Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Life (Basel) ; 11(8)2021 Aug 22.
Article in English | MEDLINE | ID: mdl-34440603

ABSTRACT

Murine cytomegalovirus (MCMV) initiates the stepwise establishment of the pre-assembly compartment (pre-AC) in the early phase of infection by the expansion of the early endosome (EE)/endosomal recycling compartment (ERC) interface and relocation of the Golgi complex. We depleted Vps34-derived phosphatidylinositol-3-phosphate (PI(3)P) at EEs by VPS34-IN1 and inhibited PI(3)P-associated functions by overexpression of 2xFYVE- and p40PX PI(3)P-binding modules to assess the role of PI(3)P-dependent EE domains in the pre-AC biogenesis. We monitored the accumulation of Rab10 and Evectin-2 in the inner pre-AC and the relocation of GM130-positive cis-Golgi organelles to the outer pre-AC by confocal microscopy. Although PI(3)P- and Vps34-positive endosomes build a substantial part of pre-AC, the PI(3)P depletion and the inhibition of PI(3)P-associated functions did not prevent the establishment of infection and progression through the early phase. The PI(3)P depletion in uninfected and MCMV-infected cells rapidly dispersed PI(3)P-bond proteins and reorganized EEs, including ablation of EE-to-ERC transport and relocation of Rab11 endosomes. The PI(3)P depletion one hour before pre-AC initiation and overexpression of 2xFYVE and p40PX domains neither prevented Rab10- and Evectin-2 accumulation, nor Golgi unlinking and relocation. These data demonstrate that PI(3)P-dependent functions, including the Rab11-dependent EE-to-ERC route, are dispensable for pre-AC initiation. Nevertheless, the virus growth was drastically reduced in PI(3)P-depleted cells, indicating that PI(3)P-associated functions are essential for the late phase of infection.

2.
Int J Mol Sci ; 22(9)2021 Apr 30.
Article in English | MEDLINE | ID: mdl-33946341

ABSTRACT

Platelets are components of the blood that are highly reactive, and they quickly respond to multiple physiological and pathophysiological processes. In the last decade, it became clear that platelets are the key components of circulation, linking hemostasis, innate, and acquired immunity. Protein composition, localization, and activity are crucial for platelet function and regulation. The current state of mass spectrometry-based proteomics has tremendous potential to identify and quantify thousands of proteins from a minimal amount of material, unravel multiple post-translational modifications, and monitor platelet activity during drug treatments. This review focuses on the role of proteomics in understanding the molecular basics of the classical and newly emerging functions of platelets. including the recently described role of platelets in immunology and the development of COVID-19.The state-of-the-art proteomic technologies and their application in studying platelet biogenesis, signaling, and storage are described, and the potential of newly appeared trapped ion mobility spectrometry (TIMS) is highlighted. Additionally, implementing proteomic methods in platelet transfusion medicine, and as a diagnostic and prognostic tool, is discussed.


Subject(s)
Blood Platelets/metabolism , Mass Spectrometry/methods , Platelet Function Tests/methods , Proteomics/methods , Animals , Blood Platelets/cytology , Blood Platelets/immunology , COVID-19/immunology , COVID-19/metabolism , Humans , Platelet Transfusion , Protein Processing, Post-Translational , Signal Transduction , Transfusion Medicine/methods
3.
Neonatology ; 117(1): 15-23, 2020.
Article in English | MEDLINE | ID: mdl-31786577

ABSTRACT

BACKGROUND: The mechanisms underlying neonatal platelets hyporesponsiveness are not fully understood. While previous studies have demonstrated developmental impairment of agonist-induced platelet activation, differences in inhibitory signaling pathways have been scarcely investigated. OBJECTIVE: To compare neonatal and adult platelets with regard to inhibition of platelet reactivity by prostaglandin E1 (PGE1). METHODS: Platelet-rich plasma from umbilical cord (CB) or adult blood was incubated with PGE1 (0-1 µM). We assessed aggregation in response to adenosine diphosphate (ADP), collagen, and thrombin receptor activating peptide as well as cyclic adenosine 3'5'-monophosphate (cAMP) levels (ELISA). Gαs, Gαi2, and total- and phospho-protein kinase A (PKA) were evaluated in adult and CB ultrapure and washed platelets, respectively, by immunoblotting. RESULTS: Neonatal (vs. adult) platelets display hypersensitivity to inhibition by PGE1 of platelet aggregation induced by ADP and collagen (PGE1 IC50: 14 and 117 nM for ADP and collagen, respectively, vs. 149 and 491 nM in adults). They also show increased basal and PGE1-induced cAMP levels. Mechanistically, PGE1 acts by binding to the prostanoid receptor IP (prostacyclin receptor), which couples to the Gαs protein-adenylate cyclase axis and increases intracellular levels of cAMP. cAMP activates PKA, which phosphorylates different target inhibitor proteins. Neonatal platelets showed higher basal and PGE1-induced cAMP levels, higher Gαs protein expression, and a trend to increased PKA-dependent protein phosphorylation compared to adult platelets. CONCLUSION: Neonatal platelets have a functionally increased PGE1-cAMP-PKA axis. This finding supports a downregulation of inhibitory when going from neonate to adult contributing to neonatal platelet hyporesponsiveness.


Subject(s)
Age Factors , Alprostadil/pharmacology , Platelet Aggregation/drug effects , Adenosine Diphosphate/physiology , Adenylyl Cyclases/blood , Adult , Blood Platelets/drug effects , Blood Platelets/enzymology , Cyclic AMP/blood , Cyclic AMP-Dependent Protein Kinases/physiology , Humans , Infant, Newborn
4.
Blood ; 126(1): 80-8, 2015 Jul 02.
Article in English | MEDLINE | ID: mdl-25838348

ABSTRACT

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.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Blood Platelets , Cell Membrane/ultrastructure , Filamins/metabolism , Megakaryocytes , Adaptor Proteins, Signal Transducing/chemistry , Animals , Blood Platelets/metabolism , Blood Platelets/ultrastructure , Cell Membrane/metabolism , Cells, Cultured , Filamins/physiology , HEK293 Cells , Humans , Megakaryocytes/metabolism , Megakaryocytes/ultrastructure , Mice , Mice, Inbred C57BL , Mice, Transgenic , Protein Binding/physiology , Protein Interaction Domains and Motifs/physiology , Pseudopodia/metabolism
5.
Nat Med ; 21(1): 47-54, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25485912

ABSTRACT

The hepatic Ashwell-Morell receptor (AMR) can bind and remove desialylated platelets. Here we demonstrate that platelets become desialylated as they circulate and age in blood. Binding of desialylated platelets to the AMR induces hepatic expression of thrombopoietin (TPO) mRNA and protein, thereby regulating platelet production. Endocytic AMR controls TPO expression through Janus kinase 2 (JAK2) and the acute phase response signal transducer and activator of transcription 3 (STAT3) in vivo and in vitro. Recognition of this newly identified physiological feedback mechanism illuminates the pathophysiology of platelet diseases, such as essential thrombocythemia and immune thrombocytopenia, and contributes to an understanding of the mechanisms of thrombocytopenia observed with JAK1/2 inhibition.


Subject(s)
Asialoglycoprotein Receptor/metabolism , Blood Platelets/metabolism , Janus Kinase 2/metabolism , STAT3 Transcription Factor/metabolism , Thrombopoietin/metabolism , Animals , Asialoglycoprotein Receptor/genetics , Blood Platelets/pathology , Feedback, Physiological , Humans , Janus Kinase 2/genetics , Liver/metabolism , Mice , Purpura, Thrombocytopenic, Idiopathic/genetics , Purpura, Thrombocytopenic, Idiopathic/pathology , STAT3 Transcription Factor/genetics , Signal Transduction , Thrombocythemia, Essential/genetics , Thrombocythemia, Essential/pathology , Thrombopoietin/genetics
6.
Exp Hematol ; 41(1): 91-101.e4, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22981933

ABSTRACT

The cyclic nucleotides cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP) regulate the activity of protein kinase A (PKA) and protein kinase G (PKG), respectively. This process helps maintain circulating platelets in a resting state. Here we studied the role of cAMP and cGMP in the regulation of megakaryocyte (MK) differentiation and platelet formation. Cultured, platelet-producing MKs were differentiated from fetal livers harvested from 13.5 days postcoital mouse embryos. MK development was accompanied by a dramatic increase in cAMP production and expression of soluble guanylate cyclase, PKG, and PKA as well as their downstream targets vasodilator-stimulated phosphoprotein (VASP) and MENA. Stimulation of prostaglandin E(1) receptor/adenylyl cyclase or soluble guanylate cyclase/PKG in cultured MKs increased VASP phosphorylation, indicating that these components share a common signaling pathway. To dissect out the role of cyclic nucleotides in MK differentiation, cAMP/PKA and cGMP/PKG signaling were alternately blocked in cultured MKs. Down-regulation of cAMP pathway effectors decreased MK numbers and ploidy. Notably, cGMP levels increased at the beginning of MK development and returned to basal levels in parallel with MK maturation. However, inhibition of cGMP pathway effectors had no effect on MK development. In addition, platelet release from mature MKs was enhanced by cGMP and inhibited by cAMP. Our data suggest that cAMP plays an important role in MK differentiation, while cAMP and cGMP have opposite effects on platelet production. Identifying the signaling pathways that underpin MK development and proplatelet formation will provide greater insights into thrombopoiesis and may potentially yield useful therapeutic targets.


Subject(s)
Blood Platelets/physiology , Cyclic AMP/physiology , Cyclic GMP/physiology , Megakaryocytes/physiology , Animals , Cell Adhesion Molecules/metabolism , Cell Differentiation , Cyclic AMP-Dependent Protein Kinases/analysis , Cytoskeletal Proteins/analysis , Female , Mice , Mice, Inbred C57BL , Microfilament Proteins/metabolism , Phosphoproteins/metabolism , Phosphorylation , Pregnancy , Thrombopoietin/physiology
7.
Nat Commun ; 3: 852, 2012 May 22.
Article in English | MEDLINE | ID: mdl-22617292

ABSTRACT

Megakaryocytes release large preplatelet intermediates into the sinusoidal blood vessels. Preplatelets convert into barbell-shaped proplatelets in vitro to undergo repeated abscissions that yield circulating platelets. These observations predict the presence of circular-preplatelets and barbell-proplatelets in blood, and two fundamental questions in platelet biology are what are the forces that determine barbell-proplatelet formation, and how is the final platelet size established. Here we provide insights into the terminal mechanisms of platelet production. We quantify circular-preplatelets and barbell-proplatelets in human blood in high-resolution fluorescence images, using a laser scanning cytometry assay. We demonstrate that force constraints resulting from cortical microtubule band diameter and thickness determine barbell-proplatelet formation. Finally, we provide a mathematical model for the preplatelet to barbell conversion. We conclude that platelet size is limited by microtubule bundling, elastic bending, and actin-myosin-spectrin cortex forces.


Subject(s)
Microtubules/metabolism , Actins/metabolism , Humans , Microscopy, Fluorescence , Models, Theoretical , Myosins/metabolism , Thrombopoiesis/physiology
8.
J Exp Med ; 207(9): 1967-79, 2010 Aug 30.
Article in English | MEDLINE | ID: mdl-20713593

ABSTRACT

Filamin A (FlnA) cross-links actin filaments and connects the Von Willebrand factor receptor GPIb-IX-V to the underlying cytoskeleton in platelets. Because FlnA deficiency is embryonic lethal, mice lacking FlnA in platelets were generated by breeding FlnA(loxP/loxP) females with GATA1-Cre males. FlnA(loxP/y) GATA1-Cre males have a macrothrombocytopenia and increased tail bleeding times. FlnA-null platelets have decreased expression and altered surface distribution of GPIbalpha because they lack the normal cytoskeletal linkage of GPIbalpha to underlying actin filaments. This results in approximately 70% less platelet coverage on collagen-coated surfaces at shear rates of 1,500/s, compared with wild-type platelets. Unexpectedly, however, immunoreceptor tyrosine-based activation motif (ITAM)- and ITAM-like-mediated signals are severely compromised in FlnA-null platelets. FlnA-null platelets fail to spread and have decreased alpha-granule secretion, integrin alphaIIbbeta3 activation, and protein tyrosine phosphorylation, particularly that of the protein tyrosine kinase Syk and phospholipase C-gamma2, in response to stimulation through the collagen receptor GPVI and the C-type lectin-like receptor 2. This signaling defect was traced to the loss of a novel FlnA-Syk interaction, as Syk binds to FlnA at immunoglobulin-like repeat 5. Our findings reveal that the interaction between FlnA and Syk regulates ITAM- and ITAM-like-containing receptor signaling and platelet function.


Subject(s)
Blood Platelets/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Nerve Tissue Proteins/metabolism , Protein-Tyrosine Kinases/metabolism , Signal Transduction , Amino Acid Motifs , Animals , Blood Platelets/cytology , Cell Adhesion , Female , Filamins , Male , Mice , Mice, Knockout , Nerve Tissue Proteins/deficiency , Phosphotyrosine/metabolism , Protein Binding , Syk Kinase , Thrombocytosis/genetics , Thrombocytosis/metabolism
9.
Blood ; 109(2): 616-8, 2007 Jan 15.
Article in English | MEDLINE | ID: mdl-16990590

ABSTRACT

p38 MAP kinase in human platelets is activated by platelet agonists including thrombin, thromboxane A2 (TxA2), ADP, and others. However, both upstream mechanisms of p38 MAP kinase activation, and their downstream sequelae, are presently controversial and essentially unclear. Certain studies report sequential activation of cGMP-dependent protein kinase (PKG) and p38/ERK pathways by platelet agonists, leading to integrin activation and secretion, whereas others establish an essential role of Src/ERK-mediated TxA2 generation for fibrinogen receptor activation in human platelets. Here, we show that ADP secreted from platelet-dense granules, and subsequent activation of P2Y12 receptors, as well as TxA2 release are important upstream mediators of p38 MAP kinase activation by thrombin. However, p38 MAP kinase activation did not significantly contribute to calcium mobilization, P-selectin expression, alphaIIbbeta3 integrin activation, and aggregation of human platelets in response to thrombin. Finally, PKG activation did not stimulate, but rather inhibited, p38 MAP kinase in human platelets.


Subject(s)
Adenosine Diphosphate/metabolism , Blood Platelets/physiology , Cyclic GMP-Dependent Protein Kinases/physiology , Thrombin/physiology , Thromboxane A2/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Blood Platelets/drug effects , Calcium/metabolism , Enzyme Activation/drug effects , Humans , Thrombin/pharmacology , p38 Mitogen-Activated Protein Kinases/drug effects
10.
Blood ; 106(8): 2757-60, 2005 Oct 15.
Article in English | MEDLINE | ID: mdl-15976180

ABSTRACT

Platelets play a crucial role in the physiology of primary hemostasis and pathophysiologic processes such as arterial thrombosis. Accumulating evidence suggests a role of reactive oxygen species (ROSs) in platelet activation. Here we show that platelets activated with different agonists produced intracellular ROSs, which were reduced by reduced nicotinamide adenine dinucleotide (phosphate) (NAD(P)H) oxidase inhibitors and superoxide scavengers. In addition, we demonstrate that ROSs produced in platelets significantly affected alphaIIbbeta3 integrin activation but not alpha and dense granule secretion and platelet shape change. Thrombin-induced integrin alphaIIbbeta3 activation was significantly decreased after pretreatment of platelets with NAD(P)H oxidase inhibitors (diphenylene iodonium [DPI] [45% +/- 9%] and apocynin [43% +/- 11%]) and superoxide scavengers (tiron [60% +/- 9%] and Mn(III)tetrakis (1-methyl-4-pyridyl)porphyrin [MnTMPyP] [70% +/- 6%]). These inhibitors also reduced platelet aggregation and thrombus formation on collagen under high shear and achieved their effects independent of the nitric oxide/cyclic guanosine monophosphate (NO/cGMP) pathway.


Subject(s)
Blood Platelets/metabolism , Cyclic GMP/metabolism , NADPH Oxidases/metabolism , Nitric Oxide/metabolism , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction , Blood Platelets/drug effects , Blood Platelets/enzymology , Enzyme Inhibitors/pharmacology , Humans , NADPH Oxidases/antagonists & inhibitors , Superoxides/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...