Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 83
Filtrar
1.
Blood ; 127(11): 1468-80, 2016 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-26744461

RESUMO

Platelets are essential for hemostasis, and thrombocytopenia is a major clinical problem. Megakaryocytes (MKs) generate platelets by extending long processes, proplatelets, into sinusoidal blood vessels. However, very little is known about what regulates proplatelet formation. To uncover which proteins were dynamically changing during this process, we compared the proteome and transcriptome of round vs proplatelet-producing MKs by 2D difference gel electrophoresis (DIGE) and polysome profiling, respectively. Our data revealed a significant increase in a poorly-characterized MK protein, myristoylated alanine-rich C-kinase substrate (MARCKS), which was upregulated 3.4- and 5.7-fold in proplatelet-producing MKs in 2D DIGE and polysome profiling analyses, respectively. MARCKS is a protein kinase C (PKC) substrate that binds PIP2. In MKs, it localized to both the plasma and demarcation membranes. MARCKS inhibition by peptide significantly decreased proplatelet formation 53%. To examine the role of MARCKS in the PKC pathway, we treated MKs with polymethacrylate (PMA), which markedly increased MARCKS phosphorylation while significantly inhibiting proplatelet formation 84%, suggesting that MARCKS phosphorylation reduces proplatelet formation. We hypothesized that MARCKS phosphorylation promotes Arp2/3 phosphorylation, which subsequently downregulates proplatelet formation; both MARCKS and Arp2 were dephosphorylated in MKs making proplatelets, and Arp2 inhibition enhanced proplatelet formation. Finally, we used MARCKS knockout (KO) mice to probe the direct role of MARCKS in proplatelet formation; MARCKS KO MKs displayed significantly decreased proplatelet levels. MARCKS expression and signaling in primary MKs is a novel finding. We propose that MARCKS acts as a "molecular switch," binding to and regulating PIP2 signaling to regulate processes like proplatelet extension (microtubule-driven) vs proplatelet branching (Arp2/3 and actin polymerization-driven).


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Megacariócitos/metabolismo , Proteínas de Membrana/fisiologia , Processamento de Proteína Pós-Traducional , Trombopoese/fisiologia , Complexo 2-3 de Proteínas Relacionadas à Actina/metabolismo , Proteína 3 Relacionada a Actina/metabolismo , Sequência de Aminoácidos , Proteína 2 Semelhante a Angiopoietina , Proteínas Semelhantes a Angiopoietina , Angiopoietinas/metabolismo , Animais , Apoptose , Plaquetas/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/deficiência , Fígado/citologia , Fígado/embriologia , Proteínas de Membrana/deficiência , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Substrato Quinase C Rico em Alanina Miristoilada , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/farmacologia , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosforilação , Biossíntese de Proteínas , Proteína Quinase C/metabolismo , Transdução de Sinais
2.
Blood ; 125(5): 860-8, 2015 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-25411426

RESUMO

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.


Assuntos
Plaquetas/metabolismo , Dineínas do Citoplasma/metabolismo , Megacariócitos/metabolismo , Microtúbulos/metabolismo , Tubulina (Proteína)/metabolismo , Animais , Plaquetas/citologia , Diferenciação Celular , Citoplasma/metabolismo , Dineínas do Citoplasma/genética , Recuperação de Fluorescência Após Fotodegradação , Expressão Gênica , Mecanotransdução Celular , Megacariócitos/citologia , Camundongos , Microscopia de Interferência , Microtúbulos/química , Cultura Primária de Células , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Estresse Mecânico , Trombopoese/genética , Tubulina (Proteína)/genética
3.
Blood ; 126(1): 80-8, 2015 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-25838348

RESUMO

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.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Plaquetas , Membrana Celular/ultraestrutura , Filaminas/metabolismo , Megacariócitos , Proteínas Adaptadoras de Transdução de Sinal/química , Animais , Plaquetas/metabolismo , Plaquetas/ultraestrutura , Membrana Celular/metabolismo , Células Cultivadas , Filaminas/fisiologia , Células HEK293 , Humanos , Megacariócitos/metabolismo , Megacariócitos/ultraestrutura , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Ligação Proteica/fisiologia , Domínios e Motivos de Interação entre Proteínas/fisiologia , Pseudópodes/metabolismo
4.
Traffic ; 15(8): 819-38, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24891099

RESUMO

Dynamin is a 96-kDa protein that has multiple oligomerization states that influence its GTPase activity. A number of different dynamin effectors, including lipids, actin filaments, and SH3-domain-containing proteins, have been implicated in the regulation of dynamin oligomerization, though their roles in influencing dynamin oligomerization have been studied predominantly in vitro using recombinant proteins. Here, we identify higher order dynamin oligomers such as rings and helices in vitro and in live cells using fluorescence lifetime imaging microscopy (FLIM). FLIM detected GTP- and actin-dependent dynamin oligomerization at distinct cellular sites, including the cell membrane and transition zones where cortical actin transitions into stress fibers. Our study identifies a major role for direct dynamin-actin interactions and dynamin's GTPase activity in the regulation of dynamin oligomerization in cells.


Assuntos
Actinas/metabolismo , Dinaminas/metabolismo , Guanosina Trifosfato/metabolismo , Multimerização Proteica , Actinas/química , Sequência de Aminoácidos , Animais , Células COS , Chlorocebus aethiops , Dinaminas/química , Camundongos , Dados de Sequência Molecular , Ligação Proteica , Estrutura Terciária de Proteína
5.
Biochem Biophys Res Commun ; 469(3): 659-64, 2016 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-26707877

RESUMO

Filamin A (FLNA) is an actin filament crosslinking protein with multiple intracellular binding partners. Mechanical force exposes cryptic FLNA binding sites for some of these ligands. To identify new force-dependent binding interactions, we used a fusion construct composed of two FLNA domains, one of which was previously identified as containing a force-dependent binding site as a bait in a yeast two-hybrid system and identified the Rho dissociation inhibitor 2 (RhoGDI2) as a potential interacting partner. A RhoGDI2 truncate with 81 N-terminal amino acid residues and a phosphomimetic mutant, RhoGDI(Tyr153Glu) interacted with the FLNA construct. However, neither wild-type or full-length RhoGDI2 phosphorylated at Y153 interacted with FLNA. Our interpretation of these contradictions is that truncation and/or mutation of RhoGDI2 perturbs its conformation to expose a site that adventitiously binds FLNA and is not a bona-fide interaction. Therefore, previous studies reporting that a RhoGDI(Y153E) mutant suppresses the metastasis of human bladder cancer cells must be reinvestigated in light of artificial interaction of this point mutant with FLNA.


Assuntos
Filaminas/química , Filaminas/metabolismo , Inibidor beta de Dissociação do Nucleotídeo Guanina rho/química , Inibidor beta de Dissociação do Nucleotídeo Guanina rho/metabolismo , Sítios de Ligação , Células HEK293 , Humanos , Fosforilação , Ligação Proteica
6.
Blood ; 121(14): 2743-52, 2013 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-23372168

RESUMO

Three isoforms of phosphatidylinositol-4-phosphate 5-kinase (PIP5KIα, PIP5KIß, and PIP5KIγ) can each catalyze the final step in the synthesis of phosphatidylinositol-4,5-bisphosphate (PIP2), which in turn can be either converted to second messengers or bind directly to and thereby regulate proteins such as talin. A widely quoted model speculates that only p90, a longer splice form of platelet-specific PIP5KIγ, but not the shorter p87 PIP5KIγ, regulates the ligand-binding activity of integrins via talin. However, when we used mice genetically engineered to lack only p90 PIP5KIγ, we found that p90 PIP5KIγ is not critical for integrin activation or platelet adhesion on collagen. However, p90 PIP5KIγ-null platelets do have impaired anchoring of their integrins to the underlying cytoskeleton. Platelets lacking both the p90 and p87 PIP5KIγ isoforms had normal integrin activation and actin dynamics, but impaired anchoring of their integrins to the cytoskeleton. Most importantly, they formed weak shear-resistant adhesions ex vivo and unstable vascular occlusions in vivo. Together, our studies demonstrate that, although PIP5KIγ is essential for normal platelet function, individual isoforms of PIP5KIγ fulfill unique roles for the integrin-dependent integrity of the membrane cytoskeleton and for the stabilization of platelet adhesion.


Assuntos
Plaquetas/citologia , Plaquetas/enzimologia , Integrinas/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Adesividade Plaquetária/fisiologia , Trombose/enzimologia , Citoesqueleto de Actina/fisiologia , Processamento Alternativo/genética , Animais , Citoesqueleto/fisiologia , Éxons/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Isomerismo , Megacariócitos/citologia , Megacariócitos/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pinças Ópticas , Fosfotransferases (Aceptor do Grupo Álcool)/química , Fosfotransferases (Aceptor do Grupo Álcool)/deficiência , Gravidez , Talina/metabolismo , Trombose/genética
7.
J Biol Chem ; 288(48): 34352-63, 2013 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-24133214

RESUMO

Platelets are immunologically competent cells containing cytokines such as TGF-ß1 that regulate cell-mediated immunity. However, the mechanisms underlying cytokine secretion from platelets are undefined. The Wiskott-Aldrich syndrome protein (WASp) regulates actin polymerization in nucleated hematopoietic cells but has other role(s) in platelets. WASp-null (WASp(-/-)) platelets stimulated with a PAR-4 receptor agonist had increased TGF-ß1 release compared with WT platelets; inhibiting WASp function with wiskostatin augmented TRAP-induced TGF-ß1 release in human platelets. TGF-ß1 release is dissociated from α-granule secretion (P-selectin up-regulation) and occurs more gradually, with ∼10-15% released after 30-60 min. Blockade of Src family kinase-mediated WASp Tyr-291/Tyr-293 phosphorylation increased TGF-ß1 release, with no additive effect in WASp(-/-) platelets, signifying that phosphorylation is critical for WASp-limited TGF-ß1 secretion. Inhibiting F-actin assembly with cytochalasin D enhanced secretion in WT platelets and further increased TGF-ß1 release in WASp(-/-) platelets, indicating that WASp and actin assembly independently regulate TGF-ß1 release. A permeabilized platelet model was used to test the role of upstream small GTPases in TGF-ß1 release. N17Cdc42, but not Rac1 mutants, increased TGF-ß1 secretion and abrogated WASp phosphorylation. We conclude that WASp function restricts TGF-ß1 secretion in a Cdc42- and Src family kinase-dependent manner and independently of actin assembly.


Assuntos
Plaquetas/metabolismo , Matriz Extracelular/genética , Imunidade Celular/genética , Fator de Crescimento Transformador beta1/metabolismo , Proteína da Síndrome de Wiskott-Aldrich/genética , Actinas/metabolismo , Animais , Proteínas Reguladoras de Apoptose/agonistas , Proteínas Reguladoras de Apoptose/metabolismo , Plaquetas/imunologia , Carbazóis/metabolismo , Humanos , Camundongos , Propanolaminas/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta1/genética , Proteína da Síndrome de Wiskott-Aldrich/metabolismo , Proteína cdc42 de Ligação ao GTP/genética , Proteína cdc42 de Ligação ao GTP/metabolismo , Quinases da Família src/genética , Quinases da Família src/metabolismo
8.
Blood ; 119(5): 1263-73, 2012 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-22101895

RESUMO

When refrigerated platelets are rewarmed, they secrete active sialidases, including the lysosomal sialidase Neu1, and express surface Neu3 that remove sialic acid from platelet von Willebrand factor receptor (VWFR), specifically the GPIbα subunit. The recovery and circulation of refrigerated platelets is greatly improved by storage in the presence of inhibitors of sialidases. Desialylated VWFR is also a target for metalloproteinases (MPs), because GPIbα and GPV are cleaved from the surface of refrigerated platelets. Receptor shedding is inhibited by the MP inhibitor GM6001 and does not occur in Adam17(ΔZn/ΔZn) platelets expressing inactive ADAM17. Critically, desialylation in the absence of MP-mediated receptor shedding is sufficient to cause the rapid clearance of platelets from circulation. Desialylation of platelet VWFR therefore triggers platelet clearance and primes GPIbα and GPV for MP-dependent cleavage.


Assuntos
Plaquetas/fisiologia , Glicoproteínas de Membrana/metabolismo , Metaloproteases/metabolismo , Neuraminidase/metabolismo , Refrigeração , Proteínas ADAM/metabolismo , Proteínas ADAM/fisiologia , Proteína ADAM17 , Animais , Plaquetas/metabolismo , Preservação de Sangue/métodos , Ativação Enzimática , Glicosilação , Humanos , Masculino , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/fisiologia , Metaloproteases/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Neuraminidase/fisiologia , Complexo Glicoproteico GPIb-IX de Plaquetas , Processamento de Proteína Pós-Traducional/fisiologia , Proteólise , Refrigeração/métodos , Fator de von Willebrand/metabolismo
9.
Blood ; 119(26): 6335-43, 2012 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-22596262

RESUMO

Transfusion-related acute lung injury (TRALI) is the leading cause of transfusion-related death. The biologic processes contributing to TRALI are poorly understood. All blood products can cause TRALI, and no specific treatment is available. A "2-event model" has been proposed as the trigger. The first event may include surgery, trauma, or infection; the second involves the transfusion of antileukocyte antibodies or bioactive lipids within the blood product. Together, these events induce neutrophil activation in the lungs, causing endothelial damage and capillary leakage. Neutrophils, in response to pathogens or under stress, can release their chromatin coated with granule contents, thus forming neutrophil extracellular traps (NETs). Although protective against infection, these NETs are injurious to tissue. Here we show that NET biomarkers are present in TRALI patients' blood and that NETs are produced in vitro by primed human neutrophils when challenged with anti-HNA-3a antibodies previously implicated in TRALI. NETs are found in alveoli of mice experiencing antibody-mediated TRALI. DNase 1 inhalation prevents their alveolar accumulation and improves arterial oxygen saturation even when administered 90 minutes after TRALI onset. We suggest that NETs form in the lungs during TRALI, contribute to the disease process, and thus could be targeted to prevent or treat TRALI.


Assuntos
Lesão Pulmonar Aguda/etiologia , DNA/imunologia , DNA/metabolismo , Neutrófilos/imunologia , Neutrófilos/metabolismo , Reação Transfusional , Lesão Pulmonar Aguda/imunologia , Animais , Doadores de Sangue , Células Cultivadas , Espaço Extracelular/genética , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Ativação de Neutrófilo/imunologia , Neutrófilos/patologia , Imunologia de Transplantes , Transplante Homólogo/imunologia
10.
Blood ; 120(3): 626-35, 2012 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-22613794

RESUMO

Platelets are megakaryocyte subfragments that participate in hemostatic and host defense reactions and deliver pro- and antiangiogenic factors throughout the vascular system. Although they are anucleated cells that lack a complex secretory apparatus with distinct Golgi/endoplasmic reticulum compartments, past studies have shown that platelets have glycosyltransferase activities. In the present study, we show that members of 3 distinct glycosyltransferase families are found within and on the surface of platelets. Immunocytology and flow cytometry results indicated that megakaryocytes package these Golgi-derived glycosyltransferases into vesicles that are sent via proplatelets to nascent platelets, where they accumulate. These glycosyltransferases are active, and intact platelets glycosylate large exogenous substrates. Furthermore, we show that activation of platelets results in the release of soluble glycosyltransferase activities and that platelets contain sufficient levels of sugar nucleotides for detection of glycosylation of exogenously added substrates. Therefore, the results of the present study show that blood platelets are a rich source of both glycosyltransferases and donor sugar substrates that can be released to function in the extracellular space. This platelet-glycosylation machinery offers a pathway to a simple glycoengineering strategy improving storage of platelets and may serve hitherto unknown biologic functions.


Assuntos
Plaquetas/enzimologia , Glicosiltransferases/metabolismo , Megacariócitos/enzimologia , Família Multigênica/fisiologia , Animais , Galactosiltransferases/genética , Galactosiltransferases/metabolismo , Glicosilação , Complexo de Golgi/enzimologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , N-Acetilgalactosaminiltransferases/genética , N-Acetilgalactosaminiltransferases/metabolismo , Ácido N-Acetilneuramínico/metabolismo , Plasma Rico em Plaquetas/citologia , Sialiltransferases/genética , Sialiltransferases/metabolismo , Especificidade por Substrato/fisiologia , Polipeptídeo N-Acetilgalactosaminiltransferase
11.
Cell Microbiol ; 15(3): 353-367, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23083060

RESUMO

The intracellular pathogen Shigella flexneri forms membrane protrusions to spread from cell to cell. As protrusions form, myosin-X (Myo10) localizes to Shigella. Electron micrographs of immunogold-labelled Shigella-infected HeLa cells reveal that Myo10 concentrates at the bases and along the sides of bacteria within membrane protrusions. Time-lapse video microscopy shows that a full-length Myo10 GFP-construct cycles along the sides of Shigella within the membrane protrusions as these structures progressively lengthen. RNAi knock-down of Myo10 is associated with shorter protrusions with thicker stalks, and causes a >80% decrease in confluent cell plaque formation. Myo10 also concentrates in membrane protrusions formed by another intracellular bacteria, Listeria, and knock-down of Myo10 also impairs Listeria plaque formation. In Cos7 cells (contain low concentrations of Myo10), the expression of full-length Myo10 nearly doubles Shigella-induced protrusion length, and lengthening requires the head domain, as well as the tail-PH domain, but not the FERM domain. The GFP-Myo10-HMM domain localizes to the sides of Shigella within membrane protrusions and the GFP-Myo10-PH domain localizes to host cell membranes. We conclude thatMyo10 generates the force to enhance bacterial-induced protrusions by binding its head region to actin filaments and its PH tail domain to the peripheral membrane.


Assuntos
Interações Hospedeiro-Patógeno , Miosinas/metabolismo , Shigella flexneri/fisiologia , Animais , Células COS , Membrana Celular/metabolismo , Membrana Celular/microbiologia , Chlorocebus aethiops , Células HeLa , Humanos , Listeria/patogenicidade , Microscopia Imunoeletrônica , Microscopia de Vídeo
12.
Proc Natl Acad Sci U S A ; 108(28): 11464-9, 2011 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-21709252

RESUMO

The intracellular localization and shape of the nucleus plays a central role in cellular and developmental processes. In fibroblasts, nuclear movement and shape are controlled by a specific perinuclear actin network made of contractile actin filament bundles called transmembrane actin-associated nuclear (TAN) lines that form a structure called the actin cap. The identification of regulatory proteins associated with this specific actin cytoskeletal dynamic is a priority for understanding actin-based changes in nuclear shape and position in normal and pathological situations. Here, we first identify a unique family of actin regulators, the refilin proteins (RefilinA and RefilinB), that stabilize specifically perinuclear actin filament bundles. We next identify the actin-binding filamin A (FLNA) protein as the downstream effector of refilins. Refilins act as molecular switches to convert FLNA from an actin branching protein into one that bundles. In NIH 3T3 fibroblasts, the RefilinB/FLNA complex organizes the perinuclear actin filament bundles forming the actin cap. Finally, we demonstrate that in epithelial normal murine mammary gland (NmuMG) cells, the RefilinB/FLNA complex controls formation of a new perinuclear actin network that accompanies nuclear shape changes during the epithelial-mesenchymal transition (EMT). Our studies open perspectives for further functional analyses of this unique actin-based network and shed light on FLNA function during development and in human syndromes associated with FLNA mutations.


Assuntos
Actinas/metabolismo , Proteínas de Transporte/metabolismo , Núcleo Celular/metabolismo , Proteínas Contráteis/metabolismo , Proteínas dos Microfilamentos/metabolismo , Sequência de Aminoácidos , Animais , Astrocitoma/metabolismo , Astrocitoma/ultraestrutura , Sequência de Bases , Proteínas de Transporte/química , Proteínas de Transporte/genética , Linhagem Celular , Linhagem Celular Tumoral , Núcleo Celular/ultraestrutura , Dimerização , Transição Epitelial-Mesenquimal , Feminino , Filaminas , Humanos , Glândulas Mamárias Animais/citologia , Glândulas Mamárias Animais/metabolismo , Camundongos , Proteínas dos Microfilamentos/química , Proteínas dos Microfilamentos/genética , Microscopia Eletrônica de Transmissão , Dados de Sequência Molecular , Complexos Multiproteicos , Células NIH 3T3 , Domínios e Motivos de Interação entre Proteínas , RNA Interferente Pequeno/genética , Deleção de Sequência
13.
Blood ; 118(8): 2285-95, 2011 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-21652675

RESUMO

Filamin A (FlnA) is a large cytoplasmic protein that crosslinks actin filaments and anchors membrane receptors and signaling intermediates. FlnA(loxP) PF4-Cre mice that lack FlnA in the megakaryocyte (MK) lineage have a severe macrothrombocytopenia because of accelerated platelet clearance. Macrophage ablation by injection of clodronate-encapsulated liposomes increases blood platelet counts in FlnA(loxP) PF4-Cre mice and reveals the desintegration of FlnA-null platelets into microvesicles, a process that occurs spontaneously during storage. FlnA(loxP) PF4-Cre bone marrows and spleens have a 2.5- to 5-fold increase in MK numbers, indicating increased thrombopoiesis in vivo. Analysis of platelet production in vitro reveals that FlnA-null MKs prematurely convert their cytoplasm into large CD61(+) platelet-sized particles, reminiscent of the large platelets observed in vivo. FlnA stabilizes the platelet von Willebrand factor receptor, as surface expression of von Willebrand factor receptor components is normal on FlnA-null MKs but decreased on FlnA-null platelets. Further, FlnA-null platelets contain multiple GPIbα degradation products and have increased expression of the ADAM17 and MMP9 metalloproteinases. Together, the findings indicate that FlnA-null MKs prematurely release large and fragile platelets that are removed rapidly from the circulation by macrophages.


Assuntos
Plaquetas/citologia , Megacariócitos/citologia , Proteínas do Tecido Nervoso/deficiência , Proteínas ADAM/sangue , Proteína ADAM17 , Animais , Plaquetas/metabolismo , Plaquetas/ultraestrutura , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Tamanho Celular , Feminino , Filaminas , Integrina beta3/sangue , Masculino , Metaloproteinase 9 da Matriz/sangue , Megacariócitos/metabolismo , Megacariócitos/ultraestrutura , Camundongos , Camundongos Knockout , Microscopia Eletrônica de Transmissão , Microtúbulos/ultraestrutura , Proteínas do Tecido Nervoso/sangue , Proteínas do Tecido Nervoso/genética , Contagem de Plaquetas , Complexo Glicoproteico GPIb-IX de Plaquetas/metabolismo , Glicoproteínas da Membrana de Plaquetas/fisiologia , Gravidez , Estabilidade Proteica , Trombocitopenia/sangue , Trombocitopenia/etiologia , Trombopoese/genética , Trombopoese/fisiologia
14.
Blood ; 118(6): 1641-52, 2011 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-21566095

RESUMO

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.


Assuntos
Plaquetas/metabolismo , Citoesqueleto/metabolismo , Células Progenitoras de Megacariócitos/metabolismo , Megacariócitos/metabolismo , Espectrina/metabolismo , Actinas/metabolismo , Animais , Plaquetas/citologia , Plaquetas/ultraestrutura , Western Blotting , Membrana Celular/metabolismo , Membrana Celular/ultraestrutura , Células Cultivadas , Citoesqueleto/ultraestrutura , Células Eritroides/metabolismo , Células Progenitoras de Megacariócitos/citologia , Células Progenitoras de Megacariócitos/ultraestrutura , Megacariócitos/citologia , Megacariócitos/ultraestrutura , Camundongos , Microscopia Eletrônica , Microtúbulos/metabolismo , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Multimerização Proteica , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Espectrina/química , Espectrina/genética
15.
Nat Cell Biol ; 8(8): 803-14, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16862148

RESUMO

FilGAP is a newly recognized filamin A (FLNa)-binding RhoGTPase-activating protein. The GTPase-activating protein (GAP) activity of FilGAP is specific for Rac and FLNa binding targets FilGAP to sites of membrane protrusion, where it antagonizes Rac in vivo. Dominant-negative FilGAP constructs lacking GAP activity or knockdown of endogenous FilGAP by small interference RNA (siRNA) induce spontaneous lamellae formation and stimulate cell spreading on fibronectin. Knockdown of endogenous FilGAP abrogates ROCK-dependent suppression of lamellae. Conversely, forced expression of FilGAP induces numerous blebs around the cell periphery and a ROCK-specific inhibitor suppresses bleb formation. ROCK phosphorylates FilGAP, and this phosphorylation stimulates its RacGAP activity and is a requirement for FilGAP-mediated bleb formation. FilGAP is, therefore, a mediator of the well-established antagonism of Rac by RhoA that suppresses leading edge protrusion and promotes cell retraction to achieve cellular polarity.


Assuntos
Actinas/metabolismo , Proteínas Contráteis/metabolismo , Proteínas Ativadoras de GTPase/metabolismo , Proteínas dos Microfilamentos/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas rac de Ligação ao GTP/metabolismo , Proteínas rho de Ligação ao GTP/fisiologia , Células 3T3 , Amidas/farmacologia , Animais , Western Blotting , Linhagem Celular , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Proteínas Contráteis/genética , Filaminas , Proteínas Ativadoras de GTPase/genética , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Proteínas dos Microfilamentos/genética , Microscopia de Fluorescência , Modelos Biológicos , Mutação/genética , Fosforilação , Ligação Proteica , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/genética , Piridinas/farmacologia , Interferência de RNA , Spodoptera , Transfecção , Quinases Associadas a rho
16.
Proc Natl Acad Sci U S A ; 107(36): 15880-5, 2010 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-20798043

RESUMO

Neutrophil extracellular traps (NETs) are part of the innate immune response to infections. NETs are a meshwork of DNA fibers comprising histones and antimicrobial proteins. Microbes are immobilized in NETs and encounter a locally high and lethal concentration of effector proteins. Recent studies show that NETs are formed inside the vasculature in infections and noninfectious diseases. Here we report that NETs provide a heretofore unrecognized scaffold and stimulus for thrombus formation. NETs perfused with blood caused platelet adhesion, activation, and aggregation. DNase or the anticoagulant heparin dismantled the NET scaffold and prevented thrombus formation. Stimulation of platelets with purified histones was sufficient for aggregation. NETs recruited red blood cells, promoted fibrin deposition, and induced a red thrombus, such as that found in veins. Markers of extracellular DNA traps were detected in a thrombus and plasma of baboons subjected to deep vein thrombosis, an example of inflammation-enhanced thrombosis. Our observations indicate that NETs are a previously unrecognized link between inflammation and thrombosis and may further explain the epidemiological association of infection with thrombosis.


Assuntos
DNA/isolamento & purificação , Trombose/etiologia , Animais , Humanos
17.
Dev Cell ; 13(6): 828-42, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18061565

RESUMO

Organization of actin filaments into a well-organized sarcomere structure is critical for muscle development and function. However, it is not completely understood how sarcomeric actin/thin filaments attain their stereotyped lengths. In an RNAi screen in Drosophila primary muscle cells, we identified a gene, sarcomere length short (sals), which encodes an actin-binding, WH2 domain-containing protein, required for proper sarcomere size. When sals is knocked down by RNAi, primary muscles display thin myofibrils with shortened sarcomeres and increased sarcomere number. Both loss- and gain-of-function analyses indicate that SALS may influence sarcomere lengths by promoting thin-filament lengthening from pointed ends. Furthermore, the complex localization of SALS and other sarcomeric proteins in myofibrils reveals that the full length of thin filaments is achieved in a two-step process, and that SALS is required for the second elongation phase, most likely because it antagonizes the pointed-end capping protein Tropomodulin.


Assuntos
Citoesqueleto de Actina/fisiologia , Proteínas de Drosophila/metabolismo , Drosophila/crescimento & desenvolvimento , Proteínas dos Microfilamentos/metabolismo , Músculo Estriado/fisiologia , Miofibrilas/fisiologia , Sarcômeros/fisiologia , Sequência de Aminoácidos , Animais , Animais Geneticamente Modificados , Células Cultivadas , Drosophila/metabolismo , Proteínas de Drosophila/antagonistas & inibidores , Proteínas de Drosophila/genética , Imunofluorescência , Proteínas dos Microfilamentos/antagonistas & inibidores , Proteínas dos Microfilamentos/genética , Dados de Sequência Molecular , Fenótipo , RNA Interferente Pequeno/farmacologia , Homologia de Sequência de Aminoácidos
18.
Blood ; 116(10): 1767-75, 2010 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-20530287

RESUMO

The cellular and molecular mechanisms orchestrating the complex process by which bone marrow megakaryocytes form and release platelets remain poorly understood. Mature megakaryocytes generate long cytoplasmic extensions, proplatelets, which have the capacity to generate platelets. Although microtubules are the main structural component of proplatelets and microtubule sliding is known to drive proplatelet elongation, the role of actin dynamics in the process of platelet formation has remained elusive. Here, we tailored a mouse model lacking all ADF/n-cofilin-mediated actin dynamics in megakaryocytes to specifically elucidate the role of actin filament turnover in platelet formation. We demonstrate, for the first time, that in vivo actin filament turnover plays a critical role in the late stages of platelet formation from megakaryocytes and the proper sizing of platelets in the periphery. Our results provide the genetic proof that platelet production from megakaryocytes strictly requires dynamic changes in the actin cytoskeleton.


Assuntos
Actinas/metabolismo , Plaquetas/metabolismo , Cofilina 1/metabolismo , Destrina/metabolismo , Citoesqueleto de Actina/efeitos dos fármacos , Citoesqueleto de Actina/metabolismo , Animais , Plaquetas/citologia , Plaquetas/ultraestrutura , Western Blotting , Forma Celular , Tamanho Celular , Sobrevivência Celular , Cofilina 1/genética , Citoesqueleto/efeitos dos fármacos , Citoesqueleto/metabolismo , Destrina/genética , Fibrinogênio/metabolismo , Megacariócitos/citologia , Megacariócitos/metabolismo , Megacariócitos/ultraestrutura , Camundongos , Camundongos Knockout , Microscopia Eletrônica de Varredura , Microscopia Eletrônica de Transmissão , Contagem de Plaquetas , Esplenomegalia/genética , Esplenomegalia/metabolismo , Esplenomegalia/patologia , Trombina/farmacologia , Fatores de Tempo
19.
Blood ; 115(14): 2947-55, 2010 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-20154219

RESUMO

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


Assuntos
Plaquetas/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Lectinas Tipo C/metabolismo , Glicoproteínas de Membrana/metabolismo , Ativação Plaquetária/fisiologia , Multimerização Proteica/fisiologia , Proteínas Tirosina Quinases/metabolismo , Transdução de Sinais/fisiologia , Motivos de Aminoácidos , Ativação Enzimática/fisiologia , Humanos , Fosfolipase C gama/metabolismo , Quinase Syk , Domínios de Homologia de src
20.
Am J Hematol ; 87(2): 161-6, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22228373

RESUMO

Protease-activated receptors (PAR)-1 and -4 are the principal receptors for thrombin-mediated platelet activation. Functional genetic variation has been described in the human PAR1 gene, but not in the PAR4 gene (F2RL3). We sought to identify variants in and around F2RL3 and to determine their association with perioperative myocardial injury (PMI) after coronary artery bypass graft surgery. We further explored possible mechanisms for F2RL3 single nucleotide polymorphism (SNP) associations with PMI including altered receptor expression and platelet activation. Twenty-three SNPs in the F2RL3 gene region were genotyped in two phases in 934 Caucasian subjects. Platelets from 43 subjects (23 major allele, 20 risk allele) homozygous for rs773857 (SNP with the strongest association with PMI) underwent flow cytometry to assess PAR4 receptor number and response to activation by a specific PAR4 activating peptide (AYPGKF) measured by von Willebrand factor (vWf) binding and P-selectin release and PAC-1 binding. We identified a novel association of SNP rs773857 with PMI (OR = 2.4, P = 0.004). rs773857 risk allele homozygotes have significantly increased platelet counts and platelets showed a significant increase in P-selectin release after activation (P = 0.004). We conclude that rs773857 risk allele homozygotes are associated with risk for increased platelet count and hyperactivity.


Assuntos
Plaquetas/metabolismo , Ponte de Artéria Coronária/efeitos adversos , Doença da Artéria Coronariana/cirurgia , Traumatismos Cardíacos/sangue , Traumatismos Cardíacos/genética , Polimorfismo de Nucleotídeo Único , Receptores de Trombina/genética , Idoso , Plaquetas/efeitos dos fármacos , Plaquetas/patologia , Doença da Artéria Coronariana/sangue , Doença da Artéria Coronariana/patologia , Fosfatase 2 de Especificidade Dupla/metabolismo , Feminino , Expressão Gênica , Traumatismos Cardíacos/etiologia , Homozigoto , Humanos , Pessoa de Meia-Idade , Oligopeptídeos/metabolismo , Oligopeptídeos/farmacologia , Selectina-P/metabolismo , Período Perioperatório , Ativação Plaquetária/efeitos dos fármacos , Contagem de Plaquetas , Ligação Proteica , Fatores de Risco , Fator de von Willebrand/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA