation is a third, tightly regulated, critical process akin to cytokinesis that regulates platelet count. We show that deficiency in cytokine receptor-like factor 3 (CRLF3) in mice leads to an isolated and sustained 25% to 48% reduction in the platelet count without any effect on other blood cell lineages. We show that Crlf3-/- preplatelets have increased microtubule stability, possibly because of increased microtubule glutamylation via the interaction of CRLF3 with key members of the Hippo pathway. Using a mouse model of JAK2 V617F essential thrombocythemia, we show that a lack of CRLF3 leads to long-term lineage-specific normalization of the platelet count. We thereby postulate that targeting CRLF3 has therapeutic potential for treatment of thrombocythemia.


Subject(s)
Blood Platelets , Thrombocythemia, Essential , Blood Platelets/metabolism , Humans , Megakaryocytes/metabolism , Microtubules , Platelet Count , Receptors, Cytokine , Thrombocythemia, Essential/drug therapy , Thrombopoiesis/genetics
3.
Blood ; 131(6): 649-661, 2018 02 08.
Article in English | MEDLINE | ID: mdl-29282219

ABSTRACT

Somatic mutations in the endoplasmic reticulum chaperone calreticulin (CALR) are detected in approximately 40% of patients with essential thrombocythemia (ET) and primary myelofibrosis (PMF). Multiple different mutations have been reported, but all result in a +1-bp frameshift and generate a novel protein C terminus. In this study, we generated a conditional mouse knockin model of the most common CALR mutation, a 52-bp deletion. The mutant novel human C-terminal sequence is integrated into the otherwise intact mouse CALR gene and results in mutant CALR expression under the control of the endogenous mouse locus. CALRdel/+ mice develop a transplantable ET-like disease with marked thrombocytosis, which is associated with increased and morphologically abnormal megakaryocytes and increased numbers of phenotypically defined hematopoietic stem cells (HSCs). Homozygous CALRdel/del mice developed extreme thrombocytosis accompanied by features of MF, including leukocytosis, reduced hematocrit, splenomegaly, and increased bone marrow reticulin. CALRdel/+ HSCs were more proliferative in vitro, but neither CALRdel/+ nor CALRdel/del displayed a competitive transplantation advantage in primary or secondary recipient mice. These results demonstrate the consequences of heterozygous and homozygous CALR mutations and provide a powerful model for dissecting the pathogenesis of CALR-mutant ET and PMF.


Subject(s)
Calreticulin/genetics , Cell Self Renewal/genetics , Hematopoietic Stem Cells/physiology , Primary Myelofibrosis/genetics , Thrombocytosis/genetics , Animals , Cells, Cultured , Homozygote , Leukocytosis/genetics , Leukocytosis/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation, Missense , Splenomegaly/genetics , Splenomegaly/pathology , Thrombocythemia, Essential/genetics , Thrombocythemia, Essential/pathology
4.
Haematologica ; 104(5): 1036-1045, 2019 05.
Article in English | MEDLINE | ID: mdl-30467204

ABSTRACT

Sphingolipids are fundamental to membrane trafficking, apoptosis, and cell differentiation and proliferation. KDSR or 3-keto-dihydrosphingosine reductase is an essential enzyme for de novo sphingolipid synthesis, and pathogenic mutations in KDSR result in the severe skin disorder erythrokeratodermia variabilis et progressiva-4 Four of the eight reported cases also had thrombocytopenia but the underlying mechanism has remained unexplored. Here we expand upon the phenotypic spectrum of KDSR deficiency with studies in two siblings with novel compound heterozygous variants associated with thrombocytopenia, anemia, and minimal skin involvement. We report a novel phenotype of progressive juvenile myelofibrosis in the propositus, with spontaneous recovery of anemia and thrombocytopenia in the first decade of life. Examination of bone marrow biopsies showed megakaryocyte hyperproliferation and dysplasia. Megakaryocytes obtained by culture of CD34+ stem cells confirmed hyperproliferation and showed reduced proplatelet formation. The effect of KDSR insufficiency on the sphingolipid profile was unknown, and was explored in vivo and in vitro by a broad metabolomics screen that indicated activation of an in vivo compensatory pathway that leads to normalization of downstream metabolites such as ceramide. Differentiation of propositus-derived induced pluripotent stem cells to megakaryocytes followed by expression of functional KDSR showed correction of the aberrant cellular and biochemical phenotypes, corroborating the critical role of KDSR in proplatelet formation. Finally, Kdsr depletion in zebrafish recapitulated the thrombocytopenia and showed biochemical changes similar to those observed in the affected siblings. These studies support an important role for sphingolipids as regulators of cytoskeletal organization during megakaryopoiesis and proplatelet formation.


Subject(s)
Alcohol Oxidoreductases/deficiency , Blood Platelets/pathology , Induced Pluripotent Stem Cells/pathology , Megakaryocytes/pathology , Sphingolipids/metabolism , Thrombocytopenia/etiology , Alcohol Oxidoreductases/genetics , Animals , Blood Platelets/metabolism , Cell Differentiation , Cells, Cultured , Child , Female , Humans , Induced Pluripotent Stem Cells/metabolism , Male , Megakaryocytes/metabolism , Metabolomics , Mutation , Pedigree , Prognosis , Thrombocytopenia/metabolism , Thrombocytopenia/pathology , Zebrafish
5.
Transfusion ; 59(1): 32-38, 2019 01.
Article in English | MEDLINE | ID: mdl-30394544

ABSTRACT

BACKGROUND: Beyond their role in hemostasis and thrombosis, platelets are also important mediators of inflammation by the release of hundreds of factors stored in their α-granules. Mutations in Nbeal2 cause gray platelet syndrome (GPS), characterized by the lack of platelet α-granules. This study aims to evaluate the immunological (proinflammatory) effects of platelet α-granules. STUDY DESIGN AND METHODS: We performed an experiment using Nbeal2-/- mice, the mouse model of GPS. Systemic inflammation was induced by intravenous injection of lipopolysaccharide (LPS). Inflammatory response was assessed by quantification of inflammatory soluble factors and platelet biological response modifiers. RESULTS: The lack of Nbeal2 (in Nbeal2 -/- mice, compared with controls) significantly reduced the recruitment of circulating neutrophils and monocytes. Moreover, after LPS injection, there was a significant increase in neutrophil and monocyte counts in control animals, compared with Nbeal2 -/- mice. The control of inflammation, evaluated by the production of anti-inflammatory cytokines, appeared to be greater in Nbeal2-/- mice compared with controls. Conversely, the production of certain inflammatory-soluble mediators known to characterize normal platelet secretion, such as soluble CD40 ligand (sCD40L), was decreased under experimental inflammation in Nbeal2 -/- mice. CONCLUSIONS: These results show that α-granules play a direct role in platelet-mediated inflammation balance, confirming the need to further investigate platelet-associated inflammatory pathophysiology and inflammatory adverse events related to blood transfusion.


Subject(s)
Blood Proteins/metabolism , Gray Platelet Syndrome/immunology , Lipopolysaccharides/toxicity , Animals , Blood Proteins/genetics , CD40 Ligand/genetics , CD40 Ligand/metabolism , Disease Models, Animal , Gray Platelet Syndrome/genetics , Gray Platelet Syndrome/metabolism , Immunoassay , Inflammation/chemically induced , Inflammation/genetics , Inflammation/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation/genetics
6.
Pediatr Res ; 85(6): 874-884, 2019 05.
Article in English | MEDLINE | ID: mdl-30742030

ABSTRACT

BACKGROUND: Neonatal haemorrhaging is often co-observed with thrombocytopenia; however, no evidence of a causal relationship with low platelet count has been reported. Regardless, the administration of a platelet transfusion is often based upon this parameter. Accurate measurement of platelet function in small volumes of adult blood samples by flow cytometry is well established and we propose that the use of the same technology could provide complementary information to guide the administration of platelet transfusions in premature neonates. METHODS: In 28 neonates born at 27-41 weeks gestation, platelet function after stimulation agonists was measured using fibrinogen binding and P-selectin expression (a marker of degranulation). RESULTS: Platelets of neonates with gestation of ≤36 weeks (n = 20) showed reduced fibrinogen binding and degranulation with ADP, and reduced degranulation with CRP-XL. Degranulation Scores of 7837 ± 5548, 22,408 ± 5301 and 53,131 ± 12,102 (mean ± SEM) identified significant differences between three groups: <29, 29-36 and >36 weeks gestation). Fibrinogen binding and degranulation responses to ADP were significantly reduced in suspected septic neonates (n = 6) and the Fibrinogen Binding scores clearly separated the septic and healthy group (88.2 ± 10.3 vs 38.6 ± 12.2, P = 0.03). CONCLUSIONS: Flow cytometric measurement of platelet function identified clinically different neonatal groups and may eventually contribute to assessment of neonates requiring platelet transfusion.


Subject(s)
Flow Cytometry/methods , Infant, Premature/blood , Platelet Function Tests/methods , Platelet Transfusion , Cell Degranulation , Female , Fibrinogen/metabolism , Hemorrhage/blood , Hemorrhage/therapy , Humans , Infant, Newborn , Male , Neonatal Sepsis/blood , P-Selectin/blood , Platelet Activation , Platelet Count , Platelet Function Tests/standards , Thrombocytopenia, Neonatal Alloimmune/blood , Thrombocytopenia, Neonatal Alloimmune/therapy
8.
Blood ; 127(9): e1-e11, 2016 Mar 03.
Article in English | MEDLINE | ID: mdl-26660425

ABSTRACT

In platelets, splicing and translation occur in the absence of a nucleus. However, the integrity and stability of mRNAs derived from megakaryocyte progenitor cells remain poorly quantified on a transcriptome-wide level. As circular RNAs (circRNAs) are resistant to degradation by exonucleases, their abundance relative to linear RNAs can be used as a surrogate marker for mRNA stability in the absence of transcription. Here we show that circRNAs are enriched in human platelets 17- to 188-fold relative to nucleated tissues and 14- to 26-fold relative to samples digested with RNAse R to selectively remove linear RNA. We compare RNAseq read depths inside and outside circRNAs to provide in silico evidence of transcript circularity, show that exons within circRNAs are enriched on average 12.7 times in platelets relative to nucleated tissues and identify 3162 genes significantly enriched for circRNAs, including some where all RNAseq reads appear to be derived from circular molecules. We also confirm that this is a feature of other anucleate cells through transcriptome sequencing of mature erythrocytes, demonstrate that circRNAs are not enriched in cultured megakaryocytes, and demonstrate that linear RNAs decay more rapidly than circRNAs in platelet preparations. Collectively, these results suggest that circulating platelets have lost >90% of their progenitor mRNAs and that translation in platelets occurs against the backdrop of a highly degraded transcriptome. Finally, we find that transcripts previously classified as products of reverse transcriptase template switching are both enriched in platelets and resistant to decay, countering the recent suggestion that up to 50% of rearranged RNAs are artifacts.


Subject(s)
Blood Platelets/metabolism , RNA Stability/genetics , RNA/genetics , Transcriptome/genetics , Exons/genetics , Exoribonucleases/metabolism , Humans , Megakaryocytes/metabolism , RNA, Circular , Real-Time Polymerase Chain Reaction , Reproducibility of Results
9.
Blood ; 127(23): 2791-803, 2016 06 09.
Article in English | MEDLINE | ID: mdl-27084890

ABSTRACT

Inherited bleeding, thrombotic, and platelet disorders (BPDs) are diseases that affect ∼300 individuals per million births. With the exception of hemophilia and von Willebrand disease patients, a molecular analysis for patients with a BPD is often unavailable. Many specialized tests are usually required to reach a putative diagnosis and they are typically performed in a step-wise manner to control costs. This approach causes delays and a conclusive molecular diagnosis is often never reached, which can compromise treatment and impede rapid identification of affected relatives. To address this unmet diagnostic need, we designed a high-throughput sequencing platform targeting 63 genes relevant for BPDs. The platform can call single nucleotide variants, short insertions/deletions, and large copy number variants (though not inversions) which are subjected to automated filtering for diagnostic prioritization, resulting in an average of 5.34 candidate variants per individual. We sequenced 159 and 137 samples, respectively, from cases with and without previously known causal variants. Among the latter group, 61 cases had clinical and laboratory phenotypes indicative of a particular molecular etiology, whereas the remainder had an a priori highly uncertain etiology. All previously detected variants were recapitulated and, when the etiology was suspected but unknown or uncertain, a molecular diagnosis was reached in 56 of 61 and only 8 of 76 cases, respectively. The latter category highlights the need for further research into novel causes of BPDs. The ThromboGenomics platform thus provides an affordable DNA-based test to diagnose patients suspected of having a known inherited BPD.


Subject(s)
Blood Platelet Disorders/genetics , Genetic Predisposition to Disease , Hemorrhage/genetics , High-Throughput Nucleotide Sequencing/methods , Thrombosis/genetics , Case-Control Studies , DNA Copy Number Variations , Female , Genetic Association Studies/methods , Humans , Male , Mutation , Polymorphism, Single Nucleotide , Sequence Analysis, DNA/methods
10.
J Biol Chem ; 291(19): 9872-81, 2016 May 06.
Article in English | MEDLINE | ID: mdl-27006395

ABSTRACT

The biosynthesis of endogenous brain-derived neurotrophic factor (BDNF) has thus far been examined in neurons where it is expressed at very low levels, in an activity-dependent fashion. In humans, BDNF has long been known to accumulate in circulating platelets, at levels far higher than in the brain. During the process of blood coagulation, BDNF is released from platelets, which has led to its extensive use as a readily accessible biomarker, under the assumption that serum levels may somehow reflect brain levels. To identify the cellular origin of BDNF in platelets, we established primary cultures of megakaryocytes, the progenitors of platelets, and we found that human and rat megakaryocytes express the BDNF gene. Surprisingly, the pattern of mRNA transcripts is similar to neurons. In the presence of thapsigargin and external calcium, the levels of the mRNA species leading to efficient BDNF translation rapidly increase. Under these conditions, pro-BDNF, the obligatory precursor of biologically active BDNF, becomes readily detectable. Megakaryocytes store BDNF in α-granules, with more than 80% of them also containing platelet factor 4. By contrast, BDNF is undetectable in mouse megakaryocytes, in line with the absence of BDNF in mouse serum. These findings suggest that alterations of BDNF levels in human serum as reported in studies dealing with depression or physical exercise may primarily reflect changes occurring in megakaryocytes and platelets, including the ability of the latter to retain and release BDNF.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Megakaryocytes/metabolism , Secretory Vesicles/metabolism , Animals , Blood Coagulation/physiology , Blood Platelets/cytology , Blood Platelets/metabolism , COS Cells , Calcium/pharmacology , Chlorocebus aethiops , Humans , Megakaryocytes/cytology , Mice , Neurons/metabolism , RNA, Messenger/metabolism , Rats , Rats, Wistar , Species Specificity , Thapsigargin/pharmacology
11.
Blood ; 126(10): 1234-6, 2015 Sep 03.
Article in English | MEDLINE | ID: mdl-26209661

ABSTRACT

Incompatibility of the human platelet antigen-1 (HPA-1) system is the most common cause of fetal/neonatal alloimmune thrombocytopenia (F/NAIT) and is thought to be mediated by accelerated clearance of antibody-opsonized fetal platelets. We evaluated the effect of maternal sera containing anti-HPA-1a antibodies (F/NAIT sera) on in vitro megakaryopoiesis. Compared with control maternal sera, 14 out of 17 F/NAIT sera significantly reduced megakaryocyte (MK) number. This finding was associated with increased apoptosis and cell death of early MKs/MK progenitors, but normal maturation and differentiation of surviving MKs. An analysis of platelet counts in infants born to mothers following antenatal intravenous immunoglobulin (IVIG) ± prednisone therapy demonstrated a significant and moderately strong correlation between the MK growth in cultures and the infants' platelet counts at birth. These findings suggest that maternal anti-HPA-1a antibodies can suppress fetal megakaryopoiesis by inducing early cell death and that this influences the neonatal platelet count. Thus, the ability of maternal antibodies to suppress MK growth is a potential predictive factor for the fetal response to maternal IVIG therapy.


Subject(s)
Antigens, Human Platelet/immunology , Hematopoiesis/immunology , Megakaryocytes/immunology , Thrombocytopenia, Neonatal Alloimmune/immunology , Female , Humans , In Vitro Techniques , Integrin beta3 , Pregnancy , Thrombocytopenia, Neonatal Alloimmune/physiopathology
12.
J Immunol ; 194(12): 5751-60, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25972474

ABSTRACT

Human platelet Ag (HPA)-1a, located on integrin ß3, is the main target for alloantibodies responsible for fetal and neonatal alloimmune thrombocytopenia (FNAIT) in the white population. There are ongoing efforts to develop an Ab prophylaxis and therapy to prevent or treat FNAIT. In this study, an mAb specific for HPA-1a, named 26.4, was derived from an immortalized B cell from an alloimmunized woman who had an infant affected by FNAIT. It is the only HPA-1a-specific human mAb with naturally paired H and L chains. Specific binding of mAb 26.4, both native and recombinant forms, to platelets and to purified integrins αIIbß3 (from platelets) and αVß3 (from trophoblasts) from HPA-1a(+) donors was demonstrated by flow cytometry and surface plasmon resonance technology, respectively. No binding to HPA-1a(-) platelets or integrins was detected. Moreover, the Ab binds with higher affinity to integrin αVß3 compared with a second HPA-1a-specific human mAb, B2G1. Further in vitro experimentation demonstrated that mAb 26.4 can opsonize HPA-1a(+) platelets for enhanced phagocytosis by monocytes, inhibit binding of maternal polyclonal anti-HPA-1a Abs, and weakly inhibit aggregation of HPA-1a-heterozygous platelets, the latter with no predicted clinical relevance. Thus, mAb 26.4 is highly specific for HPA-1a and could potentially be explored for use as a prophylactic or therapeutic reagent for FNAIT intervention and as a phenotyping reagent to identify women at risk for immunization.


Subject(s)
Antibodies, Monoclonal/immunology , Antibody Formation/immunology , Antigens, Human Platelet/immunology , B-Lymphocytes/immunology , Isoantibodies/immunology , Amino Acid Sequence , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/pharmacology , Antibody Affinity/immunology , Antibody Specificity , B-Lymphocytes/metabolism , Base Sequence , Blood Platelets/drug effects , Blood Platelets/immunology , Blood Platelets/metabolism , Cell Line, Transformed , Female , Humans , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Immunoglobulin Variable Region/chemistry , Immunoglobulin Variable Region/genetics , Immunologic Memory , Integrin alphaVbeta3/metabolism , Integrin beta3 , Molecular Sequence Data , Mutation , Phagocytosis/drug effects , Phagocytosis/immunology , Platelet Aggregation/drug effects , Platelet Aggregation/immunology , Pregnancy , Protein Binding/immunology , Recombinant Proteins/genetics , Recombinant Proteins/immunology
13.
Blood ; 124(24): 3624-35, 2014 Dec 04.
Article in English | MEDLINE | ID: mdl-25258341

ABSTRACT

NBEAL2 encodes a multidomain scaffolding protein with a putative role in granule ontogeny in human platelets. Mutations in NBEAL2 underlie gray platelet syndrome (GPS), a rare inherited bleeding disorder characterized by a lack of α-granules within blood platelets and progressive bone marrow fibrosis. We present here a novel Nbeal2(-/-) murine model of GPS and demonstrate that the lack of α-granules is due to their loss from platelets/mature megakaryocytes (MKs), and not by initial impaired formation. We show that the lack of Nbeal2 confers a proinflammatory phenotype to the bone marrow MKs, which in combination with the loss of proteins from α-granules drives the development of bone marrow fibrosis. In addition, we demonstrate that α-granule deficiency impairs platelet function beyond their purely hemostatic role and that Nbeal2 deficiency has a protective effect against cancer metastasis.


Subject(s)
Gray Platelet Syndrome/metabolism , Megakaryocytes/metabolism , Animals , Blood Proteins/genetics , Blood Proteins/metabolism , Disease Models, Animal , Gray Platelet Syndrome/genetics , Gray Platelet Syndrome/pathology , Humans , Megakaryocytes/pathology , Mice , Mice, Knockout , Mutation , Neoplasm Metastasis , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Primary Myelofibrosis/genetics , Primary Myelofibrosis/metabolism , Primary Myelofibrosis/pathology , Secretory Vesicles
14.
Blood ; 123(20): 3139-51, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24692758

ABSTRACT

Genomic regions of acquired uniparental disomy (UPD) are common in malignancy and frequently harbor mutated oncogenes. Homozygosity for such gain-of-function mutations is thought to modulate tumor phenotype, but direct evidence has been elusive. Polycythemia vera (PV) and essential thrombocythemia (ET), 2 subtypes of myeloproliferative neoplasms, are associated with an identical acquired JAK2V617F mutation but the mechanisms responsible for distinct clinical phenotypes remain unclear. We provide direct genetic evidence and demonstrate that homozygosity for human JAK2V617F in knock-in mice results in a striking phenotypic switch from an ET-like to PV-like phenotype. The resultant erythrocytosis is driven by increased numbers of early erythroid progenitors and enhanced erythroblast proliferation, whereas reduced platelet numbers are associated with impaired platelet survival. JAK2V617F-homozygous mice developed a severe hematopoietic stem cell defect, suggesting that additional lesions are needed to sustain clonal expansion. Together, our results indicate that UPD for 9p plays a causal role in the PV phenotype in patients as a consequence of JAK2V617F homozygosity. The generation of a JAK2V617F allelic series of mice with a dose-dependent effect on hematopoiesis provides a powerful model for studying the consequences of mutant JAK2 homozygosity.


Subject(s)
Janus Kinase 2/genetics , Mutation , Polycythemia Vera/genetics , Thrombocythemia, Essential/genetics , Animals , Blood Platelets/metabolism , Blood Platelets/pathology , Erythroblasts/metabolism , Erythroblasts/pathology , Female , Gene Knock-In Techniques , Homozygote , Humans , Male , Megakaryocytes/metabolism , Megakaryocytes/pathology , Mice , Mice, Inbred C57BL , Phenotype , Polycythemia Vera/pathology , Thrombocythemia, Essential/pathology , Uniparental Disomy/genetics , Uniparental Disomy/pathology
15.
Arterioscler Thromb Vasc Biol ; 35(12): 2554-61, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26471268

ABSTRACT

OBJECTIVE: Platelets are increasingly implicated in processes beyond hemostasis and thrombosis, such as vascular remodeling. Members of the matrix metalloproteinase (MMP) family not only remodel the extracellular matrix but also modulate platelet function. Here, we made a systematic comparison of the roles of MMP family members in acute thrombus formation under flow conditions and assessed platelet-dependent collagenolytic activity over time. APPROACH AND RESULTS: Pharmacological inhibition of MMP-1 or MMP-2 (human) or deficiency in MMP-2 (mouse) suppressed collagen-dependent platelet activation and thrombus formation under flow, whereas MMP-9 inhibition/deficiency stimulated these processes. The absence of MMP-3 was without effect. Interestingly, MMP-14 inhibition led to the formation of larger thrombi, which occurred independently of its capacity to activate MMP-2. Platelet thrombi exerted local collagenolytic activity capable of cleaving immobilized dye-quenched collagen and fibrillar collagen fibers within hours, with loss of the majority of the platelet adhesive properties of collagen as a consequence. This collagenolytic activity was redundantly mediated by platelet-associated MMP-1, MMP-2, MMP-9, and MMP-14 but occurred independently of platelet α-granule release (Nbeal2(-/-) mice). The latter was in line with subcellular localization experiments, which indicated a granular distribution of MMP-1 and MMP-2 in platelets, distinct from α-granules. Whereas MMP-9 protein could not be detected inside platelets, activated platelets did bind plasma-derived MMP-9 to their plasma membrane. Overall, platelet MMP activity was predominantly membrane-associated and influenced by platelet activation status. CONCLUSIONS: Platelet-associated MMP-1, MMP-2, MMP-9, and MMP-14 differentially modulate acute thrombus formation and at later time points limit thrombus formation by exerting collagenolytic activity.


Subject(s)
Blood Platelets/enzymology , Collagen/metabolism , Collagenases/blood , Thrombosis/enzymology , Animals , Blood Platelets/drug effects , Blood Proteins/genetics , Blood Proteins/metabolism , Collagenases/deficiency , Collagenases/genetics , Disease Models, Animal , Humans , Matrix Metalloproteinase Inhibitors/pharmacology , Mice, Inbred C57BL , Mice, Knockout , Platelet Activation , Platelet Membrane Glycoproteins/metabolism , Proteolysis , Thrombosis/blood , Thrombosis/genetics , Time Factors
16.
Eur J Immunol ; 44(3): 905-14, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24285214

ABSTRACT

G1Δnab is a mutant human IgG1 constant region with a lower ability to interact with FcγR than the natural IgG constant regions. Radiolabelled RBCs and platelets sensitised with specific G1Δnab Abs were cleared more slowly from human circulation than IgG1-sensitised counterparts. However, non-destructive splenic retention of G1Δnab-coated RBCs required investigation and plasma radioactivities now suggest this also occurred for platelets sensitised with an IgG1/G1Δnab mixture. In vitro assays with human cells showed that G1Δnab-sensitised RBCs did not cause FcγRI-mediated monocyte activation, FcγRIIIa-mediated antibody-dependent cell-mediated cytotoxicity (ADCC) or macrophage phagocytosis although they did adhere to macrophages. Thus, FcγRII was implicated in the adhesion despite the Δnab mutation reducing the already low-affinity binding to this receptor class. Additional contacts via P-selectin enhance the interaction of sensitised platelets with monocytes and this system provided evidence of FcγRII-dependent activation by G1Δnab. These results emphasise the physiological relevance of low-affinity interactions: It appears that FcγRII interactions of G1Δnab allowed splenic retention of G1Δnab-coated RBCs with inhibitory FcγRIIb binding preventing RBC destruction and that FcγRIIb engagement by G1Δnab on IgG1/G1Δnab-sensitised platelets overcame activation by IgG1. Considering therapeutic blocking Abs, G1Δnab offers lower FcγR binding and a greater bias towards inhibition than IgG2 and IgG4 constant regions.


Subject(s)
Blood Platelets/immunology , Blood Platelets/metabolism , Erythrocytes/immunology , Erythrocytes/metabolism , Immunoglobulin G/immunology , Receptors, IgG/metabolism , Antigens, Human Platelet/immunology , Cell Survival/immunology , Cell Survival/radiation effects , Humans , Immunoglobulin G/metabolism , Integrin beta3 , Monocytes/immunology , Nuclear Proteins/immunology , Protein Binding , Rh-Hr Blood-Group System/immunology , Transcription Factors/immunology
17.
Blood ; 122(3): 313-20, 2013 Jul 18.
Article in English | MEDLINE | ID: mdl-23656729

ABSTRACT

Fetomaternal alloimmune thrombocytopenia, caused by the maternal generation of antibodies against fetal human platelet antigen-1a (HPA-1a), can result in intracranial hemorrhage and intrauterine death. We have developed a therapeutic human recombinant high-affinity HPA-1a antibody (B2G1Δnab) that competes for binding to the HPA-1a epitope but carries a modified constant region that does not bind to Fcγ receptors. In vitro studies with a range of clinical anti-HPA-1a sera have shown that B2G1Δnab blocks monocyte chemiluminescence by >75%. In this first-in-man study, we demonstrate that HPA-1a1b autologous platelets (matching fetal phenotype) sensitized with B2G1Δnab have the same intravascular survival as unsensitized platelets (190 hours), while platelets sensitized with a destructive immunoglobulin G1 version of the antibody (B2G1) are cleared from the circulation in 2 hours. Mimicking the situation in fetuses receiving B2G1Δnab as therapy, we show that platelets sensitized with a combination of B2G1 (representing destructive HPA-1a antibody) and B2G1Δnab survive 3 times as long in circulation compared with platelets sensitized with B2G1 alone. This confirms the therapeutic potential of B2G1Δnab. The efficient clearance of platelets sensitized with B2G1 also opens up the opportunity to carry out studies of prophylaxis to prevent alloimmunization in HPA-1a-negative mothers.


Subject(s)
Antibodies/therapeutic use , Recombinant Proteins/therapeutic use , Thrombocytopenia, Neonatal Alloimmune/drug therapy , Antigens, Human Platelet/immunology , Blood Platelets/immunology , Blood Vessels/pathology , Cell Survival/immunology , Female , Flow Cytometry , Humans , Immunoglobulin G/blood , Integrin beta3 , Male , Mutant Proteins/immunology , Software , Thrombocytopenia, Neonatal Alloimmune/blood , Thrombocytopenia, Neonatal Alloimmune/immunology
18.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL