Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Cell ; 158(5): 1033-1044, 2014 Aug 28.
Article in English | MEDLINE | ID: mdl-25171405

ABSTRACT

Although tyrosine phosphorylation of extracellular proteins has been reported to occur extensively in vivo, no secreted protein tyrosine kinase has been identified. As a result, investigation of the potential role of extracellular tyrosine phosphorylation in physiological and pathological tissue regulation has not been possible. Here, we show that VLK, a putative protein kinase previously shown to be essential in embryonic development, is a secreted protein kinase, with preference for tyrosine, that phosphorylates a broad range of secreted and ER-resident substrate proteins. We find that VLK is rapidly and quantitatively secreted from platelets in response to stimuli and can tyrosine phosphorylate coreleased proteins utilizing endogenous as well as exogenous ATP sources. We propose that discovery of VLK activity provides an explanation for the extensive and conserved pattern of extracellular tyrosine phosphophorylation seen in vivo, and extends the importance of regulated tyrosine phosphorylation into the extracellular environment.


Subject(s)
Blood Platelets/enzymology , Embryo, Mammalian/enzymology , Protein Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Amino Acid Sequence , Animals , Embryonic Development , Glycosylation , Humans , Mice , Molecular Sequence Data , Phosphorylation , Protein Kinases/chemistry , Protein Kinases/genetics , Protein Processing, Post-Translational , Protein Structure, Tertiary , Protein-Tyrosine Kinases/chemistry , Secretory Pathway
2.
Haematologica ; 107(2): 519-531, 2022 02 01.
Article in English | MEDLINE | ID: mdl-33567808

ABSTRACT

Antisense oligonucleotides (ASO) are DNA-based, disease-modifying drugs. Clinical trials with 2'-O-methoxyethyl (2'MOE) ASO have shown dose- and sequence-specific lowering of platelet counts according to two phenotypes. Phenotype 1 is a moderate (but not clinically severe) drop in platelet count. Phenotype 2 is rare, severe thrombocytopenia. This article focuses on the underlying cause of the more common phenotype 1, investigating the effects of ASO on platelet production and platelet function. Five phosphorothioate ASO were studied: three 2'MOE sequences; 487660 (no effects on platelet count), 104838 (associated with phenotype 1), and 501861 (effects unknown) and two CpG sequences; 120704 and ODN 2395 (known to activate platelets). Human cord bloodderived megakaryocytes were treated with these ASO to study their effects on proplatelet production. Platelet activation (determined by surface Pselectin) and platelet-leukocyte aggregates were analyzed in ASO-treated blood from healthy human volunteers. None of the ASO inhibited proplatelet production by human megakaryocytes. All the ASO were shown to bind to the platelet receptor glycoprotein VI (KD ~0.2-1.5 mM). CpG ASO had the highest affinity to glycoprotein VI, the most potent platelet-activating effects and led to the greatest formation of platelet-leukocyte aggregates. 2'MOE ASO 487660 had no detectable platelet effects, while 2'MOE ASOs 104838 and 501861 triggered moderate platelet activation and SYKdependent formation of platelet-leukocyte aggregates. Donors with higher platelet glycoprotein VI levels had greater ASO-induced platelet activation. Sequence-dependent ASO-induced platelet activation and platelet-leukocyte aggregates may explain phenotype 1 (moderate drops in platelet count). Platelet glycoprotein VI levels could be useful as a screening tool to identify patients at higher risk of ASO-induced platelet side effects.


Subject(s)
Blood Platelets , Oligonucleotides, Antisense , Humans , Leukocytes , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/pharmacology , Oligonucleotides, Antisense/therapeutic use , Platelet Activation , Platelet Count
3.
Trends Biochem Sci ; 42(5): 327-329, 2017 05.
Article in English | MEDLINE | ID: mdl-28385393

ABSTRACT

Translating basic research discoveries through entrepreneurship must be scientist driven and institutionally supported to be successful (not the other way around). Here, we describe why scientists should engage in entrepreneurship, where institutional support for scientist-founders falls short, and how these challenges can be overcome.


Subject(s)
Entrepreneurship , Research Personnel
4.
Trends Biochem Sci ; 39(12): 571-3, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25458606

ABSTRACT

An entrepreneurial movement within science strives to invert the classical trajectory of academic research careers by positioning trainees at the apex of burgeoning industries. Young scientists today have nothing to lose and everything to gain by pursuing this 'third road', and academic institutes and established companies only stand to benefit from supporting this emerging movement of discovery research with economic purpose.


Subject(s)
Biochemistry , Career Choice , Education, Graduate , Entrepreneurship , Biochemistry/economics , Biochemistry/education , Canada , Education, Graduate/economics , Education, Graduate/trends , Humans , Salaries and Fringe Benefits , United States , Workforce
5.
Blood ; 125(5): 860-8, 2015 Jan 29.
Article in English | MEDLINE | ID: mdl-25411426

ABSTRACT

Bone marrow megakaryocytes produce platelets by extending long cytoplasmic protrusions, designated proplatelets, into sinusoidal blood vessels. Although microtubules are known to regulate platelet production, the underlying mechanism of proplatelet elongation has yet to be resolved. Here we report that proplatelet formation is a process that can be divided into repetitive phases (extension, pause, and retraction), as revealed by differential interference contrast and fluorescence loss after photoconversion time-lapse microscopy. Furthermore, we show that microtubule sliding drives proplatelet elongation and is dependent on cytoplasmic dynein under static and physiological shear stress by using fluorescence recovery after photobleaching in proplatelets with fluorescence-tagged ß1-tubulin. A refined understanding of the specific mechanisms regulating platelet production will yield strategies to treat patients with thrombocythemia or thrombocytopenia.


Subject(s)
Blood Platelets/metabolism , Cytoplasmic Dyneins/metabolism , Megakaryocytes/metabolism , Microtubules/metabolism , Tubulin/metabolism , Animals , Blood Platelets/cytology , Cell Differentiation , Cytoplasm/metabolism , Cytoplasmic Dyneins/genetics , Fluorescence Recovery After Photobleaching , Gene Expression , Mechanotransduction, Cellular , Megakaryocytes/cytology , Mice , Microscopy, Interference , Microtubules/chemistry , Primary Cell Culture , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Stress, Mechanical , Thrombopoiesis/genetics , Tubulin/genetics
6.
Platelets ; 28(5): 472-477, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28112988

ABSTRACT

Platelets, responsible for clot formation and blood vessel repair, are produced by megakaryocytes in the bone marrow. Platelets are critical for hemostasis and wound healing, and are often provided following surgery, chemotherapy, and major trauma. Despite their importance, platelets today are derived exclusively from human volunteer donors. They have a shelf life of just five days, making platelet shortages common during long weekends, civic holidays, bad weather, and during major emergencies when platelets are needed most. Megakaryocytes in the bone marrow generate platelets by extruding long cytoplasmic extensions called proplatelets through gaps/fenestrations in blood vessels. Proplatelets serve as assembly lines for platelet production by sequentially releasing platelets and large discoid-shaped platelet intermediates called preplatelets into the circulation. Recent advances in platelet bioreactor development have aimed to mimic the key physiological characteristics of bone marrow, including extracellular matrix composition/stiffness, blood vessel architecture comprising tissue-specific microvascular endothelium, and shear stress. Nevertheless, how complex interactions within three-dimensional (3D) microenvironments regulate thrombopoiesis remains poorly understood, and the technical challenges associated with designing and manufacturing biomimetic microfluidic devices are often under-appreciated and under-reported. We have previously reviewed the major cell culture, platelet quality assessment, and regulatory roadblocks that must be overcome to make human platelet production possible for clinical use [1]. This review builds on our previous manuscript by: (1) detailing the historical evolution of platelet bioreactor design to recapitulate native platelet production ex vivo, and (2) identifying the associated challenges that still need to be addressed to further scale and validate these devices for commercial application. While platelets are among the first cells whose ex vivo production is spearheading major engineering advancements in microfluidic design, the resulting discoveries will undoubtedly extend to the production of other human tissues. This work is critical to identify the physiological characteristics of relevant 3D tissue-specific microenvironments that drive cell differentiation and elaborate upon how these are disrupted in disease. This is a burgeoning field whose future will define not only the ex vivo production of platelets and development of targeted therapies for thrombocytopenia, but the promise of regenerative medicine for the next century.


Subject(s)
Bioreactors , Blood Platelets , Cell Culture Techniques , Megakaryocytes , Animals , Blood Platelets/cytology , Blood Platelets/metabolism , Cell Culture Techniques/instrumentation , Cell Culture Techniques/methods , Humans , Megakaryocytes/cytology , Megakaryocytes/metabolism
7.
Am J Hum Genet ; 93(5): 906-14, 2013 Nov 07.
Article in English | MEDLINE | ID: mdl-24119684

ABSTRACT

We used exome sequencing to identify mutations in sideroflexin 4 (SFXN4) in two children with mitochondrial disease (the more severe case also presented with macrocytic anemia). SFXN4 is an uncharacterized mitochondrial protein that localizes to the mitochondrial inner membrane. sfxn4 knockdown in zebrafish recapitulated the mitochondrial respiratory defect observed in both individuals and the macrocytic anemia with megaloblastic features of the more severe case. In vitro and in vivo complementation studies with fibroblasts from the affected individuals and zebrafish demonstrated the requirement of SFXN4 for mitochondrial respiratory homeostasis and erythropoiesis. Our findings establish mutations in SFXN4 as a cause of mitochondriopathy and macrocytic anemia.


Subject(s)
Anemia, Macrocytic/genetics , Membrane Proteins/genetics , Mitochondrial Diseases/genetics , Adolescent , Animals , Child , Erythropoiesis/genetics , Exome , Female , Gene Knockdown Techniques , Humans , Mitochondrial Proteins/genetics , Mutation , Zebrafish/genetics
8.
Blood ; 124(12): 1857-67, 2014 Sep 18.
Article in English | MEDLINE | ID: mdl-25606631

ABSTRACT

Platelet transfusions total >2.17 million apheresis-equivalent units per year in the United States and are derived entirely from human donors, despite clinically significant immunogenicity, associated risk of sepsis, and inventory shortages due to high demand and 5-day shelf life. To take advantage of known physiological drivers of thrombopoiesis, we have developed a microfluidic human platelet bioreactor that recapitulates bone marrow stiffness, extracellular matrix composition,micro-channel size, hemodynamic vascular shear stress, and endothelial cell contacts, and it supports high-resolution live-cell microscopy and quantification of platelet production. Physiological shear stresses triggered proplatelet initiation, reproduced ex vivo bone marrow proplatelet production, and generated functional platelets. Modeling human bone marrow composition and hemodynamics in vitro obviates risks associated with platelet procurement and storage to help meet growing transfusion needs.


Subject(s)
Bioreactors , Blood Platelets , Microfluidic Analytical Techniques , Animals , Biomimetic Materials , Blood Platelets/cytology , Blood Platelets/physiology , Equipment Design , Humans , Megakaryocytes/cytology , Megakaryocytes/physiology , Mice , Models, Biological , Platelet Transfusion , Thrombopoiesis
9.
Blood ; 122(7): 1305-11, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23838351

ABSTRACT

During thrombopoiesis, megakaroycytes undergo extensive cytoskeletal remodeling to form proplatelet extensions that eventually produce mature platelets. Proplatelet formation is a tightly orchestrated process that depends on dynamic regulation of both tubulin reorganization and Rho-associated, coiled-coil containing protein kinase/RhoA activity. A disruption in tubulin dynamics or RhoA activity impairs proplatelet formation and alters platelet morphology. We previously observed that protein kinase Cepsilon (PKCε), a member of the protein kinase C family of serine/threonine-kinases, expression varies during human megakaryocyte differentiation and modulates megakaryocyte maturation and platelet release. Here we used an in vitro model of murine platelet production to investigate a potential role for PKCε in proplatelet formation. By immunofluorescence we observed that PKCε colocalizes with α/ß-tubulin in specific areas of the marginal tubular-coil in proplatelets. Moreover, we found that PKCε expression escalates during megakarocyte differentiation and remains elevated in proplatelets, whereas the active form of RhoA is substantially downregulated in proplatelets. PKCε inhibition resulted in lower proplatelet numbers and larger diameter platelets in culture as well as persistent RhoA activation. Finally, we demonstrate that pharmacological inhibition of RhoA is capable of reversing the proplatelet defects mediated by PKCε inhibition. Collectively, these data indicate that by regulating RhoA activity, PKCε is a critical mediator of mouse proplatelet formation in vitro.


Subject(s)
Blood Platelets/cytology , Megakaryocytes/cytology , Protein Kinase C-epsilon/metabolism , Thrombopoiesis/physiology , Tubulin/metabolism , rhoA GTP-Binding Protein/antagonists & inhibitors , Animals , Blood Platelets/metabolism , Blotting, Western , Cell Differentiation , Cells, Cultured , Fetus/cytology , Fetus/metabolism , Flow Cytometry , Fluorescent Antibody Technique , Humans , Liver/cytology , Liver/metabolism , Megakaryocytes/metabolism , Mice , RNA, Small Interfering/genetics , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
10.
Blood ; 121(1): 188-96, 2013 Jan 03.
Article in English | MEDLINE | ID: mdl-23160460

ABSTRACT

Wnt signaling is involved in numerous aspects of vertebrate development and homeostasis, including the formation and function of blood cells. Here, we show that canonical and noncanonical Wnt signaling pathways are present and functional in megakaryocytes (MKs), with several Wnt effectors displaying MK-restricted expression. Using the CHRF288-11 cell line as a model for human MKs, the canonical Wnt3a signal was found to induce a time and dose-dependent increase in ß-catenin expression. ß-catenin accumulation was inhibited by the canonical antagonist dickkopf-1 (DKK1) and by the noncanonical agonist Wnt5a. Whole genome expression analysis demonstrated that Wnt3a and Wnt5a regulated distinct patterns of gene expression in MKs, and revealed a further interplay between canonical and noncanonical Wnt pathways. Fetal liver cells derived from low-density-lipoprotein receptor-related protein 6-deficient mice (LRP6(-/-)), generated dramatically reduced numbers of MKs in culture of lower ploidy (2N and 4N) than wild-type controls, implicating LRP6-dependent Wnt signaling in MK proliferation and maturation. Finally, in wild-type mature murine fetal liver-derived MKs, Wnt3a potently induced proplatelet formation, an effect that could be completely abrogated by DKK1. These data identify novel extrinsic regulators of proplatelet formation, and reveal a profound role for Wnt signaling in platelet production.


Subject(s)
Megakaryocytes/cytology , Thrombopoiesis/physiology , Wnt Signaling Pathway/physiology , Animals , Blood Platelets/cytology , Cell Line , Cells, Cultured/drug effects , Humans , Intercellular Signaling Peptides and Proteins/pharmacology , Liver/embryology , Low Density Lipoprotein Receptor-Related Protein-6/deficiency , Low Density Lipoprotein Receptor-Related Protein-6/genetics , Megakaryocytes/drug effects , Megakaryocytes/metabolism , Mice , Mice, Knockout , Recombinant Proteins/pharmacology , Thrombopoiesis/genetics , Wnt Proteins/pharmacology , Wnt3A Protein/pharmacology , beta Catenin/biosynthesis , beta Catenin/genetics
11.
Br J Haematol ; 165(2): 227-36, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24499183

ABSTRACT

Platelets are essential for haemostasis, and thrombocytopenia (platelet counts <150 × 10(9) /l) is a major clinical problem encountered across a number of conditions, including immune thrombocytopenic purpura, myelodysplastic syndromes, chemotherapy, aplastic anaemia, human immunodeficiency virus infection, complications during pregnancy and delivery, and surgery. Circulating blood platelets are specialized cells that function to prevent bleeding and minimize blood vessel injury. Platelets circulate in their quiescent form, and upon stimulation, activate to release their granule contents and spread on the affected tissue to create a physical barrier that prevents blood loss. The current model of platelet formation states that large progenitor cells in the bone marrow, called megakaryocytes, release platelets by extending long, branching processes, designated proplatelets, into sinusoidal blood vessels. This review will focus on different factors that impact megakaryocyte development, proplatelet formation and platelet release. It will highlight recent studies on thrombopoeitin-dependent megakaryocyte maturation, endomitosis and granule formation, cytoskeletal contributions to proplatelet formation, the role of apoptosis, and terminal platelet formation and release.


Subject(s)
Megakaryocytes/physiology , Thrombopoiesis/physiology , Actins/metabolism , Animals , Apoptosis , Blood Platelets/cytology , Blood Platelets/metabolism , Cell Membrane/metabolism , Humans , Microtubules/metabolism , Protein Kinase C/metabolism , rho GTP-Binding Proteins/metabolism
12.
Blood ; 120(8): 1552-61, 2012 Aug 23.
Article in English | MEDLINE | ID: mdl-22665937

ABSTRACT

Platelet (PLT) production represents the final stage of megakaryocyte (MK) development. During differentiation, bone marrow MKs extend and release long, branched proPLTs into sinusoidal blood vessels, which undergo repeated abscissions to yield circulating PLTs. Circular-prePLTs are dynamic intermediate structures in this sequence that have the capacity to reversibly convert into barbell-proPLTs and may be related to "young PLTs" and "large PLTs" of both inherited and acquired macrothrombocytopenias. Conversion is regulated by the diameter and thickness of the peripheral microtubule coil, and PLTs are capable of enlarging in culture to generate barbell-proPLTs that divide to yield 2 smaller PLT products. Because PLT number and size are inversely proportional, this raises the question: do macrothrombocytopenias represent a failure in the intermediate stages of PLT production? This review aims to bring together and contextualize our current understanding of terminal PLT production against the backdrop of human macrothrombocytopenias to establish how "large PLTs" observed in both conditions are similar, how they are different, and what they can teach us about PLT formation. A better understanding of the cytoskeletal mechanisms that regulate PLT formation and determine PLT size offers the promise of improved therapies for clinical disorders of PLT production and an important source of PLTs for infusion.


Subject(s)
Blood Platelets/cytology , Blood Platelets/pathology , Thrombocytopenia/pathology , Thrombopoiesis , Animals , Blood Platelets/metabolism , Cell Size , Cytoskeleton/genetics , Cytoskeleton/metabolism , Cytoskeleton/pathology , Gene Expression Regulation , Humans , Thrombocytopenia/genetics , Thrombocytopenia/metabolism
13.
Blood ; 120(10): 1975-84, 2012 Sep 06.
Article in English | MEDLINE | ID: mdl-22665936

ABSTRACT

Proplatelet production represents a terminal stage of megakaryocyte development during which long, branching processes composed of platelet-sized swellings are extended and released into the surrounding culture. Whereas the cytoskeletal mechanics driving these transformations have been the focus of many studies, significant limitations in our ability to quantify the rate and extent of proplatelet production have restricted the field to qualitative analyses of a limited number of cells over short intervals. A novel high-content, quantitative, live-cell imaging assay using the IncuCyte system (Essen BioScience) was therefore developed to measure the rate and extent of megakaryocyte maturation and proplatelet production under live culture conditions for extended periods of time. As proof of concept, we used this system in the present study to establish a mechanism by which trastuzumab emtansine (T-DM1), an Ab-drug conjugate currently in clinical development for cancer, affects platelet production. High-content analysis of primary cell cultures revealed that T-DM1 is taken up by mouse megakaryocytes, inhibits megakaryocyte differentiation, and disrupts proplatelet formation by inducing abnormal tubulin organization and suppressing microtubule dynamic instability. Defining the pathways by which therapeutics such as T-DM1 affect megakaryocyte differentiation and proplatelet production may yield strategies to manage drug-induced thrombocytopenias.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Blood Platelets/drug effects , Cell Differentiation/drug effects , Maytansine/analogs & derivatives , Megakaryocytes/drug effects , Thrombocytopenia/chemically induced , Ado-Trastuzumab Emtansine , Animals , Biological Assay , Blood Platelets/physiology , Blood Platelets/ultrastructure , Cell Differentiation/physiology , Fetus , Flow Cytometry , Humans , Liver/cytology , Liver/drug effects , Liver/physiology , Maytansine/pharmacology , Megakaryocytes/physiology , Megakaryocytes/ultrastructure , Mice , Microscopy, Fluorescence , Microtubules/drug effects , Microtubules/ultrastructure , Molecular Imaging , Platelet Count , Primary Cell Culture , Thrombocytopenia/prevention & control , Thrombopoiesis/drug effects , Thrombopoiesis/physiology , Trastuzumab , Tubulin/metabolism
14.
Blood ; 120(11): 2317-29, 2012 Sep 13.
Article in English | MEDLINE | ID: mdl-22806889

ABSTRACT

Serum response factor and its transcriptional cofactor MKL1 are critical for megakaryocyte maturation and platelet formation. We show that MKL2, a homologue of MKL1, is expressed in megakaryocytes and plays a role in megakaryocyte maturation. Using a megakaryocyte-specific Mkl2 knockout (KO) mouse on the conventional Mkl1 KO background to produce double KO (DKO) megakaryocytes and platelets, a critical role for MKL2 is revealed. The decrease in megakaryocyte ploidy and platelet counts of DKO mice is more severe than in Mkl1 KO mice. Platelet dysfunction in DKO mice is revealed by prolonged bleeding times and ineffective platelet activation in vitro in response to adenosine 5'-diphosphate. Electron microscopy and immunofluorescence of DKO megakaryocytes and platelets indicate abnormal cytoskeletal and membrane organization with decreased granule complexity. Surprisingly, the DKO mice have a more extreme thrombocytopenia than mice lacking serum response factor (SRF) expression in the megakaryocyte compartment. Comparison of gene expression reveals approximately 4400 genes whose expression is differentially affected in DKO compared with megakaryocytes deficient in SRF, strongly suggesting that MKL1 and MKL2 have both SRF-dependent and SRF-independent activity in megakaryocytopoiesis.


Subject(s)
Blood Platelets/cytology , Blood Platelets/metabolism , Hematopoiesis , Megakaryocytes/cytology , Megakaryocytes/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism , Adenosine Diphosphate/metabolism , Animals , Bleeding Time , Blood Platelets/ultrastructure , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cells, Cultured , Crosses, Genetic , Cytoplasm/metabolism , Cytoplasm/ultrastructure , Cytoskeleton/metabolism , Cytoskeleton/ultrastructure , Gene Expression Profiling , Megakaryocytes/ultrastructure , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Platelet Activation , Thrombocytopenia/etiology , Trans-Activators/genetics , Transcription Factors/genetics
15.
Blood ; 120(24): 4859-68, 2012 Dec 06.
Article in English | MEDLINE | ID: mdl-22972982

ABSTRACT

We recently identified 68 genomic loci where common sequence variants are associated with platelet count and volume. Platelets are formed in the bone marrow by megakaryocytes, which are derived from hematopoietic stem cells by a process mainly controlled by transcription factors. The homeobox transcription factor MEIS1 is uniquely transcribed in megakaryocytes and not in the other lineage-committed blood cells. By ChIP-seq, we show that 5 of the 68 loci pinpoint a MEIS1 binding event within a group of 252 MK-overexpressed genes. In one such locus in DNM3, regulating platelet volume, the MEIS1 binding site falls within a region acting as an alternative promoter that is solely used in megakaryocytes, where allelic variation dictates different levels of a shorter transcript. The importance of dynamin activity to the latter stages of thrombopoiesis was confirmed by the observation that the inhibitor Dynasore reduced murine proplatelet for-mation in vitro.


Subject(s)
Blood Platelets/metabolism , Dynamin III/genetics , Genome, Human/genetics , Homeodomain Proteins/genetics , Megakaryocytes/metabolism , Neoplasm Proteins/genetics , Promoter Regions, Genetic/genetics , Animals , Binding Sites/genetics , Blood Platelets/drug effects , Cell Line, Tumor , Cell Lineage/genetics , Cells, Cultured , Chromatin Immunoprecipitation , Gene Expression , Genetic Variation , Homeodomain Proteins/metabolism , Humans , Hydrazones/pharmacology , Mice , Myeloid Ecotropic Viral Integration Site 1 Protein , Neoplasm Proteins/metabolism , Platelet Count , Polymorphism, Single Nucleotide , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA , Transcription Initiation Site , Transcription, Genetic
16.
Blood ; 118(6): 1641-52, 2011 Aug 11.
Article in English | MEDLINE | ID: mdl-21566095

ABSTRACT

Megakaryocytes generate platelets by remodeling their cytoplasm first into proplatelets and then into preplatelets, which undergo fission to generate platelets. Although the functions of microtubules and actin during platelet biogenesis have been defined, the role of the spectrin cytoskeleton is unknown. We investigated the function of the spectrin-based membrane skeleton in proplatelet and platelet production in murine megakaryocytes. Electron microscopy revealed that, like circulating platelets, proplatelets have a dense membrane skeleton, the main fibrous component of which is spectrin. Unlike other cells, megakaryocytes and their progeny express both erythroid and nonerythroid spectrins. Assembly of spectrin into tetramers is required for invaginated membrane system maturation and proplatelet extension, because expression of a spectrin tetramer-disrupting construct in megakaryocytes inhibits both processes. Incorporation of this spectrin-disrupting fragment into a novel permeabilized proplatelet system rapidly destabilizes proplatelets, causing blebbing and swelling. Spectrin tetramers also stabilize the "barbell shapes" of the penultimate stage in platelet production, because addition of the tetramer-disrupting construct converts these barbell shapes to spheres, demonstrating that membrane skeletal continuity maintains the elongated, pre-fission shape. The results of this study provide evidence for a role for spectrin in different steps of megakaryocyte development through its participation in the formation of invaginated membranes and in the maintenance of proplatelet structure.


Subject(s)
Blood Platelets/metabolism , Cytoskeleton/metabolism , Megakaryocyte Progenitor Cells/metabolism , Megakaryocytes/metabolism , Spectrin/metabolism , Actins/metabolism , Animals , Blood Platelets/cytology , Blood Platelets/ultrastructure , Blotting, Western , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Cells, Cultured , Cytoskeleton/ultrastructure , Erythroid Cells/metabolism , Megakaryocyte Progenitor Cells/cytology , Megakaryocyte Progenitor Cells/ultrastructure , Megakaryocytes/cytology , Megakaryocytes/ultrastructure , Mice , Microscopy, Electron , Microtubules/metabolism , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Multimerization , Reverse Transcriptase Polymerase Chain Reaction , Spectrin/chemistry , Spectrin/genetics
17.
Blood ; 115(6): 1267-76, 2010 Feb 11.
Article in English | MEDLINE | ID: mdl-19846887

ABSTRACT

The spontaneous mouse mutation "thrombocytopenia and cardiomyopathy" (trac) causes macrothrombocytopenia, prolonged bleeding times, anemia, leukopenia, infertility, cardiomyopathy, and shortened life span. Homozygotes show a 20-fold decrease in platelet numbers and a 3-fold increase in platelet size with structural alterations and functional impairments in activation and aggregation. Megakaryocytes in trac/trac mice are present in increased numbers, have poorly developed demarcation membrane systems, and have decreased polyploidy. The thrombocytopenia is not intrinsic to defects at the level of hematopoietic progenitor cells but is associated with a microenvironmental abnormality. The trac mutation maps to mouse chromosome 17, syntenic with human chromosome 2p21-22. A G to A mutation in exon 10 of the adenosine triphosphate (ATP)-binding cassette subfamily G, member 5 (Abcg5) gene, alters a tryptophan codon (UGG) to a premature stop codon (UAG). Crosses with mice doubly transgenic for the human ABCG5 and ABCG8 genes rescued platelet counts and volumes. ABCG5 and ABCG8 form a functional complex that limits dietary phytosterol accumulation. Phytosterolemia in trac/trac mice confirmed a functional defect in the ABCG5/ABCG8 transport system. The trac mutation provides a new clinically significant animal model for human phytosterolemia and provides a new means for studying the role of phytosterols in hematologic diseases and testing therapeutic interventions.


Subject(s)
ATP-Binding Cassette Transporters/physiology , Cardiomyopathies/genetics , Disease Models, Animal , Lipid Metabolism, Inborn Errors/genetics , Lipoproteins/physiology , Mutation/genetics , Phytosterols/metabolism , Sitosterols/metabolism , Thrombocytopenia/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 5 , ATP Binding Cassette Transporter, Subfamily G, Member 8 , ATP-Binding Cassette Transporters/genetics , Animals , Bleeding Time , Cardiomyopathies/pathology , Cells, Cultured , Colony-Forming Units Assay , Crosses, Genetic , Female , Fetus/cytology , Fetus/metabolism , Lipid Metabolism, Inborn Errors/pathology , Lipoproteins/genetics , Liver/cytology , Liver/metabolism , Male , Megakaryocytes/cytology , Megakaryocytes/metabolism , Mice , Mice, Inbred A , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Thrombocytopenia/pathology
18.
Handb Exp Pharmacol ; (210): 3-22, 2012.
Article in English | MEDLINE | ID: mdl-22918725

ABSTRACT

Platelets are anucleate, discoid cells, roughly 2-3 µm in diameter that function primarily as regulators of hemostasis, but also play secondary roles in angiogensis and innate immunity. Although human adults contain nearly one trillion platelets in circulation that are turned over every 8-10 days, our understanding of the mechanisms involved in platelet production is still incomplete. Platelets stem from large (30-100 µm) nucleated cells called megakaryocytes that reside primarily in the bone marrow. During maturation megakaryocytes extend long proplatelet elongations into sinusoidal blood vessels from which platelets ultimately release. During this process, platelets develop a number of distinguishable structural elements including: a delimited plasma membrane; invaginations of the surface membrane that form the open canalicular system (OCS); a closed-channel network of residual endoplasmic reticulum that form the dense tubular system (DTS); a spectrin-based membrane skeleton; an actin-based cytoskeletal network; a peripheral band of microtubules; and numerous organelles including α-granules, dense-granules, peroxisomes, lysosomes, and mitochondria. Proplatelet elongation and platelet production is an elaborate and complex process that defines the morphology and ultrastructure of circulating platelets, and is critical in understanding their increasingly numerous and varied biological functions.


Subject(s)
Blood Platelets/physiology , Blood Platelets/ultrastructure , Animals , Blood Platelets/cytology , Cytoskeleton/ultrastructure , Humans , Megakaryocytes/physiology , Organelles/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL