Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Blood ; 138(25): 2607-2620, 2021 12 23.
Article in English | MEDLINE | ID: mdl-34293122

ABSTRACT

In addition to their hemostatic role, platelets play a significant role in immunity. Once activated, platelets release extracellular vesicles (EVs) formed by the budding of their cytoplasmic membranes. Because of their heterogeneity, platelet EVs (PEVs) are thought to perform diverse functions. It is unknown, however, whether the proteasome is transferred from platelets to PEVs or whether its function is retained. We hypothesized that functional protein processing and antigen presentation machinery are transferred to PEVs by activated platelets. Using molecular and functional assays, we found that the active 20S proteasome was enriched in PEVs, along with major histocompatibility complex class I (MHC-I) and lymphocyte costimulatory molecules (CD40L and OX40L). Proteasome-containing PEVs were identified in healthy donor blood, but did not increase in platelet concentrates that caused adverse transfusion reactions. They were augmented, however, after immune complex injections in mice. The complete biodistribution of murine PEVs after injection into mice revealed that they principally reached lymphoid organs, such as spleen and lymph nodes, in addition to the bone marrow, and to a lesser extent, liver and lungs. The PEV proteasome processed exogenous ovalbumin (OVA) and loaded its antigenic peptide onto MHC-I molecules, which promoted OVA-specific CD8+ T-lymphocyte proliferation. These results suggest that PEVs contribute to adaptive immunity through cross-presentation of antigens and have privileged access to immune cells through the lymphatic system, a tissue location that is inaccessible to platelets.


Subject(s)
Blood Platelets/immunology , Extracellular Vesicles/immunology , Histocompatibility Antigens Class I/immunology , Proteasome Endopeptidase Complex/immunology , Animals , Antigen Presentation , Blood Platelets/chemistry , Extracellular Vesicles/chemistry , Histocompatibility Antigens Class I/analysis , Humans , Mice , Mice, Inbred C57BL , Proteasome Endopeptidase Complex/analysis
2.
Sci Transl Med ; 13(581)2021 02 17.
Article in English | MEDLINE | ID: mdl-33597264

ABSTRACT

The accumulation of DNA and nuclear components in blood and their recognition by autoantibodies play a central role in the pathophysiology of systemic lupus erythematosus (SLE). Despite the efforts, the sources of circulating autoantigens in SLE are still unclear. Here, we show that in SLE, platelets release mitochondrial DNA, the majority of which is associated with the extracellular mitochondrial organelle. Mitochondrial release in patients with SLE correlates with platelet degranulation. This process requires the stimulation of platelet FcγRIIA, a receptor for immune complexes. Because mice lack FcγRIIA and murine platelets are completely devoid of receptor capable of binding IgG-containing immune complexes, we used transgenic mice expressing FcγRIIA for our in vivo investigations. FcγRIIA expression in lupus-prone mice led to the recruitment of platelets in kidneys and to the release of mitochondria in vivo. Using a reporter mouse with red fluorescent protein targeted to the mitochondrion, we confirmed platelets as a source of extracellular mitochondria driven by FcγRIIA and its cosignaling by the fibrinogen receptor α2bß3 in vivo. These findings suggest that platelets might be a key source of mitochondrial antigens in SLE and might be a therapeutic target for treating SLE.


Subject(s)
Blood Platelets , Lupus Erythematosus, Systemic , Animals , Antigen-Antibody Complex , Autoantibodies/metabolism , Blood Platelets/metabolism , Humans , Lupus Erythematosus, Systemic/metabolism , Mice , Mitochondria , Receptors, IgG/metabolism
3.
Blood ; 136(25): 2933-2945, 2020 12 17.
Article in English | MEDLINE | ID: mdl-33331924

ABSTRACT

Systemic lupus erythematosus (SLE) is an autoimmune inflammatory disease characterized by deposits of immune complexes (ICs) in organs and tissues. The expression of FcγRIIA by human platelets, which is their unique receptor for immunoglobulin G antibodies, positions them to ideally respond to circulating ICs. Whereas chronic platelet activation and thrombosis are well-recognized features of human SLE, the exact mechanisms underlying platelet activation in SLE remain unknown. Here, we evaluated the involvement of FcγRIIA in the course of SLE and platelet activation. In patients with SLE, levels of ICs are associated with platelet activation. Because FcγRIIA is absent in mice, and murine platelets do not respond to ICs in any existing mouse model of SLE, we introduced the FcγRIIA (FCGR2A) transgene into the NZB/NZWF1 mouse model of SLE. In mice, FcγRIIA expression by bone marrow cells severely aggravated lupus nephritis and accelerated death. Lupus onset initiated major changes to the platelet transcriptome, both in FcγRIIA-expressing and nonexpressing mice, but enrichment for type I interferon response gene changes was specifically observed in the FcγRIIA mice. Moreover, circulating platelets were degranulated and were found to interact with neutrophils in FcγRIIA-expressing lupus mice. FcγRIIA expression in lupus mice also led to thrombosis in lungs and kidneys. The model recapitulates hallmarks of human SLE and can be used to identify contributions of different cellular lineages in the manifestations of SLE. The study further reveals a role for FcγRIIA in nephritis and in platelet activation in SLE.


Subject(s)
Autoantibodies/immunology , Blood Platelets/immunology , Immunoglobulin G/immunology , Lupus Nephritis/immunology , Platelet Activation/immunology , Receptors, IgG/immunology , Animals , Autoantibodies/genetics , Blood Platelets/pathology , Disease Models, Animal , Immunoglobulin G/genetics , Lupus Nephritis/genetics , Lupus Nephritis/pathology , Mice , Mice, Transgenic , Platelet Activation/genetics , Receptors, IgG/genetics
4.
Proc Natl Acad Sci U S A ; 115(7): E1550-E1559, 2018 02 13.
Article in English | MEDLINE | ID: mdl-29386381

ABSTRACT

There is a growing appreciation for the contribution of platelets to immunity; however, our knowledge mostly relies on platelet functions associated with vascular injury and the prevention of bleeding. Circulating immune complexes (ICs) contribute to both chronic and acute inflammation in a multitude of clinical conditions. Herein, we scrutinized platelet responses to systemic ICs in the absence of tissue and endothelial wall injury. Platelet activation by circulating ICs through a mechanism requiring expression of platelet Fcγ receptor IIA resulted in the induction of systemic shock. IC-driven shock was dependent on release of serotonin from platelet-dense granules secondary to platelet outside-in signaling by αIIbß3 and its ligand fibrinogen. While activated platelets sequestered in the lungs and leaky vasculature of the blood-brain barrier, platelets also sequestered in the absence of shock in mice lacking peripheral serotonin. Unexpectedly, platelets returned to the blood circulation with emptied granules and were thereby ineffective at promoting subsequent systemic shock, although they still underwent sequestration. We propose that in response to circulating ICs, platelets are a crucial mediator of the inflammatory response highly relevant to sepsis, viremia, and anaphylaxis. In addition, platelets recirculate after degranulation and sequestration, demonstrating that in adaptive immunity implicating antibody responses, activated platelets are longer lived than anticipated and may explain platelet count fluctuations in IC-driven diseases.


Subject(s)
Anaphylaxis/immunology , Antigen-Antibody Complex/immunology , Blood Platelets/immunology , Serotonin/immunology , Shock, Septic/immunology , Adult , Anaphylaxis/blood , Anaphylaxis/genetics , Animals , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Platelet Activation , Platelet Count , Platelet Glycoprotein GPIIb-IIIa Complex/genetics , Platelet Glycoprotein GPIIb-IIIa Complex/immunology , Receptors, IgG/genetics , Receptors, IgG/immunology , Shock, Septic/blood , Shock, Septic/genetics , Young Adult
5.
J Clin Med ; 6(2)2017 Feb 09.
Article in English | MEDLINE | ID: mdl-28208757

ABSTRACT

Immune thrombocytopenia (ITP) is a complex autoimmune disease characterized by low  platelet counts. The pathogenesis of ITP remains unclear although both antibody-mediated and/or  T cell-mediated platelet destruction are key processes. In addition, impairment of T cells, cytokine  imbalances, and the contribution of the bone marrow niche have now been recognized to be  important. Treatment strategies are aimed at the restoration of platelet counts compatible with  adequate hemostasis rather than achieving physiological platelet counts. The first line treatments  focus on the inhibition of autoantibody production and platelet degradation, whereas second-line  treatments include immunosuppressive drugs, such as Rituximab, and splenectomy. Finally, thirdline treatments aim to stimulate platelet production by megakaryocytes. This review discusses the  pathophysiology  of  ITP  and  how  the  different  treatment  modalities  affect  the  pathogenic  mechanisms.

6.
Blood ; 129(18): 2557-2569, 2017 05 04.
Article in English | MEDLINE | ID: mdl-28202460

ABSTRACT

Transfusion-related acute lung injury (TRALI) is the leading cause of transfusion-related fatalities and is characterized by acute respiratory distress following blood transfusion. Donor antibodies are frequently involved; however, the pathogenesis and protective mechanisms in the recipient are poorly understood, and specific therapies are lacking. Using newly developed murine TRALI models based on injection of anti-major histocompatibility complex class I antibodies, we found CD4+CD25+FoxP3+ T regulatory cells (Tregs) and CD11c+ dendritic cells (DCs) to be critical effectors that protect against TRALI. Treg or DC depletion in vivo resulted in aggravated antibody-mediated acute lung injury within 90 minutes with 60% mortality upon DC depletion. In addition, resistance to antibody-mediated TRALI was associated with increased interleukin-10 (IL-10) levels, and IL-10 levels were found to be decreased in mice suffering from TRALI. Importantly, IL-10 injection completely prevented and rescued the development of TRALI in mice and may prove to be a promising new therapeutic approach for alleviating lung injury in this serious complication of transfusion.


Subject(s)
Acute Lung Injury , Dendritic Cells/immunology , Interleukin-10 , T-Lymphocytes, Regulatory/immunology , Transfusion Reaction , Acute Lung Injury/etiology , Acute Lung Injury/immunology , Acute Lung Injury/prevention & control , Animals , Antibodies/immunology , Dendritic Cells/pathology , Interleukin-10/immunology , Interleukin-10/pharmacology , Mice , Mice, Inbred BALB C , Mice, Knockout , T-Lymphocytes, Regulatory/pathology
7.
Platelets ; 28(3): 214-221, 2017 May.
Article in English | MEDLINE | ID: mdl-28102737

ABSTRACT

Interest in cell-derived extracellular vesicles and their physiological and pathological implications is constantly growing. Microvesicles, also known as microparticles, are small extracellular vesicles released by cells in response to activation or apoptosis. Among the different microvesicles present in the blood of healthy individuals, platelet-derived microvesicles (PMVs) are the most abundant. Their characterization has revealed a heterogeneous cargo that includes a set of adhesion molecules. Similarly to platelets, PMVs are also involved in thrombosis through support of the coagulation cascade. The levels of circulatory PMVs are altered during several disease manifestations such as coagulation disorders, rheumatoid arthritis, systemic lupus erythematosus, cancers, cardiovascular diseases, and infections, pointing to their potential contribution to disease and their development as a biomarker. This review highlights recent findings in the field of PMV research and addresses their contribution to both healthy and diseased states.


Subject(s)
Blood Coagulation/physiology , Blood Platelets/metabolism , Cell-Derived Microparticles/metabolism , Platelet Activation/physiology , Platelet Adhesiveness/physiology , Apoptosis , Arthritis, Rheumatoid/blood , Arthritis, Rheumatoid/diagnosis , Arthritis, Rheumatoid/pathology , Biomarkers/blood , Blood Platelets/pathology , Cardiovascular Diseases/blood , Cardiovascular Diseases/diagnosis , Cardiovascular Diseases/pathology , Cell-Derived Microparticles/chemistry , Humans , Lupus Erythematosus, Systemic/blood , Lupus Erythematosus, Systemic/diagnosis , Lupus Erythematosus, Systemic/pathology , Neoplasms/blood , Neoplasms/diagnosis , Neoplasms/pathology , Neovascularization, Pathologic/blood , Neovascularization, Pathologic/diagnosis , Neovascularization, Pathologic/pathology , Thrombosis/blood , Thrombosis/diagnosis , Thrombosis/pathology
8.
Blood Adv ; 1(20): 1773-1785, 2017 Sep 12.
Article in English | MEDLINE | ID: mdl-29296823

ABSTRACT

Megakaryocytes (MKs) are bone marrow-derived cells that are primarily responsible for generating platelets for the maintenance of hemostasis. Although MK can variably express major histocompatibility complex (MHC) class I and II molecules during their differentiation, little is known whether they can elicit nonhemostatic immune functions such as T-cell activation. Here, we demonstrate that mature CD34- MHC class II- CD41+ MKs can endocytose exogenous ovalbumin (OVA) and proteolytically generate its immunogenic peptide ligand, which is crosspresented on their surface in association with MHC class I molecules. This crosspresentation triggered in vitro and in vivo OVA-specific CD8+ T-cell activation and proliferation. In addition, the OVA-MHC class I complexes were transferred from MK to pro-platelets upon thrombopoiesis in vitro. MK could also present endogenous MK-associated (CD61) peptides to activate CD61-specific CD8+ T cells and mediate immune thrombocytopenia in vivo. These results suggest that, in addition to their hemostatic role, mature MKs can significantly affect antigen-specific CD8+ T-cell responses via antigen presentation and are able to spread this immunogenic information through platelets.

9.
Exp Hematol ; 44(10): 924-930.e1, 2016 10.
Article in English | MEDLINE | ID: mdl-27417974

ABSTRACT

For many years, splenectomy has been used to treat immune thrombocytopenia (ITP), and this procedure benefits approximately two-thirds of the treated patients. Although splenectomy may raise platelet counts, antibody-coated platelets and cytotoxic T lymphocytes appear to persist or can change over time. To better understand how the spleen may affect anti-platelet immune responses, we used a murine model of ITP demonstrating both antibody-mediated and T lymphocyte-mediated thrombocytopenia. Mice with severe combined immunodeficiency (SCID) were either splenectomized or not and transfused with splenocytes from CD61 (GPIIIa) knockout mice immunized against CD61(+) platelets. Platelet counts and anti-platelet antibody levels were performed weekly. After 4 weeks, the mice were sacrificed, and lymphoid organs were harvested and examined by flow cytometry to quantify CD4(+)CD25(+)FoxP3(+) Tregs and conventional cross-presenting XCR1(+) and tolerizing SIRPα+ dendritic cells. The results indicate that compared with control non-splenectomized mice, thrombocytopenia was improved and anti-platelet antibody production was significantly diminished in all splenectomized mice that received immune splenocytes. Splenectomized SCID mice also had a marked reduction in Tregs in the thymus together with an increased proportion of both thymic dendritic cell subsets that correlated with increased platelet counts. Of interest, although splenectomy diminished anti-platelet antibody production and raised platelet counts, marrow megakaryocyte densities were still significantly reduced in mice that received immune splenocytes. These results suggest that the spleen in murine ITP not only is the primary site responsible for platelet destruction, but it also controls, to a significant extent, the antibody response against platelets and the migration patterns of lymphocyte subsets.


Subject(s)
Autoantibodies/immunology , Blood Platelets/immunology , Lymphocytes/immunology , Purpura, Thrombocytopenic, Idiopathic/immunology , Spleen/immunology , Animals , Biomarkers , Bone Marrow/pathology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Disease Models, Animal , Female , Histocompatibility Antigens Class I/immunology , Integrin beta3/genetics , Integrin beta3/immunology , Leukocyte Count , Lymphocytes/metabolism , Lymphocytes/pathology , Megakaryocytes/metabolism , Mice , Mice, Knockout , Phenotype , Platelet Count , Purpura, Thrombocytopenic, Idiopathic/metabolism , Purpura, Thrombocytopenic, Idiopathic/pathology , Purpura, Thrombocytopenic, Idiopathic/therapy , Spleen/pathology , Splenectomy , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/pathology , Thymocytes/immunology , Thymocytes/metabolism
10.
Hum Genet ; 135(4): 403-414, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26883867

ABSTRACT

Platelet reactivity (PR) is variable between individuals and modulates clinical outcome in cardiovascular (CV) patients treated with antiplatelet drugs. Although several data point to a genetic control of platelet reactivity, the genes contributing to the modulation of this phenotype are not clearly identified. Integration of data derived from high-throughput technologies may yield novel insights into the molecular mechanisms that govern platelet reactivity. The aim of this study is to identify candidate genes modulating platelet reactivity in aspirin-treated CV patients using an integrative network-based approach. Patients with extreme high (n = 6) or low PR (n = 6) were selected and data derived from quantitative proteomic of platelets and platelet sub-cellular fractions, as well as from transcriptomic analysis were integrated with a network biology approach. Two modules within the network containing 123 and 182 genes were identified. We then specifically assessed the level of miRNAs in these two groups of patients. Among the 12 miRNAs differentially expressed, 2 (miR-135a-5p and miR-204-5p) correlated with PR. The predicted targets of these miRNAs were mapped onto the network, allowing the identification of seven overlapping genes (THBS1, CDC42, CORO1C, SPTBN1, TPM3, GTPBP2, and MAPRE2), suggesting a synergistic effect of these two miRNAs on these predicted targets. Integration of several omics data sets allowed the identification of 2 candidate miRNAs and 7 candidate genes regulating platelet reactivity in aspirin-treated CV patients.


Subject(s)
Aspirin/pharmacology , Blood Platelets/drug effects , Humans , MicroRNAs/genetics , Proteomics , RNA, Messenger/genetics
11.
Platelets ; 27(3): 196-202, 2016.
Article in English | MEDLINE | ID: mdl-26270593

ABSTRACT

Serum thromboxane B2 (TxB2) is a specific marker of platelet inhibition by aspirin. Yet, TxB2 levels differ by up to 10-fold between some aspirin-treated patient cohorts. This study aimed to identify factors responsible for differences in serum TxB2 between cohorts in the ADRIE study (n = 657) and the BOSTON study (n = 678) of aspirin-treated cardiovascular patients originally tested with different ELISA assays. TxB2 levels were assessed in representative subgroups of the two cohorts (34 samples in BOSTON and 39 in ADRIE) by both ELISAs, as well as liquid chromatography and tandem mass spectroscopy (MS). A multivariate analysis was performed on the whole cohort database to identify determinants of the difference of TxB2 levels between cohorts. There was no systematic bias between the original ELISA TxB2 values and the MS values and the median difference was small, 0.12 ng/ml, thus not explaining the difference between median TxB2 levels in the two study populations (7 and 0.6 ng/ml in the ADRIE and BOSTON studies, respectively). In the combined dataset of the ADRIE and BOSTON cohorts (n = 1342), body mass index, age, gender, aspirin dose, time from aspirin intake to blood draw, NSAID intake, platelet count and C-reactive protein were significantly associated with TxB2 levels. After adjustment for patient characteristics, the difference between cohorts did not decrease. Unexplained differences in serum TxB2 levels in different populations of aspirin-treated cardiovascular patients suggest that further studies are needed to confirm the role of serum TxB2 level as a prognostic factor or rather as a marker of therapeutic observance.


Subject(s)
Aspirin/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Thromboxane B2/blood , Aged , Aspirin/therapeutic use , Biomarkers , Blood Platelets/drug effects , Blood Platelets/metabolism , Cardiovascular Diseases/blood , Cardiovascular Diseases/drug therapy , Chromatography, Liquid , Enzyme-Linked Immunosorbent Assay , Female , Humans , Male , Middle Aged , Platelet Aggregation Inhibitors/therapeutic use , Tandem Mass Spectrometry , Treatment Outcome
12.
Blood ; 127(6): 735-8, 2016 Feb 11.
Article in English | MEDLINE | ID: mdl-26556550

ABSTRACT

Immune thrombocytopenia (ITP) is an autoimmune bleeding disorder with a complex pathogenesis, which includes both antibody- and T-cell-mediated effector mechanisms. Rituximab (an anti-human CD20 monoclonal antibody [mAb]) is one of the treatments for ITP and is known to deplete B cells but may also work by affecting the T-cell compartments. Here, we investigated the outcome of B-cell depletion (Bdep) therapy on CD8(+) T-cell-mediated ITP using a murine model. CD61 knockout (KO) mice were immunized with CD61(+) platelets, and T-cell-mediated ITP was initiated by transfer of their splenocytes into severe combined immunodeficiency (SCID) mice. The CD61 KO mice were administrated an anti-mouse CD20 mAb either before or after CD61(+) platelet immunization. This resulted in efficient Bdep in vivo, accompanied by significant increases in splenic and lymph node CD4(+) and CD8(+) T cells and proportional increases of FOXP3(+) in CD4(+)and CD8(+) T cells. Moreover, Bdep therapy resulted in significantly decreased splenic CD8(+) T-cell proliferation in vitro that could be rescued by interleukin-2. This correlated with normalization of in vivo platelet counts in the transferred SCID mice suggesting that anti-CD20 therapy significantly reduces the ability of CD8(+) T cells to activate and mediate ITP.


Subject(s)
Antigens, CD20/metabolism , B-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , Lymphocyte Depletion/methods , Purpura, Thrombocytopenic, Idiopathic/therapy , Animals , B-Lymphocytes/pathology , Integrin beta3/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, SCID , Platelet Count , Purpura, Thrombocytopenic, Idiopathic/immunology , Purpura, Thrombocytopenic, Idiopathic/pathology
13.
J Immunol ; 194(12): 5579-87, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-26048965

ABSTRACT

Platelets are small cellular fragments with the primary physiological role of maintaining hemostasis. In addition to this well-described classical function, it is becoming increasingly clear that platelets have an intimate connection with infection and inflammation. This stems from several platelet characteristics, including their ability to bind infectious agents and secrete many immunomodulatory cytokines and chemokines, as well as their expression of receptors for various immune effector and regulatory functions, such as TLRs, which allow them to sense pathogen-associated molecular patterns. Furthermore, platelets contain RNA that can be nascently translated under different environmental stresses, and they are able to release membrane microparticles that can transport inflammatory cargo to inflammatory cells. Interestingly, acute infections can also result in platelet breakdown and thrombocytopenia. This report highlights these relatively new aspects of platelets and, thus, their nonhemostatic nature in an inflammatory setting.


Subject(s)
Blood Platelets/immunology , Blood Platelets/metabolism , Inflammation/immunology , Inflammation/metabolism , Animals , CD40 Ligand/metabolism , Cell-Derived Microparticles/metabolism , Cytokines/metabolism , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Inflammation/genetics , Thrombocytopenia/genetics , Thrombocytopenia/immunology , Thrombocytopenia/metabolism , Toll-Like Receptors/metabolism , Transcriptome
14.
J Proteomics ; 114: 125-35, 2015 Jan 30.
Article in English | MEDLINE | ID: mdl-25464367

ABSTRACT

The competition effect between aspirin-mediated acetylation and protein glycation has been a matter of concern for decades. However, the exact interactions between these two post-translational modifications are still not well understood. Several efforts have been made to explain how aspirin prevents glycation, but the influence of prior protein glycation on the action of aspirin has never been investigated. This study involved qualitative and quantitative analyses to: 1) identify acetylated and glycated proteins; 2) quantify rates of acetylation and glycation; and 3) elucidate the common modification sites. Human plasma was incubated with 30mM glucose and then 500µM aspirin. A label-free mass spectrometry approach indicated an increase in the acetylation level after this sequential glucose-then-aspirin incubation; these results were also confirmed by Western blot. Interestingly, for several proteins, decreases in glycation levels were evidenced after aspirin incubation. The common modification sites, where both acetylation and glycation took place, were also identified. The influence that glycation and acetylation processes have on each other could reflect conformational changes induced by glucose and aspirin. In future studies, in order to better understand the interactions between these two PTMs, we intend to apply this strategy to other blood compartments and to diabetic patients. BIOLOGICAL SIGNIFICANCE: Non-enzymatic glycation represents an early stage in the development of the long-lasting complications that are associated with diabetes. Aspirin has been shown to prevent this process in a few reference proteins, but how the two post-translational modifications (PTMs) of aspirin-mediated acetylation and protein glycation interact with each other remains poorly investigated. This study used a label-free quantitative proteomic approach to characterise the extent of aspirin-induced acetylation and protein glycation in human plasma. The results clearly supported a mutual influence between these PTMs, which lead us to propose a potential model based on structural conformational changes.


Subject(s)
Aspirin/pharmacology , Blood Proteins/drug effects , Blood Proteins/metabolism , Protein Processing, Post-Translational/drug effects , Acetylation/drug effects , Amino Acid Sequence , Blood Proteins/chemistry , Glucose/pharmacology , Glycoproteins/chemistry , Glycoproteins/metabolism , Glycosylation/drug effects , Humans , Male , Models, Molecular , Molecular Sequence Data
15.
J Proteomics ; 101: 130-40, 2014 Apr 14.
Article in English | MEDLINE | ID: mdl-24549006

ABSTRACT

In the present study, we performed an extensive qualitative characterization of the platelet granule proteome using subcellular fractionation followed by mass spectrometry analysis and functional annotation. Eight-hundred-and-twenty-seven proteins were identified, most of them being associated to granules and to the granule's secretory machinery. Functional pathway analysis revealed 30 pathways, including the major histocompatibility complex class 1 (MHC I) presenting antigen pathway. This pathway was of particular interest for its potential interrelation between platelets and the immune system. Key proteins belonging to this metabolic route such as ß-2-microglobulin, 26S protease regulatory subunit 10B from the proteasome and proteins 1 and 2 of the transporter associated with antigen processing were shown to co-localize with von Willebrand factor in resting platelets and to be located on the plasma membrane when platelets were activated. Key proteins of the MHC1 antigen-presenting pathway are located in platelet alpha-granules. These results suggest a possible functional role of platelet granules in platelet-related immune modulation. BIOLOGICAL SIGNIFICANCE: In this study, we described the largest dataset related to platelet granule proteins. We performed a functional pathway analysis that evidenced several expected granule-related pathways. We also highlighted the "Antigen processing and presentation" pathway that has drawn our attention. Using immunofluorescence technique, we confirmed the presence of several key proteins for antigen presentation in platelet granules. This study suggests a putative functional role of MHC1 and platelet granules in the immune modulation.


Subject(s)
Blood Platelets/metabolism , Blood Platelets/ultrastructure , Blood Proteins/analysis , Cytoplasmic Granules/metabolism , Histocompatibility Antigens Class I/metabolism , Proteome/analysis , Adult , Antigen Presentation , Blood Proteins/metabolism , Humans , Male , Mass Spectrometry , Proteome/metabolism , Signal Transduction
16.
J Proteomics ; 79: 146-60, 2013 Feb 21.
Article in English | MEDLINE | ID: mdl-23277275

ABSTRACT

High throughput protein identification and quantification analysis based on mass spectrometry are fundamental steps in most proteomics projects. Here, we present EasyProt (available at http://easyprot.unige.ch), a new platform for mass spectrometry data processing, protein identification, quantification and unexpected post-translational modification characterization. EasyProt provides a fully integrated graphical experience to perform a large part of the proteomic data analysis workflow. Our goal was to develop a software platform that would fulfill the needs of scientists in the field, while emphasizing ease-of-use for non-bioinformatician users. Protein identification is based on OLAV scoring schemes and protein quantification is implemented for both, isobaric labeling and label-free methods. Additional features are available, such as peak list processing, isotopic correction, spectra filtering, charge-state deconvolution and spectra merging. To illustrate the EasyProt platform, we present two identification and quantification workflows based on isobaric tagging and label-free methods.


Subject(s)
Proteomics/methods , Sequence Analysis, Protein/methods , Software , Mass Spectrometry/methods , Protein Processing, Post-Translational , Proteins/analysis
17.
Mass Spectrom Rev ; 31(2): 331-51, 2012.
Article in English | MEDLINE | ID: mdl-22009795

ABSTRACT

Platelets are small cell fragments, produced by megakaryocytes, in the bone marrow. They play an important role in hemostasis and diverse thrombotic disorders. They are therefore primary targets of antithrombotic therapies. They are implicated in several pathophysiological pathways, such as inflammation or wound repair. In blood circulation, platelets are activated by several pathways including subendothelial matrix and thrombin, triggering the formation of the platelet plug. Studying their proteome is a powerful approach to understand their biology and function. However, particular attention must be paid to different experimental parameters, such as platelet quality and purity. Several technologies are involved during the platelet proteome processing, yielding information on protein identification, characterization, localization, and quantification. Recent technical improvements in proteomics combined with inter-disciplinary strategies, such as metabolomic, transcriptomics, and bioinformatics, will help to understand platelets biological mechanisms. Therefore, a comprehensive analysis of the platelet proteome under different environmental conditions may contribute to elucidate complex processes relevant to platelet function regarding bleeding disorders or platelet hyperreactivity and identify new targets for antiplatelet therapy.


Subject(s)
Blood Platelets/chemistry , Blood Platelets/metabolism , Proteome/analysis , Proteomics/methods , Animals , Blood Platelets/cytology , Blood Platelets/pathology , Humans , Platelet Activation , Proteome/isolation & purification , Proteome/metabolism
18.
Thromb Haemost ; 106(3): 466-74, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21725581

ABSTRACT

In healthy subjects, platelet hyperreactivity is a global phenomenon--as opposed to agonist-specific--and epinephrine-induced platelet aggregation (EPA) is a reliable marker of this phenotype. Few data are available on platelet reactivity and the relationship between EPA and aggregation induced by other agonists in cardiovascular patients. It was the objective of this study to characterise platelet reactivity in stable cardiovascular patients treated with aspirin and to derive a composite index integrating several aggregation pathways, suitable for selecting patients with extreme phenotypes for further proteomics analysis. Platelet reactivity to agonists was assessed in 110 patients twice, two weeks apart. Factorial analysis was used to determine whether the results obtained with the different agonists could be summarised in a single composite index. EPA correlated with the aggregation values obtained with each of the other agonists, with correlation coefficients of 0.44 to 0.55 (p < 0.001). We constructed a composite "platelet reactivity" index that included 60% of the information provided by each agonist. The results obtained at the first patient visit were consistent with those obtained at the second visit (r = 0.78, p<0.01). No clinical or biological parameters correlated with the composite index. The extreme phenotypes of six selected subjects were confirmed 12 months after the second visit. In conclusion, platelet reactivity in aspirin-treated cardiovascular patients is a global phenomenon that can be summarised by a composite index based on the aggregation responses to various agonists and integrating several activation pathways. This index is not dependent on clinical or biological variables, suggesting that genetic factors regulate platelet reactivity in these patients.


Subject(s)
Aspirin/administration & dosage , Cardiovascular Diseases/drug therapy , Epinephrine/metabolism , Platelet Activation , Adenosine Diphosphate/pharmacology , Aged , Aspirin/adverse effects , Cardiovascular Diseases/pathology , Cardiovascular Diseases/physiopathology , Female , Follow-Up Studies , Humans , Male , Middle Aged , Platelet Activation/drug effects , Platelet Function Tests , Practice Guidelines as Topic , Proteomics , Severity of Illness Index
19.
Forensic Sci Int ; 167(2-3): 242-6, 2007 Apr 11.
Article in English | MEDLINE | ID: mdl-16884878

ABSTRACT

Pattern recognition techniques can be very useful in forensic sciences to point out to relevant sets of events and potentially encourage an intelligence-led style of policing. In this study, these techniques have been applied to categorical data corresponding to cutting agents found in heroin seizures. An application of graph theoretic methods has been performed, in order to highlight the possible relationships between the location of seizures and co-occurrences of particular heroin cutting agents. An analysis of the co-occurrences to establish several main combinations has been done. Results illustrate the practical potential of mathematical models in forensic data analysis.

SELECTION OF CITATIONS
SEARCH DETAIL
...