Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
PLoS One ; 18(4): e0284402, 2023.
Article in English | MEDLINE | ID: mdl-37104252

ABSTRACT

Membranes contain lipids that are composed of fatty acids (FA) and a polar head. Membrane homeostasis is crucial for optimal bacterial growth and interaction with the environment. Bacteria synthesize their FAs via the FASII pathway. Gram-positive bacteria can incorporate exogenous FAs which need to be phosphorylated to become substrate of the lipid biosynthetic pathway. In many species including staphylococci, streptococci and enterococci, this phosphorylation is carried out by the Fak complex, which is composed of two subunits, FakA and FakB. FakA is the kinase. FakB proteins are members of the DegV family, proteins known to bind FAs. Two or three FakB types have been identified depending on the bacterial species and characterized by their affinity for saturated and/or unsaturated FAs. Some species such as Streptococcus pyogenes, which causes a wide variety of diseases ranging from mild non-invasive to severe invasive infections, possess an uncharacterized additional DegV protein. We identify here this DegV member as a fourth FakB protein, named FakB4. The fakB4 gene is co-regulated with FASII genes suggesting an interaction with endogenous fatty acids. fakB4 deletion has no impact on membrane phospholipid composition nor on the percentage of other major lipids. However, the fakB4 mutant strain produced more lipids and more extracellular membrane vesicles than the wild-type strain. This suggests that FakB4 is involved in endogenous FA binding and controls FA storage or catabolism resulting in a limitation of extracellular FA release via membrane vesicles.


Subject(s)
Membrane Lipids , Streptococcus pyogenes , Membrane Lipids/metabolism , Streptococcus pyogenes/metabolism , Bacterial Proteins/metabolism , Fatty Acids/metabolism , Phospholipids/metabolism
2.
Cell Mol Life Sci ; 79(7): 365, 2022 Jun 16.
Article in English | MEDLINE | ID: mdl-35708858

ABSTRACT

SARS-CoV-2, although not being a circulatory virus, spread from the respiratory tract resulting in multiorgan failures and thrombotic complications, the hallmarks of fatal COVID-19. A convergent contributor could be platelets that beyond hemostatic functions can carry infectious viruses. Here, we profiled 52 patients with severe COVID-19 and demonstrated that circulating platelets of 19 out 20 non-survivor patients contain SARS-CoV-2 in robust correlation with fatal outcome. Platelets containing SARS-CoV-2 might originate from bone marrow and lung megakaryocytes (MKs), the platelet precursors, which were found infected by SARS-CoV-2 in COVID-19 autopsies. Accordingly, MKs undergoing shortened differentiation and expressing anti-viral IFITM1 and IFITM3 RNA as a sign of viral sensing were enriched in the circulation of deadly COVID-19. Infected MKs reach the lung concomitant with a specific MK-related cytokine storm rich in VEGF, PDGF and inflammatory molecules, anticipating fatal outcome. Lung macrophages capture SARS-CoV-2-containing platelets in vivo. The virus contained by platelets is infectious as capture of platelets carrying SARS-CoV-2 propagates infection to macrophages in vitro, in a process blocked by an anti-GPIIbIIIa drug. Altogether, platelets containing infectious SARS-CoV-2  alter COVID-19 pathogenesis and provide a powerful fatality marker. Clinical targeting of platelets might prevent viral spread, thrombus formation and exacerbated inflammation at once and increase survival in COVID-19.


Subject(s)
COVID-19 , Thrombosis , Blood Platelets , Humans , Lung , Megakaryocytes , Membrane Proteins , RNA-Binding Proteins , SARS-CoV-2
3.
Sci Transl Med ; 12(535)2020 03 18.
Article in English | MEDLINE | ID: mdl-32188724

ABSTRACT

In addition to hemostasis, human platelets have several immune functions and interact with infectious pathogens including HIV in vitro. Here, we report that platelets from HIV-infected individuals on combined antiretroviral drug therapy (ART) with low blood CD4+ T cell counts (<350 cells/µl) contained replication-competent HIV despite viral suppression. In vitro, human platelets harboring HIV propagated the virus to macrophages, a process that could be prevented with the biologic abciximab, an anti-integrin αIIb/ß3 Fab. Furthermore, in our cohort, 88% of HIV-infected individuals on ART with viral suppression and with platelets containing HIV were poor immunological responders with CD4+ T cell counts remaining below <350 cells/µl for more than one year. Our study suggests that platelets may be transient carriers of HIV and may provide an alternative pathway for HIV dissemination in HIV-infected individuals on ART with viral suppression and poor CD4+ T cell recovery.


Subject(s)
Blood Platelets , HIV Infections , Anti-Retroviral Agents/therapeutic use , CD4-Positive T-Lymphocytes , HIV Infections/drug therapy , Humans , Macrophages , Viral Load
5.
J Bone Miner Res ; 31(3): 498-513, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26426912

ABSTRACT

Claudin-16 protein (CLDN16) is a component of tight junctions (TJ) with a restrictive distribution so far demonstrated mainly in the kidney. Here, we demonstrate the expression of CLDN16 also in the tooth germ and show that claudin-16 gene (CLDN16) mutations result in amelogenesis imperfecta (AI) in the 5 studied patients with familial hypomagnesemia with hypercalciuria and nephrocalcinosis (FHHNC). To investigate the role of CLDN16 in tooth formation, we studied a murine model of FHHNC and showed that CLDN16 deficiency led to altered secretory ameloblast TJ structure, lowering of extracellular pH in the forming enamel matrix, and abnormal enamel matrix protein processing, resulting in an enamel phenotype closely resembling human AI. This study unravels an association of FHHNC owing to CLDN16 mutations with AI, which is directly related to the loss of function of CLDN16 during amelogenesis. Overall, this study indicates for the first time the importance of a TJ protein in tooth formation and underlines the need to establish a specific dental follow-up for these patients.


Subject(s)
Ameloblasts/metabolism , Claudins/deficiency , Dental Enamel/abnormalities , Dental Enamel/metabolism , Tight Junctions/metabolism , Adult , Ameloblasts/pathology , Amelogenesis Imperfecta/metabolism , Amelogenesis Imperfecta/pathology , Animals , Child , Claudins/genetics , Dental Enamel/pathology , Female , Humans , Hydrogen-Ion Concentration , Male , Mice , Middle Aged , Mutation/genetics , Phenotype , Syndrome , Young Adult
6.
Autophagy ; 11(7): 1130-45, 2015.
Article in English | MEDLINE | ID: mdl-26039325

ABSTRACT

The glomerulus is a highly specialized capillary tuft, which under pressure filters large amounts of water and small solutes into the urinary space, while retaining albumin and large proteins. The glomerular filtration barrier (GFB) is a highly specialized filtration interface between blood and urine that is highly permeable to small and midsized solutes in plasma but relatively impermeable to macromolecules such as albumin. The integrity of the GFB is maintained by molecular interplay between its 3 layers: the glomerular endothelium, the glomerular basement membrane and podocytes, which are highly specialized postmitotic pericytes forming the outer part of the GFB. Abnormalities of glomerular ultrafiltration lead to the loss of proteins in urine and progressive renal insufficiency, underlining the importance of the GFB. Indeed, albuminuria is strongly predictive of the course of chronic nephropathies especially that of diabetic nephropathy (DN), a leading cause of renal insufficiency. We found that high glucose concentrations promote autophagy flux in podocyte cultures and that the abundance of LC3B II in podocytes is high in diabetic mice. Deletion of Atg5 specifically in podocytes resulted in accelerated diabetes-induced podocytopathy with a leaky GFB and glomerulosclerosis. Strikingly, genetic alteration of autophagy on the other side of the GFB involving the endothelial-specific deletion of Atg5 also resulted in capillary rarefaction and accelerated DN. Thus autophagy is a key protective mechanism on both cellular layers of the GFB suggesting autophagy as a promising new therapeutic strategy for DN.


Subject(s)
Autophagy , Diabetic Nephropathies/pathology , Diabetic Nephropathies/prevention & control , Endothelial Cells/pathology , Podocytes/pathology , Animals , Apoptosis/drug effects , Autophagy/drug effects , Autophagy-Related Protein 5 , Cells, Cultured , Diabetic Nephropathies/physiopathology , Endothelial Cells/drug effects , Endothelial Cells/ultrastructure , Gene Deletion , Glomerular Filtration Rate/drug effects , Glucose/pharmacology , Integrases/metabolism , Mesangial Cells/drug effects , Mesangial Cells/pathology , Mesangial Cells/ultrastructure , Mice, Inbred C57BL , Microtubule-Associated Proteins/deficiency , Microtubule-Associated Proteins/metabolism , Phenotype , Podocytes/drug effects , Podocytes/ultrastructure
7.
J Am Soc Nephrol ; 25(5): 1050-62, 2014 May.
Article in English | MEDLINE | ID: mdl-24722437

ABSTRACT

The endothelin system has emerged as a novel target for the treatment of diabetic nephropathy. Endothelin-1 promotes mesangial cell proliferation and sclerosis. However, no direct pathogenic effect of endothelin-1 on podocytes has been shown in vivo and endothelin-1 signaling in podocytes has not been investigated. This study investigated endothelin effects in podocytes during experimental diabetic nephropathy. Stimulation of primary mouse podocytes with endothelin-1 elicited rapid calcium transients mediated by endothelin type A receptors (ETARs) and endothelin type B receptors (ETBRs). We then generated mice with a podocyte-specific double deletion of ETAR and ETBR (NPHS2-Cre×Ednra(lox/lox)×Ednrb(lox/lox) [Pod-ETRKO]). In vitro, treatment with endothelin-1 increased total ß-catenin and phospho-NF-κB expression in wild-type glomeruli, but this effect was attenuated in Pod-ETRKO glomeruli. After streptozotocin injection to induce diabetes, wild-type mice developed mild diabetic nephropathy with microalbuminuria, mesangial matrix expansion, glomerular basement membrane thickening, and podocyte loss, whereas Pod-ETRKO mice presented less albuminuria and were completely protected from glomerulosclerosis and podocyte loss, even when uninephrectomized. Moreover, glomeruli from normal and diabetic Pod-ETRKO mice expressed substantially less total ß-catenin and phospho-NF-κB compared with glomeruli from counterpart wild-type mice. This evidence suggests that endothelin-1 drives development of glomerulosclerosis and podocyte loss through direct activation of endothelin receptors and NF-κB and ß-catenin pathways in podocytes. Notably, both the expression and function of the ETBR subtype were found to be important. Furthermore, these results indicate that activation of the endothelin-1 pathways selectively in podocytes mediates pathophysiologic crosstalk that influences mesangial architecture and sclerosis.


Subject(s)
Diabetic Nephropathies/etiology , Endothelin-1/physiology , Podocytes/metabolism , Podocytes/pathology , Animals , Diabetic Nephropathies/genetics , Diabetic Nephropathies/pathology , Down-Regulation/genetics , Down-Regulation/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Receptor, Endothelin A/genetics , Receptor, Endothelin A/metabolism , Receptor, Endothelin B/genetics , Receptor, Endothelin B/metabolism , Signal Transduction/genetics , Signal Transduction/physiology , beta Catenin/metabolism
8.
Biochim Biophys Acta ; 1830(6): 3719-33, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23500070

ABSTRACT

BACKGROUND: Hereditary optic neuropathies (HONs) are a heterogeneous group of disorders that affect retinal ganglion cells (RGCs) and axons that form the optic nerve. Leber's Hereditary Optic Neuropathy and the autosomal dominant optic atrophy related to OPA1 mutations are the most common forms. Nonsyndromic autosomal recessive optic neuropathies are rare and their existence has been long debated. We recently identified the first gene responsible for these conditions, TMEM126A. This gene is highly expressed in retinal cellular compartments enriched in mitochondria and supposed to encode a mitochondrial transmembrane protein of unknown function. METHODS: A specific polyclonal antibody targeting the TMEM126A protein has been generated. Quantitative fluorescent in situ hybridization, cellular fractionation, mitochondrial membrane association study, mitochondrial sub compartmentalization analysis by both proteolysis assays and transmission electron microscopy, and expression analysis of truncated TMEM126A constructs by immunofluorescence confocal microscopy were carried out. RESULTS: TMEM126A mRNAs are strongly enriched in the vicinity of mitochondria and encode an inner mitochondrial membrane associated cristae protein. Moreover, the second transmembrane domain of TMEM126A is required for its mitochondrial localization. CONCLUSIONS: TMEM126A is a mitochondrial located mRNA (MLR) that may be translated in the mitochondrial surface and the protein is subsequently imported to the inner membrane. These data constitute the first step toward a better understanding of the mechanism of action of TMEM126A in RGCs and support the importance of mitochondrial dysfunction in the pathogenesis of HON. GENERAL SIGNIFICANCE: Local translation of nuclearly encoded mitochondrial mRNAs might be a mechanism for rapid onsite supply of mitochondrial membrane proteins.


Subject(s)
Membrane Proteins/biosynthesis , Mitochondrial Membranes/metabolism , Mitochondrial Proteins/biosynthesis , Protein Biosynthesis , Retinal Ganglion Cells/metabolism , Animals , COS Cells , Chlorocebus aethiops , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/metabolism , Genetic Diseases, Inborn/pathology , Humans , Membrane Proteins/genetics , Mitochondrial Membranes/pathology , Mitochondrial Proteins/genetics , Mutation , Optic Nerve Diseases/genetics , Optic Nerve Diseases/metabolism , Optic Nerve Diseases/pathology , Protein Structure, Tertiary , RNA, Messenger/genetics , RNA, Messenger/metabolism , Retinal Ganglion Cells/pathology
9.
PLoS One ; 7(1): e30130, 2012.
Article in English | MEDLINE | ID: mdl-22272285

ABSTRACT

BACKGROUND: Cell-to-cell virus transmission of Human immunodeficiency virus type-1 (HIV-1) is predominantly mediated by cellular structures such as the virological synapse (VS). The VS formed between an HIV-1-infected T cell and a target T cell shares features with the immunological synapse (IS). We have previously identified the human homologue of the Drosophila Discs Large (Dlg1) protein as a new cellular partner for the HIV-1 Gag protein and a negative regulator of HIV-1 infectivity. Dlg1, a scaffolding protein plays a key role in clustering protein complexes in the plasma membrane at cellular contacts. It is implicated in IS formation and T cell signaling, but its role in HIV-1 cell-to-cell transmission was not studied before. METHODOLOGY/PRINCIPAL FINDINGS: Kinetics of HIV-1 infection in Dlg1-depleted Jurkat T cells show that Dlg1 modulates the replication of HIV-1. Single-cycle infectivity tests show that this modulation does not take place during early steps of the HIV-1 life cycle. Immunofluorescence studies of Dlg1-depleted Jurkat T cells show that while Dlg1 depletion affects IS formation, it does not affect HIV-1-induced VS formation. Co-culture assays and quantitative cell-to-cell HIV-1 transfer analyses show that Dlg1 depletion does not modify transfer of HIV-1 material from infected to target T cells, or HIV-1 transmission leading to productive infection via cell contact. Dlg1 depletion results in increased virus yield and infectivity of the viral particles produced. Particles with increased infectivity present an increase in their cholesterol content and during the first hours of T cell infection these particles induce higher accumulation of total HIV-1 DNA. CONCLUSION: Despite its role in the IS formation, Dlg1 does not affect the VS and cell-to-cell spread of HIV-1, but plays a role in HIV-1 cell-free virus transmission. We propose that the effect of Dlg1 on HIV-1 infectivity is at the stage of virus entry.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Communication , HIV-1/metabolism , Membrane Proteins/metabolism , T-Lymphocytes/metabolism , Adaptor Proteins, Signal Transducing/genetics , Cell Line, Tumor , Cells, Cultured , Cholesterol/metabolism , Coculture Techniques , DNA, Viral/genetics , DNA, Viral/metabolism , Discs Large Homolog 1 Protein , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , HIV-1/genetics , HIV-1/physiology , HeLa Cells , Host-Pathogen Interactions , Humans , Immunological Synapses/metabolism , Immunological Synapses/virology , Jurkat Cells , Kinetics , Membrane Proteins/genetics , Microscopy, Confocal , Microscopy, Electron , RNA Interference , T-Lymphocytes/ultrastructure , T-Lymphocytes/virology , Virus Replication/genetics
10.
Biol Cell ; 103(11): 531-42, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21810080

ABSTRACT

BACKGROUND INFORMATION: The identification of a source of stem cells able to regenerate skeletal muscle was the goal of numerous studies with the aim to develop new therapeutic approaches for genetic muscle diseases or muscle injuries. A series of studies have demonstrated that stem cells derived from various tissues may have a role in the regeneration of damaged muscles, but this contribution is always very weak. Thus we established a project aiming to reprogramme non-muscle cells into the skeletal striated differentiation pathway. RESULTS: We transduced several human primary adult stem or progenitor cells using a recombinant lentivirus containing the coding sequence of the Myf5 gene considered as a master gene for the determination of skeletal striated muscle. These original results are the first demonstration of a myogenic conversion of human mesenchymal and endothelial cells by Myf5. CONCLUSIONS: The procedure described in the present paper could be used to develop new research protocols with the prospect of using these cells as therapeutic agents.


Subject(s)
Cellular Reprogramming , Endothelial Cells/cytology , Endothelial Cells/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Myoblasts/cytology , Myogenic Regulatory Factor 5/metabolism , Cell Differentiation , Cell Proliferation , Cells, Cultured , Epithelial Cells/cytology , Epithelial Cells/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Myoblasts/metabolism , Myogenic Regulatory Factor 5/genetics
11.
Stem Cells ; 25(6): 1571-7, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17379771

ABSTRACT

Megakaryocytic (MK) lineage is an attractive target for cell/gene therapy approaches, aiming at correcting platelet protein deficiencies. However, MK cells are short-lived cells, and their permanent modification requires modification of hematopoietic stem cells with an integrative vector such as a lentiviral vector. Glycoprotein (Gp) IIb promoter, the most studied among the MK regulatory sequences, is also active in stem cells. To strictly limit transgene expression to the MK lineage after transduction of human CD34(+) hematopoietic cells with a lentiviral vector, we looked for a promoter activated later during MK differentiation. Human cord blood, bone marrow, and peripheral-blood mobilized CD34(+) cells were transduced with a human immunodeficiency virus-derived self-inactivating lentiviral vector encoding the green fluorescent protein (GFP) under the transcriptional control of GpIbalpha, GpIIb, or EF1alpha gene regulatory sequences. Both GpIbalpha and GpIIb promoters restricted GFP expression (analyzed by flow cytometry and immunoelectron microscopy) in MK cells among the maturing progeny of transduced cells. However, only the GpIbalpha promoter was strictly MK-specific, whereas GpIIb promoter was leaky in immature progenitor cells not yet engaged in MK cell lineage differentiation. We thus demonstrate the pertinence of using a 328-base-pair fragment of the human GpIbalpha gene regulatory sequence, in the context of a lentiviral vector, to tightly restrict transgene expression to the MK lineage after transduction of human CD34(+) hematopoietic cells. Disclosure of potential conflicts of interest is found at the end of this article.


Subject(s)
Genetic Vectors , Hematopoietic Stem Cells/cytology , Lentivirus , Megakaryocytes/metabolism , Platelet Glycoprotein GPIb-IX Complex/genetics , Promoter Regions, Genetic/physiology , Transduction, Genetic , Animals , Antigens, CD34/metabolism , Cell Differentiation/genetics , Cell Lineage/genetics , Cells, Cultured , Gene Expression Regulation , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/metabolism , Humans , Megakaryocytes/cytology , Mice , Mice, Inbred NOD , Mice, SCID , Organ Specificity , Virus Inactivation
12.
Eur J Haematol ; 77(1): 74-9, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16827885

ABSTRACT

We report the unusual transformation of a case of Waldenström's macroglobulinemia (WM) into IgM multiple myeloma (MM). The initial clinical and biological presentation of the disease was typical smouldering WM, with lymphocytic infiltration of the bone marrow. Five years later, signs of transformation appeared: the patient presented with diffuse osteolytic bone lesions without organomegaly, and the bone marrow was infiltrated with characteristic malignant plasma cells. Electron microscopy (EM) examination showed that the endoplasmic reticulum (ER) of the dysmorphic plasma cells contained monoclonal IgM. Immunolabeling for calreticulin, a resident protein of the ER, demonstrated unequivocally that the characteristic intranuclear inclusions were indeed part of ER. Flow cytometry revealed an MM profile for the cellular proliferation. Molecular biology performed on the final marrow could only retrieve a single cellular clone. In conclusion, this is the first documented description of the transformation of typical WM into an aggressive form of MM.


Subject(s)
Cell Transformation, Neoplastic , Multiple Myeloma/etiology , Waldenstrom Macroglobulinemia/pathology , Bone Marrow/pathology , Disease Progression , Endoplasmic Reticulum/pathology , Female , Humans , Middle Aged , Multiple Myeloma/complications , Osteolysis/etiology , Plasma Cells/pathology , Plasma Cells/ultrastructure
13.
Platelets ; 17(1): 14-9, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16308182

ABSTRACT

We report the case of a 60-year-old woman who was newly diagnosed for the gray platelet syndrome (GPS). This patient had long-term thrombocytopenia which had been initially misdiagnosed as idiopathic thrombocytopenic purpura (ITP). Blood smear displayed characteristic gray platelets, allowing the diagnosis to be made, which was confirmed by electron microscopy (EM). Polymorphonuclear neutrophils (PMN) appeared poorly granulated on the May-Grunwald-Giemsa-stained blood smear. Flow cytometry analysis of PMN demonstrated increased expression of CD35, CD11b and CD18 at resting PMN surface, without any changes after fMLP stimulation. Ultrastructural study retrieved a decreased number of myeloperoxidase (MPO)-negative secondary granules in PMN. Immunolabeling confirmed the presence of membrane proteins and the absence of soluble content in platelet and megakaryocyte (MK) alpha-granules, and the decrease of secondary granules and secretory vesicles in PMN. This new observation demonstrates that the impairment of the secretory compartment of PMN is definitely a hallmark of GPS, and that the detection of these subtle abnormalities should be searched with adequate and up-to-date technical approaches.


Subject(s)
Blood Platelet Disorders/diagnosis , Blood Platelet Disorders/pathology , Blood Platelets/pathology , Neutrophils/metabolism , Blood Platelet Disorders/blood , Blood Platelets/ultrastructure , Female , Humans , Middle Aged , Neutrophils/pathology , Syndrome
14.
Thromb Haemost ; 94(5): 1004-11, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16363244

ABSTRACT

Multimerin 1 (MMRN1) is a large, soluble, polymeric, factor V binding protein and member of the EMILIN protein family. In vivo, MMRN1 is found in platelets, megakaryocytes, endothelium and extracellular matrix fibers, but not in plasma. To address the mechanism of MMRN1 binding to activated platelets and endothelial cells, we investigated the identity of the major MMRN1 receptors on these cells using wild-type and RGE-forms of recombinant MMRN1. Ligand capture, cell adhesion, ELISA and flow cytometry analyses of platelet-MMRN1 binding, indicated that MMRN1 binds to integrins alphaIIbbeta3 and alphavbeta3. Endothelial cell binding to MMRN1 was predominantly mediated by alphavbeta3 and did not require the MMRN1 RGD site or cellular activation. Like many other alphavbeta3 ligands, MMRN1 had the ability to support adhesion of additional cell types, including stimulated neutrophils. Expression studies, using a cell line capable of endothelial-like MMRN1 processing, indicated that MMRN1 adhesion to cellular receptors enhanced its extracellular matrix fiber assembly. These studies implicate integrin-mediated binding in MMRN1 attachment to cells and indicate that MMRN1 is a ligand for alphaIIbbeta3 and alphavbeta3.


Subject(s)
Blood Platelets/metabolism , Blood Proteins/metabolism , Integrin alphaVbeta3/metabolism , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Cell Line , Endothelial Cells/metabolism , Endothelial Cells/ultrastructure , Extracellular Matrix/metabolism , Extracellular Matrix/ultrastructure , Humans , In Vitro Techniques , Kidney/cytology , Ligands , Megakaryocytes/metabolism , Microscopy, Immunoelectron
15.
Thromb Haemost ; 94(3): 585-92, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16268476

ABSTRACT

Factor V is an essential coagulation cofactor that circulates in plasma and platelet alpha-granules where it is stored complexed to multimerin I (MMRN1). To gain insights into the origin and processing of human platelet factor V, and factor V-MMRN I complexes, we studied factorV in cultured megakaryocytes. Factor V mRNA was detected in all megakaryocyte cultures. However, like albumin, IgG and fibrinogen, factorV protein was detectable only in megakaryocytes cultured with exogenous protein. The amount of factor V associated with megakaryocytes was influenced by the exogenous factorV concentration. Similar to platelet factor V, megakaryocyte factor V was proteolyzed and complexed with megakaryocyte-synthesized MMRN1. With secretagogues, megakaryocytes released factor V, IgG, fibrinogen and MMRN1. Immunofluorescent and electron microscopy confirmed factorV uptake by endocytosis and its trafficking to megakaryocyte alpha-granules. These data provide direct evidence that human megakaryocytes process plasma-derived factor V into alpha-granules and generate factorV-MMRN I complexes from endogenously and exogenously synthesized proteins.


Subject(s)
Endocytosis , Factor V/metabolism , Megakaryocytes/metabolism , RNA, Messenger/metabolism , Blood Platelets/metabolism , Blood Proteins/metabolism , Bone Marrow Cells/metabolism , Cells, Cultured , Cytoplasmic Granules/metabolism , Factor V/genetics , Fibrinogen/metabolism , Humans , Immunoglobulin G/metabolism , Plasma/metabolism , Platelet Membrane Glycoprotein IIb/analysis , Time Factors
16.
Platelets ; 16(1): 13-8, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15763891

ABSTRACT

Shear stress encountered in stenosed human arteries is able to induce a certain range of platelet activation. In order to determine the extent of platelet shape change induced by high shear rate conditions, we used electron microscopy (EM) and immuno-EM to study platelet ultrastructure from blood flowing in vivo through stenosed arteries. Then it was compared with platelets from healthy controls exposed in vitro to a shear rate of 4000 s(-1). Six patients with stenosed arteries (iliac, femoral and renal) were investigated at the time of transcutaneous angiography. Blood was harvested from the same catheter in the stenosed artery and in the abdominal aortic artery (control sample), each patient being its own control. The percentage of platelets with shape changes (loss of discoid form, pseudopod emission, organelle centralisation) significantly increased in samples from stenosed arteries. Shape change was concomitant with the membrane glycoprotein IIb-IIIa distribution at the pseudopod extremities. These activated platelets had not completed secretion and were maintained in a reversible activation state. Similar results were obtained on platelets from healthy donors submitted in vitro to a high shear rate. In conclusion, this study shows that the high shear rate encountered in human stenosed arteries is able to induce shape change and reversible activation of platelets in vivo.


Subject(s)
Arteries/pathology , Blood Platelets/pathology , Constriction, Pathologic/blood , Platelet Activation , Platelet Membrane Glycoproteins/metabolism , Aged , Blood Platelets/ultrastructure , Case-Control Studies , Cell Shape , Female , Humans , Male , Microscopy, Electron , Protein Transport , Stress, Mechanical
17.
J Biol Chem ; 280(7): 5843-53, 2005 Feb 18.
Article in English | MEDLINE | ID: mdl-15576378

ABSTRACT

Although leukosialin (CD43) membrane expression decreases during neutrophil apoptosis, the CD43 molecule, unexpectedly, is neither proteolyzed nor internalized. We thus wondered whether it could be shed on bleb-derived membrane vesicles. Membrane blebbing is a transient event, hardly appreciated during the asynchronous, spontaneous apoptosis of neutrophils. Cell pre-synchronization at 15 degrees C made it possible to observe numerous blebbing neutrophils for a short 1-h period at 37 degrees C. CD43 down-regulation co-occurred with the blebbing stage and phosphatidylserine externalization, shortly after mitochondria depolarization and before nuclear condensation. Blebs detaching from the cell body were observed by time lapse fluorescence microscopy, and the release of bleb-derived vesicles was followed by flow cytometry. Phosphatidylserine externalization required caspases and protein kinase C (PKC) but not the myosin light chain kinase (MLCK). By contrast, bleb formation and release was caspase- and PKC-independent but required an active MLCK, whereas CD43 down-regulation involved caspases but neither PKC nor MLCK. Furthermore, CD43 appeared mostly excluded from membrane blebs by electron microscopy. Thus, CD43 down-regulation does not result from the release of bleb-derived vesicles. Ultracentrifugation of apoptotic cell supernatants made it possible to recover <1 microM microparticles, which contained the entire CD43 molecule. These microparticles expressed neutrophil membrane markers such as CD11b, CD66b, and CD63, together with CD43. In conclusion, we show that the three early membrane events of apoptosis, namely blebbing, phosphatidylserine externalization, and CD43 down-regulation, result from different signaling pathways and can occur independently from one another. CD43 down-regulation results from the shedding of microparticles released during apoptosis but unrelated to the blebbing.


Subject(s)
Antigens, CD/metabolism , Apoptosis , Cell Surface Extensions/physiology , Down-Regulation , Neutrophils/cytology , Phospholipids/metabolism , Sialoglycoproteins/metabolism , Signal Transduction , Annexins/metabolism , CD11b Antigen/metabolism , Caspases/metabolism , Cell Membrane/metabolism , Cells, Cultured , Cytoplasmic Vesicles/physiology , Endocytosis , Exocytosis , Flow Cytometry , Leukosialin , Mitochondria/physiology , Myosin-Light-Chain Kinase/metabolism , Neutrophils/metabolism , Phosphatidylserines/metabolism , Protein Kinase C/metabolism , Receptors, Cell Surface/metabolism
18.
Thromb Haemost ; 90(5): 844-52, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14597980

ABSTRACT

alpha-Granule protein storage is important for producing platelets with normal haemostatic function. The low to undetectable levels of several megakaryocyte-synthesized alpha-granule proteins in normal plasma suggest megakaryocytes are important to sequester these proteins in vivo. alpha-Granule protein storage in vitro has been studied using other cell types, with differences observed in how some proteins are processed compared to platelets. Human megakaryocytes, cultured from cord blood CD34(+) cells and grown in serum-free media containing thrombopoietin, were investigated to determine if they could be used as a model for studying normal alpha-granule protein processing and storage. ELISA indicated that cultured megakaryocytes contained the alpha-granule proteins multimerin, von Willebrand factor, thrombospondin-1, beta-thromboglobulin and platelet factor 4, but no detectable fibrinogen and factor V. A significant proportion of the alpha-granule protein in megakaryocyte cultures was contained within the cells (averages: 41-71 %), consistent with storage. Detailed analyses of multimerin and von Willebrand factor confirmed that alpha-granule proteins were processed to mature forms and were predominantly located in the alpha-granules of cultured megakaryocytes.Thrombopoietin-stimulated cultured megakaryocytes provide a useful model for studying alpha-granule protein processing and storage.


Subject(s)
Cytoplasmic Granules/chemistry , Megakaryocytes/chemistry , Proteins/metabolism , Blood Proteins/analysis , Blood Proteins/metabolism , Cells, Cultured , Humans , Megakaryocytes/cytology , Megakaryocytes/ultrastructure , Platelet Factor 4/analysis , Platelet Factor 4/metabolism , Proteins/analysis , Thrombopoietin/pharmacology , Thrombospondin 1/analysis , Thrombospondin 1/metabolism , beta-Thromboglobulin/analysis , beta-Thromboglobulin/metabolism , von Willebrand Factor/analysis , von Willebrand Factor/metabolism
19.
Leuk Lymphoma ; 43(4): 719-24, 2002 Apr.
Article in English | MEDLINE | ID: mdl-12153156

ABSTRACT

The study presented here, performed on the bone marrow from patients with idiopathic myelofibrosis (MF) and on a murine model of MF, demonstrates a pathological interaction between PMN leukocytes and megakaryocyte (Mk), correlated with MF development. The data obtained revealed abnormal subcellular P-selectin distribution, which appeared to correlate with excessive and pathological emperipolesis of PMN leukocytes within Mk, leading to the destruction of Mk storage organelles and leakage of alpha-granular contents into the bone marrow microenvironment. The prominent role of growth factors, PDGF and TGFbeta, stored in the Mk alpha-granular compartment in the generation of MF has been previously largely documented. Both growth factors are essential for the Mk-dependent fibroblast proliferation. The destructive mutual cellular interaction of Mk and PMN leading to the pathological release of PDGF and TGFbeta within the bone marrow microenvironment may participate, through fibroblast activation, to the generation of MF. Therefore, this study provides insight into the possible pathophysiological mechanisms for the genesis of MF.


Subject(s)
Cell Communication , Megakaryocytes/physiology , Neutrophils/physiology , Primary Myelofibrosis/etiology , Animals , Bone Marrow/ultrastructure , Cell Division , Disease Models, Animal , Humans , Megakaryocytes/ultrastructure , Mice , Microscopy, Electron , P-Selectin/analysis , Primary Myelofibrosis/pathology , Thrombopoietin/genetics
20.
Blood ; 99(10): 3637-45, 2002 May 15.
Article in English | MEDLINE | ID: mdl-11986218

ABSTRACT

Vascular endothelial cells are thought to be the main source of plasma tissue-type plasminogen activator (t-PA) and von Willebrand factor (VWF). Previous studies have suggested that both t-PA and VWF are acutely released in response to the same stimuli, both in cultured endothelial cells and in vivo. However, the subcellular storage compartment in endothelial cells has not been definitively established. We tested the hypothesis that t-PA is localized in Weibel-Palade (WP) bodies, the specialized endothelial storage granules for VWF. In cultured human umbilical vein endothelial cells (HUVECs), t-PA was expressed in a minority of cells and found in WP bodies by immunofluorescence. After up-regulation of t-PA synthesis either by vascular endothelial growth factor (VEGF) and retinoic acid or by sodium butyrate, there was a large increase in t-PA-positive cells. t-PA was exclusively located to WP bodies, an observation confirmed by immunoelectron microscopy. Incubation with histamine, forskolin, and epinephrine induced the rapid, coordinate release of both t-PA and VWF, consistent with a single storage compartment. In native human skeletal muscle, t-PA was expressed in endothelial cells from arterioles and venules, along with VWF. The 2 proteins were found to be colocalized in WP bodies by immunoelectron microscopy. These data indicate that t-PA and VWF are colocalized in WP bodies, both in HUVECs and in vivo. Release of both t-PA and VWF from the same storage pool likely accounts for the coordinate increase in the plasma level of the 2 proteins in response to numerous stimuli, such as physical activity, beta-adrenergic agents, and 1-deamino-8d-arginine vasopressin (DDAVP) among others.


Subject(s)
Endothelium, Vascular/chemistry , Tissue Plasminogen Activator/analysis , Weibel-Palade Bodies/chemistry , Cells, Cultured , Endothelium, Vascular/ultrastructure , Humans , Microscopy, Immunoelectron , Muscle, Skeletal/cytology , Tissue Plasminogen Activator/biosynthesis , Tissue Plasminogen Activator/ultrastructure , Up-Regulation , Weibel-Palade Bodies/ultrastructure , von Willebrand Factor/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...