Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 8 de 8
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
Blood ; 136(6): 715-725, 2020 08 06.
Article de Anglais | MEDLINE | ID: mdl-32384141

RÉSUMÉ

Studies of inherited platelet disorders have provided many insights into platelet development and function. Loss of function of neurobeachin-like 2 (NBEAL2) causes gray platelet syndrome (GPS), where the absence of platelet α-granules indicates NBEAL2 is required for their production by precursor megakaryocytes. The endoplasmic reticulum is a dynamic network that interacts with numerous intracellular vesicles and organelles and plays key roles in their development. The megakaryocyte endoplasmic reticulum is extensive, and in this study we investigated its role in the biogenesis of α-granules by focusing on the membrane-resident trafficking protein SEC22B. Coimmunoprecipitation (co-IP) experiments using tagged proteins expressed in human HEK293 and megakaryocytic immortalized megakaryocyte progenitor (imMKCL) cells established binding of NBEAL2 with SEC22B, and demonstrated that NBEAL2 can simultaneously bind SEC22B and P-selectin. NBEAL2-SEC22B binding was also observed for endogenous proteins in human megakaryocytes using co-IP, and immunofluorescence microscopy detected substantial overlap. SEC22B binding was localized to a region of NBEAL2 spanning amino acids 1798 to 1903, where 2 GPS-associated missense variants have been reported: E1833K and R1839C. NBEAL2 containing either variant did not bind SEC22B coexpressed in HEK293 cells. CRISPR/Cas9-mediated knockout of SEC22B in imMKCL cells resulted in decreased NBEAL2, but not vice versa. Loss of either SEC22B or NBEAL2 expression resulted in failure of α-granule production and reduced granule proteins in imMKCL cells. We conclude that SEC22B is required for α-granule biogenesis in megakaryocytes, and that interactions with SEC22B and P-selectin facilitate the essential role of NBEAL2 in granule development and cargo stability.


Sujet(s)
Protéines du sang/physiologie , Granulations cytoplasmiques/physiologie , Réticulum endoplasmique/physiologie , Mégacaryocytes/ultrastructure , Biogenèse des organelles , Protéines R-SNARE/physiologie , Sites de fixation , Protéines du sang/déficit , Protéines du sang/génétique , Cellules cultivées , Techniques de knock-out de gènes , Syndrome des plaquettes grises/génétique , Cellules HEK293 , Humains , Immunoprécipitation , Progéniteurs mégacaryocytaires , Mégacaryocytes/métabolisme , Microscopie confocale , Microscopie de fluorescence , Mutation faux-sens , Sélectine P/physiologie , Cartographie d'interactions entre protéines , Protéines recombinantes/métabolisme
2.
Arterioscler Thromb Vasc Biol ; 38(10): 2435-2447, 2018 10.
Article de Anglais | MEDLINE | ID: mdl-30354215

RÉSUMÉ

Objective- Human and mouse megakaryocytes lacking NBEAL2 (neurobeachin-like 2) produce platelets where α-granules lack protein cargo. This cargo is mostly megakaryocyte-synthesized, but some proteins, including FGN (fibrinogen), are endocytosed. In this study, we examined the trafficking of both types of cargo within primary megakaryocytes cultured from normal and NBEAL2-null mice, to determine the role of NBEAL2 in α-granule maturation. We also examined the interaction of NBEAL2 with the granule-associated protein P-selectin in human megakaryocytes and platelets. Approach and Results- Fluorescence microscopy was used to compare uptake of labeled FGN by normal and NBEAL2-null mouse megakaryocytes, which was similar in both. NBEAL2-null cells, however, showed decreased FGN retention, and studies with biotinylated protein showed rapid loss rather than increased degradation. Intracellular tracking via fluorescence microscopy revealed that in normal megakaryocytes, endocytosed FGN sequentially associated with compartments expressing RAB5 (Ras-related protein in brain 5), RAB7 (Ras-related protein in brain 7), and P-selectin, where it was retained. A similar initial pattern was observed in NBEAL2-null megakaryocytes, but then FGN passed from the P-selectin compartment to RAB11 (Ras-related protein in brain 11)-associated endosomes before release. Megakaryocyte-synthesized VWF (Von Willebrand factor) was observed to follow the same route out of NBEAL2-null cells. Immunofluorescence microscopy revealed intracellular colocalization of NBEAL2 with P-selectin in human megakaryocytes, proplatelets, and platelets. Native NBEAL2 and P-selectin were coimmunoprecipitated from platelets and megakaryocytes. Conclusions- NBEAL2 is not required for FGN uptake by megakaryocytes. NBEAL2 is required for the retention of both endocytosed and megakaryocyte-synthesized proteins by maturing α-granules, and possibly by platelet-borne granules. This function may involve interaction of NBEAL2 with P-selectin.


Sujet(s)
Protéines du sang/métabolisme , Granulations cytoplasmiques/métabolisme , Endocytose , Fibrinogène/métabolisme , Mégacaryocytes/métabolisme , Animaux , Protéines du sang/déficit , Protéines du sang/génétique , Cellules cultivées , Endosomes/métabolisme , Femelle , Mâle , Souris knockout , Sélectine P/métabolisme , Transport des protéines , Voie de sécrétion , Protéines G rab/métabolisme , Protéines G rab5/métabolisme , Protéines Rab7 liant le GTP , Facteur de von Willebrand/métabolisme
3.
Nat Commun ; 8: 14816, 2017 04 03.
Article de Anglais | MEDLINE | ID: mdl-28368018

RÉSUMÉ

Human actin-related protein 2/3 complex (Arp2/3), required for actin filament branching, has two ARPC1 component isoforms, with ARPC1B prominently expressed in blood cells. Here we show in a child with microthrombocytopenia, eosinophilia and inflammatory disease, a homozygous frameshift mutation in ARPC1B (p.Val91Trpfs*30). Platelet lysates reveal no ARPC1B protein and greatly reduced Arp2/3 complex. Missense ARPC1B mutations are identified in an unrelated patient with similar symptoms and ARPC1B deficiency. ARPC1B-deficient platelets are microthrombocytes similar to those seen in Wiskott-Aldrich syndrome that show aberrant spreading consistent with loss of Arp2/3 function. Knockout of ARPC1B in megakaryocytic cells results in decreased proplatelet formation, and as observed in platelets from patients, increased ARPC1A expression. Thus loss of ARPC1B produces a unique set of platelet abnormalities, and is associated with haematopoietic/immune symptoms affecting cell lineages where this isoform predominates. In agreement with recent experimental studies, our findings suggest that ARPC1 isoforms are not functionally interchangeable.


Sujet(s)
Complexe Arp-2-3/déficit , Anomalies des plaquettes/métabolisme , Plaquettes/métabolisme , Inflammation/anatomopathologie , Complexe Arp-2-3/métabolisme , Actines/métabolisme , Plaquettes/effets des médicaments et des substances chimiques , Plaquettes/anatomopathologie , Plaquettes/ultrastructure , Cellules de la moelle osseuse/effets des médicaments et des substances chimiques , Cellules de la moelle osseuse/métabolisme , Cellules de la moelle osseuse/anatomopathologie , Forme de la cellule , Prédisposition aux maladies , Fibrinogène/pharmacologie , Techniques de knock-out de gènes , Humains , Mégacaryocytes/effets des médicaments et des substances chimiques , Mégacaryocytes/métabolisme , Mégacaryocytes/anatomopathologie , Mutation/génétique , Vascularite/anatomopathologie , Syndrome de Wiskott-Aldrich/anatomopathologie
4.
Platelets ; 28(2): 147-154, 2017 Mar.
Article de Anglais | MEDLINE | ID: mdl-28277061

RÉSUMÉ

Platelets are critical to hemostasis and thrombosis. Upon detecting injury, platelets show a range of responses including the release of protein cargo from α-granules. This cargo is synthesized by platelet precursor megakaryocytes or endocytosed by megakaryocytes and/or platelets. Insights into α-granule biogenesis have come from studies of hereditary conditions where these granules are immature, deficient or absent. Studies of Arthrogryposis, Renal dysfunction, and Cholestasis (ARC) syndrome identified the first proteins essential to α-granule biogenesis: VPS33B and VPS16B. VPS33B and VPS16B form a complex, and in the absence of either, platelets lack α-granules and the granule-specific membrane protein P-selectin. Gray Platelet Syndrome (GPS) platelets also lack conventionally recognizable α-granules, although P-selectin containing structures are present. GPS arises from mutations affecting NBEAL2. The GPS phenotype is more benign than ARC syndrome, but it can cause life-threatening bleeding, progressive thrombocytopenia, and myelofibrosis. We review the essential roles of VPS33B, VPS16B, and NBEAL2 in α-granule development. We also examine the existing data on their mechanisms of action, where many details remain poorly understood. VPS33B and VPS16B are ubiquitously expressed and ARC syndrome is a multisystem disorder that causes lethality early in life. Thus, VPS33B and VPS16B are clearly involved in other processes besides α-granule biogenesis. Studies of their involvement in vesicular trafficking and protein interactions are reviewed to gain insights into their roles in α-granule formation. NBEAL2 mutations primarily affect megakaryocytes and platelets, and while little is known about NBEAL2 function some insights can be gained from studies of related proteins, such as LYST.


Sujet(s)
Plaquettes/métabolisme , Granulations cytoplasmiques/métabolisme , Animaux , Arthrogrypose/diagnostic , Arthrogrypose/étiologie , Arthrogrypose/métabolisme , Transport biologique , Plaquettes/ultrastructure , Cholestase/diagnostic , Cholestase/étiologie , Cholestase/métabolisme , Granulations cytoplasmiques/ultrastructure , Syndrome des plaquettes grises/diagnostic , Syndrome des plaquettes grises/étiologie , Syndrome des plaquettes grises/métabolisme , Humains , Mégacaryocytes/métabolisme , Mutation , Phénotype , Insuffisance rénale/diagnostic , Insuffisance rénale/étiologie , Insuffisance rénale/métabolisme , Vésicules de sécrétion/métabolisme , Protéines du transport vésiculaire/génétique , Protéines du transport vésiculaire/métabolisme
5.
Arterioscler Thromb Vasc Biol ; 36(6): 1164-73, 2016 06.
Article de Anglais | MEDLINE | ID: mdl-27079884

RÉSUMÉ

OBJECTIVE: Thiol isomerases facilitate protein folding in the endoplasmic reticulum, and several of these enzymes, including protein disulfide isomerase and ERp57, are mobilized to the surface of activated platelets, where they influence platelet aggregation, blood coagulation, and thrombus formation. In this study, we examined the synthesis and trafficking of thiol isomerases in megakaryocytes, determined their subcellular localization in platelets, and identified the cellular events responsible for their movement to the platelet surface on activation. APPROACH AND RESULTS: Immunofluorescence microscopy imaging was used to localize protein disulfide isomerase and ERp57 in murine and human megakaryocytes at various developmental stages. Immunofluorescence microscopy and subcellular fractionation analysis were used to localize these proteins in platelets to a compartment distinct from known secretory vesicles that overlaps with an inner cell-surface membrane region defined by the endoplasmic/sarcoplasmic reticulum proteins calnexin and sarco/endoplasmic reticulum calcium ATPase 3. Immunofluorescence microscopy and flow cytometry were used to monitor thiol isomerase mobilization in activated platelets in the presence and absence of actin polymerization (inhibited by latrunculin) and in the presence or absence of membrane fusion mediated by Munc13-4 (absent in platelets from Unc13d(Jinx) mice). CONCLUSIONS: Platelet-borne thiol isomerases are trafficked independently of secretory granule contents in megakaryocytes and become concentrated in a subcellular compartment near the inner surface of the platelet outer membrane corresponding to the sarco/endoplasmic reticulum of these cells. Thiol isomerases are mobilized to the surface of activated platelets via a process that requires actin polymerization but not soluble N-ethylmaleimide-sensitive fusion protein attachment receptor/Munc13-4-dependent vesicular-plasma membrane fusion.


Sujet(s)
Plaquettes/enzymologie , Membrane cellulaire/enzymologie , Mégacaryocytes/enzymologie , Activation plaquettaire , Protein Disulfide-Isomerases/sang , Actines/sang , Animaux , Plaquettes/effets des médicaments et des substances chimiques , Protéines du sang/déficit , Protéines du sang/génétique , Calnexine/sang , Membrane cellulaire/effets des médicaments et des substances chimiques , Génotype , Humains , Mégacaryocytes/effets des médicaments et des substances chimiques , Fusion membranaire , Protéines membranaires/sang , Protéines membranaires/déficit , Protéines membranaires/génétique , Souris , Souris de lignée C57BL , Souris knockout , Phénotype , Activation plaquettaire/effets des médicaments et des substances chimiques , Protein Disulfide-Isomerases/biosynthèse , Transport des protéines , Sarcoplasmic Reticulum Calcium-Transporting ATPases/sang
6.
Blood ; 126(1): 80-8, 2015 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-25838348

RÉSUMÉ

Bin-Amphiphysin-Rvs (BAR) and Fes-CIP4 homology BAR (F-BAR) proteins generate tubular membrane invaginations reminiscent of the megakaryocyte (MK) demarcation membrane system (DMS), which provides membranes necessary for future platelets. The F-BAR protein PACSIN2 is one of the most abundant BAR/F-BAR proteins in platelets and the only one reported to interact with the cytoskeletal and scaffold protein filamin A (FlnA), an essential regulator of platelet formation and function. The FlnA-PACSIN2 interaction was therefore investigated in MKs and platelets. PACSIN2 associated with FlnA in human platelets. The interaction required FlnA immunoglobulin-like repeat 20 and the tip of PACSIN2 F-BAR domain and enhanced PACSIN2 F-BAR domain membrane tubulation in vitro. Most human and wild-type mouse platelets had 1 to 2 distinct PACSIN2 foci associated with cell membrane GPIbα, whereas Flna-null platelets had 0 to 4 or more foci. Endogenous PACSIN2 and transfected enhanced green fluorescent protein-PACSIN2 were concentrated in midstage wild-type mouse MKs in a well-defined invagination of the plasma membrane reminiscent of the initiating DMS and dispersed in the absence of FlnA binding. The DMS appeared less well defined, and platelet territories were not readily visualized in Flna-null MKs. We conclude that the FlnA-PACSIN2 interaction regulates membrane tubulation in MKs and platelets and likely contributes to DMS formation.


Sujet(s)
Protéines adaptatrices de la transduction du signal/métabolisme , Plaquettes , Membrane cellulaire/ultrastructure , Filamines/métabolisme , Mégacaryocytes , Protéines adaptatrices de la transduction du signal/composition chimique , Animaux , Plaquettes/métabolisme , Plaquettes/ultrastructure , Membrane cellulaire/métabolisme , Cellules cultivées , Filamines/physiologie , Cellules HEK293 , Humains , Mégacaryocytes/métabolisme , Mégacaryocytes/ultrastructure , Souris , Souris de lignée C57BL , Souris transgéniques , Liaison aux protéines/physiologie , Motifs et domaines d'intéraction protéique/physiologie , Pseudopodes/métabolisme
7.
Nat Genet ; 47(5): 535-538, 2015 May.
Article de Anglais | MEDLINE | ID: mdl-25807284

RÉSUMÉ

Some familial platelet disorders are associated with predisposition to leukemia, myelodysplastic syndrome (MDS) or dyserythropoietic anemia. We identified a family with autosomal dominant thrombocytopenia, high erythrocyte mean corpuscular volume (MCV) and two occurrences of B cell-precursor acute lymphoblastic leukemia (ALL). Whole-exome sequencing identified a heterozygous single-nucleotide change in ETV6 (ets variant 6), c.641C>T, encoding a p.Pro214Leu substitution in the central domain, segregating with thrombocytopenia and elevated MCV. A screen of 23 families with similar phenotypes identified 2 with ETV6 mutations. One family also had a mutation encoding p.Pro214Leu and one individual with ALL. The other family had a c.1252A>G transition producing a p.Arg418Gly substitution in the DNA-binding domain, with alternative splicing and exon skipping. Functional characterization of these mutations showed aberrant cellular localization of mutant and endogenous ETV6, decreased transcriptional repression and altered megakaryocyte maturation. Our findings underscore a key role for ETV6 in platelet formation and leukemia predisposition.


Sujet(s)
Hémopathies/génétique , Protéines proto-oncogènes c-ets/génétique , Protéines de répression/génétique , Thrombopénie/génétique , Adulte , Enfant d'âge préscolaire , Analyse de mutations d'ADN , Érythrocytes anormaux , Exome , Femelle , Études d'associations génétiques , Prédisposition génétique à une maladie , Mutation germinale , Cellules HEK293 , Humains , Mâle , Mutation faux-sens , Pedigree , Leucémie-lymphome lymphoblastique à précurseurs B et T/génétique ,
8.
Blood ; 122(19): 3349-58, 2013 Nov 07.
Article de Anglais | MEDLINE | ID: mdl-23861251

RÉSUMÉ

Gray platelet syndrome (GPS) is an inherited bleeding disorder associated with macrothrombocytopenia and α-granule-deficient platelets. GPS has been linked to loss of function mutations in NEABL2 (neurobeachin-like 2), and we describe here a murine GPS model, the Nbeal2(-/-) mouse. As in GPS, Nbeal2(-/-) mice exhibit splenomegaly, macrothrombocytopenia, and a deficiency of platelet α-granules and their cargo, including von Willebrand factor (VWF), thrombospondin-1, and platelet factor 4. The platelet α-granule membrane protein P-selectin is expressed at 48% of wild-type levels and externalized upon platelet activation. The presence of P-selectin and normal levels of VPS33B and VPS16B in Nbeal2(-/-) platelets suggests that NBEAL2 acts independently of VPS33B/VPS16B at a later stage of α-granule biogenesis. Impaired Nbeal2(-/-) platelet function was shown by flow cytometry, platelet aggregometry, bleeding assays, and intravital imaging of laser-induced arterial thrombus formation. Microscopic analysis detected marked abnormalities in Nbeal2(-/-) bone marrow megakaryocytes, which when cultured showed delayed maturation, decreased survival, decreased ploidy, and developmental abnormalities, including abnormal extracellular distribution of VWF. Our results confirm that α-granule secretion plays a significant role in platelet function, and they also indicate that abnormal α-granule formation in Nbeal2(-/-) mice has deleterious effects on megakaryocyte survival, development, and platelet production.


Sujet(s)
Plaquettes/anatomopathologie , Protéines du sang/génétique , Moelle osseuse/anatomopathologie , Granulations cytoplasmiques/anatomopathologie , Syndrome des plaquettes grises/anatomopathologie , Mégacaryocytes/anatomopathologie , Animaux , Plaquettes/métabolisme , Protéines du sang/déficit , Moelle osseuse/métabolisme , Protéines de transport/génétique , Protéines de transport/métabolisme , Différenciation cellulaire , Granulations cytoplasmiques/métabolisme , Modèles animaux de maladie humaine , Femelle , Expression des gènes , Syndrome des plaquettes grises/génétique , Syndrome des plaquettes grises/métabolisme , Mâle , Mégacaryocytes/métabolisme , Souris , Souris knockout , Sélectine P/génétique , Sélectine P/métabolisme , Agrégation plaquettaire , Facteur-4 plaquettaire/génétique , Facteur-4 plaquettaire/métabolisme , Isoformes de protéines/déficit , Isoformes de protéines/génétique , Thrombospondine-1/génétique , Thrombospondine-1/métabolisme , Facteur de von Willebrand/génétique , Facteur de von Willebrand/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE