Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
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
2.
Arterioscler Thromb Vasc Biol ; 40(4): 929-942, 2020 04.
Article in English | MEDLINE | ID: mdl-32102567

ABSTRACT

OBJECTIVE: The lymphatic system is a circulatory system that unidirectionally drains the interstitial tissue fluid back to blood circulation. Although lymph is utilized by leukocytes for immune surveillance, it remains inaccessible to platelets and erythrocytes. Activated cells release submicron extracellular vesicles (EV) that transport molecules from the donor cell. In rheumatoid arthritis, EV accumulate in the joint where they can interact with numerous cellular lineages. However, whether EV can exit the inflamed tissue to recirculate is unknown. Here, we investigated whether vascular leakage that occurs during inflammation could favor EV access to the lymphatic system. Approach and Results: Using an in vivo model of autoimmune inflammatory arthritis, we show that there is an influx of platelet EV, but not EV from erythrocytes or leukocytes, in joint-draining lymph. In contrast to blood platelet EV, lymph platelet EV lacked mitochondrial organelles and failed to promote coagulation. Platelet EV influx in lymph was consistent with joint vascular leakage and implicated the fibrinogen receptor α2bß3 and platelet-derived serotonin. CONCLUSIONS: These findings show that platelets can disseminate their EV in fluid that is inaccessible to platelets and beyond the joint in this disease.


Subject(s)
Arthritis, Rheumatoid/physiopathology , Blood Platelets/physiology , Extracellular Vesicles/physiology , Lymph/physiology , Animals , Blood Platelets/metabolism , Capillary Permeability , Disease Models, Animal , Mice, Inbred C57BL , Serotonin/metabolism
3.
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
4.
Neurobiol Dis ; 124: 163-175, 2019 04.
Article in English | MEDLINE | ID: mdl-30408591

ABSTRACT

The production of extracellular vesicles (EV) is a ubiquitous feature of eukaryotic cells but pathological events can affect their formation and constituents. We sought to characterize the nature, profile and protein signature of EV in the plasma of Parkinson's disease (PD) patients and how they correlate to clinical measures of the disease. EV were initially collected from cohorts of PD (n = 60; Controls, n = 37) and Huntington's disease (HD) patients (Pre-manifest, n = 11; manifest, n = 52; Controls, n = 55) - for comparative purposes in individuals with another chronic neurodegenerative condition - and exhaustively analyzed using flow cytometry, electron microscopy and proteomics. We then collected 42 samples from an additional independent cohort of PD patients to confirm our initial results. Through a series of iterative steps, we optimized an approach for defining the EV signature in PD. We found that the number of EV derived specifically from erythrocytes segregated with UPDRS scores corresponding to different disease stages. Proteomic analysis further revealed that there is a specific signature of proteins that could reliably differentiate control subjects from mild and moderate PD patients. Taken together, we have developed/identified an EV blood-based assay that has the potential to be used as a biomarker for PD.


Subject(s)
Erythrocytes/metabolism , Extracellular Vesicles/metabolism , Parkinson Disease/blood , Aged , Biomarkers/blood , Blood Cell Count , Erythrocytes/ultrastructure , Extracellular Vesicles/ultrastructure , Female , Humans , Huntington Disease/blood , Huntington Disease/diagnosis , Huntington Disease/pathology , Male , Middle Aged , Parkinson Disease/diagnosis , Parkinson Disease/pathology , Proteomics
5.
J Neurol Neurosurg Psychiatry ; 90(3): 272-283, 2019 03.
Article in English | MEDLINE | ID: mdl-30567722

ABSTRACT

Huntington's disease (HD) is a hereditary disorder that typically manifests in adulthood with a combination of motor, cognitive and psychiatric problems. The pathology is caused by a mutation in the huntingtin gene which results in the production of an abnormal protein, mutant huntingtin (mHtt). This protein is ubiquitously expressed and known to confer toxicity to multiple cell types. We have recently reported that HD brains are also characterised by vascular abnormalities, which include changes in blood vessel density/diameter as well as increased blood-brain barrier (BBB) leakage. OBJECTIVES: Seeking to elucidate the origin of these vascular and BBB abnormalities, we studied platelets that are known to play a role in maintaining the integrity of the vasculature and thrombotic pathways linked to this, given they surprisingly contain the highest concentration of mHtt of all blood cells. METHODS: We assessed the functional status of platelets by performing ELISA, western blot and RNA sequencing in a cohort of 71 patients and 68 age- and sex-matched healthy control subjects. We further performed haemostasis and platelet depletion tests in the R6/2 HD mouse model. RESULTS: Our findings indicate that the platelets in HD are dysfunctional with respect to the release of angiogenic factors and functions including thrombosis, angiogenesis and vascular haemostasis. CONCLUSION: Taken together, our results provide a better understanding for the impact of mHtt on platelet function.


Subject(s)
Blood-Brain Barrier/physiopathology , Huntingtin Protein/blood , Huntington Disease/blood , Platelet Activation/physiology , Adult , Aged , Angiogenic Proteins/blood , Animals , Blood Coagulation Factors/metabolism , Case-Control Studies , Cohort Studies , Disease Models, Animal , Female , Fibroblast Growth Factor 2/blood , Humans , Huntington Disease/complications , Male , Mice , Middle Aged , Platelet Count
6.
Prostaglandins Other Lipid Mediat ; 143: 106340, 2019 08.
Article in English | MEDLINE | ID: mdl-31129176

ABSTRACT

Phospholipase A2s (PLA2) play a key role in generation of eicosanoids. Cytosolic PLA2α (cPLA2α) is constitutively expressed in most cells, whereas IIA secreted PLA2 (sPLA2-IIA) is induced during inflammation and is present at high levels in the synovial fluid of rheumatoid arthritis patients. In mice, both cPLA2α and sPLA2-IIA have been implicated in autoimmune arthritis; however, the respective contribution of these two enzymes to the pathogenesis and production of eicosanoids is unknown. We evaluated the respective role of cPLA2α and sPLA2-IIA with regard to arthritis and eicosanoid profile in an in vivo model of arthritis. While arthritis was most severe in mice expressing both enzymes, it was abolished when both cPLA2α and sPLA2-IIA were lacking. cPLA2α played a dominant role in the severity of arthritis, although sPLA2-IIA sufficed to significantly contribute to the disease. Several eicosanoids were modulated during the course of arthritis and numerous species involved sPLA2-IIA expression. This study confirms the critical role of PLA2s in arthritis and unveils the distinct contribution of cPLA2α and sPLA2-IIA to the eicosanoid profile in arthritis.


Subject(s)
Arthritis/metabolism , Eicosanoids/biosynthesis , Group II Phospholipases A2/metabolism , Group IV Phospholipases A2/metabolism , Animals , Arthritis/enzymology , Female , Gene Expression Regulation, Enzymologic , Group II Phospholipases A2/genetics , Group IV Phospholipases A2/genetics , Inflammation/enzymology , Lipidomics , Mice
7.
Proc Natl Acad Sci U S A ; 112(27): E3564-73, 2015 Jul 07.
Article in English | MEDLINE | ID: mdl-26106157

ABSTRACT

Platelets are anucleated blood elements highly potent at generating extracellular vesicles (EVs) called microparticles (MPs). Whereas EVs are accepted as an important means of intercellular communication, the mechanisms underlying platelet MP internalization in recipient cells are poorly understood. Our lipidomic analyses identified 12(S)-hydroxyeicosatetranoic acid [12(S)-HETE] as the predominant eicosanoid generated by MPs. Mechanistically, 12(S)-HETE is produced through the concerted activity of secreted phospholipase A2 IIA (sPLA2-IIA), present in inflammatory fluids, and platelet-type 12-lipoxygenase (12-LO), expressed by platelet MPs. Platelet MPs convey an elaborate set of transcription factors and nucleic acids, and contain mitochondria. We observed that MPs and their cargo are internalized by activated neutrophils in the endomembrane system via 12(S)-HETE. Platelet MPs are found inside neutrophils isolated from the joints of arthritic patients, and are found in neutrophils only in the presence of sPLA2-IIA and 12-LO in an in vivo model of autoimmune inflammatory arthritis. Using a combination of genetically modified mice, we show that the coordinated action of sPLA2-IIA and 12-LO promotes inflammatory arthritis. These findings identify 12(S)-HETE as a trigger of platelet MP internalization by neutrophils, a mechanism highly relevant to inflammatory processes. Because sPLA2-IIA is induced during inflammation, and 12-LO expression is restricted mainly to platelets, these observations demonstrate that platelet MPs promote their internalization in recipient cells through highly regulated mechanisms.


Subject(s)
Arachidonate 12-Lipoxygenase/metabolism , Blood Platelets/metabolism , Cell-Derived Microparticles/metabolism , Group II Phospholipases A2/metabolism , Neutrophils/metabolism , 12-Hydroxy-5,8,10,14-eicosatetraenoic Acid/metabolism , Animals , Arachidonate 12-Lipoxygenase/genetics , Arthritis, Experimental/genetics , Arthritis, Experimental/metabolism , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/metabolism , Blood Platelets/enzymology , Cell Line , Cell-Derived Microparticles/enzymology , Cell-Derived Microparticles/ultrastructure , Cells, Cultured , Endocytosis , Group II Phospholipases A2/genetics , Humans , Immunoblotting , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Confocal , Microscopy, Electron , Mitochondria/metabolism , Mitochondria/ultrastructure , Neutrophils/ultrastructure , RNA/genetics , RNA/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Synovial Fluid/metabolism
8.
Blood ; 123(18): 2854-63, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24665136

ABSTRACT

Platelets play crucial functions in hemostasis and the prevention of bleeding. During H1N1 influenza A virus infection, platelets display activation markers. The platelet activation triggers during H1N1 infection remain elusive. We observed that H1N1 induces surface receptor activation, lipid mediator synthesis, and release of microparticles from platelets. These activation processes require the presence of serum/plasma, pointing to the contribution of soluble factor(s). Considering that immune complexes in the H1N1 pandemic were reported to play a pathogenic role, we assessed their contribution in H1N1-induced platelet activation. In influenza-immunized subjects, we observed that the virus scaffolds with immunoglobulin G (IgG) to form immune complexes that promote platelet activation. Mechanistically, this activation occurs through stimulation of low-affinity type 2 receptor for Fc portion of IgG (FcγRIIA), a receptor for immune complexes, independently of thrombin. Using a combination of in vitro and in vivo approaches, we found that the antibodies from H3N2-immunized mice activate transgenic mouse platelets that express FcγRIIA when put in the presence of H1N1, suggesting that cross-reacting influenza antibodies suffice. Alternatively, H1N1 can activate platelets via thrombin formation, independently of complement and FcγRIIA. These observations identify both the adaptive immune response and the innate response against pathogens as 2 intertwined processes that activate platelets during influenza infections.


Subject(s)
Blood Platelets/immunology , Blood Platelets/metabolism , Influenza A Virus, H1N1 Subtype/immunology , Platelet Activation/immunology , Receptors, IgG/metabolism , Signal Transduction , Thrombin/metabolism , Animals , Antibodies, Viral/immunology , Antigen-Antibody Complex/immunology , Humans , Immunity, Innate , Immunophenotyping , Mice , Mice, Transgenic , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/metabolism , Phenotype , Receptors, IgG/genetics
9.
Blood ; 124(14): 2173-83, 2014 Oct 02.
Article in English | MEDLINE | ID: mdl-25082876

ABSTRACT

Mitochondrial DNA (mtDNA) is a highly potent inflammatory trigger and is reportedly found outside the cells in blood in various pathologies. Platelets are abundant in blood where they promote hemostasis. Although lacking a nucleus, platelets contain functional mitochondria. On activation, platelets produce extracellular vesicles known as microparticles. We hypothesized that activated platelets could also release their mitochondria. We show that activated platelets release respiratory-competent mitochondria, both within membrane-encapsulated microparticles and as free organelles. Extracellular mitochondria are found in platelet concentrates used for transfusion and are present at higher levels in those that induced acute reactions (febrile nonhemolytic reactions, skin manifestations, and cardiovascular events) in transfused patients. We establish that the mitochondrion is an endogenous substrate of secreted phospholipase A2 IIA (sPLA2-IIA), a phospholipase otherwise specific for bacteria, likely reflecting the ancestral proteobacteria origin of mitochondria. The hydrolysis of the mitochondrial membrane by sPLA2-IIA yields inflammatory mediators (ie, lysophospholipids, fatty acids, and mtDNA) that promote leukocyte activation. Two-photon microscopy in live transfused animals revealed that extracellular mitochondria interact with neutrophils in vivo, triggering neutrophil adhesion to the endothelial wall. Our findings identify extracellular mitochondria, produced by platelets, at the midpoint of a potent mechanism leading to inflammatory responses.


Subject(s)
Blood Platelets/metabolism , Group II Phospholipases A2/metabolism , Inflammation/metabolism , Mitochondria/metabolism , Animals , DNA, Mitochondrial/metabolism , Endothelium, Vascular/metabolism , Flow Cytometry , Humans , Male , Mice , Mice, Inbred C57BL , Platelet Activation , Rickettsia prowazekii/metabolism
10.
Blood ; 122(2): 253-61, 2013 Jul 11.
Article in English | MEDLINE | ID: mdl-23652806

ABSTRACT

Platelets play a crucial role in the maintenance of hemostasis, as well as in thrombosis. Upon activation, platelets release small membrane-bound microparticles (MPs) containing bioactive proteins and genetic materials from their parental cells that may be transferred to, and exert potent biological effects in, recipient cells of the circulatory system. Platelets have been shown to contain an abundant and diverse array of microRNAs, and platelet-derived MPs are the most abundant microvesicles in the circulation. Here we demonstrate that human platelets activated with thrombin preferentially release their miR-223 content in MPs. These MPs can be internalized by human umbilical vein endothelial cells (HUVEC), leading to the accumulation of platelet-derived miR-223. Platelet MPs contain functional Argonaute 2 (Ago2)•miR-223 complexes that are capable of regulating expression of a reporter gene in recipient HUVEC. Moreover, we demonstrate a role for platelet MP-derived miR-223 in the regulation of 2 endogenous endothelial genes, both at the messenger RNA and protein levels. Our results support a scenario by which platelet MPs may act as intercellular carriers of functional Ago2•microRNA complexes that may exert heterotypic regulation of gene expression in endothelial cells, and possibly other recipient cells of the circulatory system.


Subject(s)
Argonaute Proteins/genetics , Blood Platelets/physiology , Cell-Derived Microparticles/physiology , Endothelial Cells/metabolism , MicroRNAs/genetics , Platelet Activation/physiology , RNA, Messenger/genetics , Argonaute Proteins/metabolism , Biological Transport , Gene Expression Regulation , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Macromolecular Substances/metabolism , MicroRNAs/metabolism , RNA Interference , RNA, Messenger/metabolism
11.
Blood ; 120(6): 1334-43, 2012 Aug 09.
Article in English | MEDLINE | ID: mdl-22544703

ABSTRACT

Platelets survey blood vessels, searching for endothelial damage and preventing loss of vascular integrity. However, there are circumstances where vascular permeability increases, suggesting that platelets sometimes fail to fulfill their expected function. Human inflammatory arthritis is associated with tissue edema attributed to enhanced permeability of the synovial microvasculature. Murine studies have suggested that such vascular leak facilitates entry of autoantibodies and may thereby promote joint inflammation. Whereas platelets typically help to promote microvascular integrity, we examined the role of platelets in synovial vascular permeability in murine experimental arthritis. Using an in vivo model of autoimmune arthritis, we confirmed the presence of endothelial gaps in inflamed synovium. Surprisingly, permeability in the inflamed joints was abrogated if the platelets were absent. This effect was mediated by platelet serotonin accumulated via the serotonin transporter and could be antagonized using serotonin-specific reuptake inhibitor antidepressants. As opposed to the conventional role of platelets to microvascular leakage, this demonstration that platelets are capable of amplifying and maintaining permeability adds to the rapidly growing list of unexpected functions for platelets.


Subject(s)
Blood Platelets/physiology , Capillary Permeability/physiology , Animals , Arthritis/genetics , Arthritis/immunology , Arthritis/pathology , Arthritis/physiopathology , Autoimmune Diseases/genetics , Autoimmune Diseases/pathology , Autoimmune Diseases/physiopathology , Blood Platelets/metabolism , Capillary Permeability/genetics , Female , Gap Junctions/drug effects , Gap Junctions/metabolism , Humans , Joints/metabolism , Joints/pathology , Joints/physiopathology , Joints/ultrastructure , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Serotonin/metabolism , Serotonin/pharmacology , Up-Regulation/genetics , Up-Regulation/physiology
12.
J Patient Exp ; 11: 23743735241229373, 2024.
Article in English | MEDLINE | ID: mdl-38618513

ABSTRACT

Background: The aim of this study was to develop a patient-reported experience measure (PREM) for comparing the experience of care received by ambulatory patients with acute unexpected needs presenting in emergency departments (EDs), walk-in clinics, and primary care practices. Methods: The Ambulatory Patient EXperience (APEX) questionnaire was developed using a 5-phase mixed-methods approach. The questionnaire was pretested by asking potential users to rate its clarity, usefulness, redundancy, content and face validities, and discrimination on a 9-point scale (1 = strongly disagree to 9 = strongly agree). The pre-final version was then tested in a pilot study. Results: The final questionnaire is composed of 61 questions divided into 7 sections. In the pretest (n = 25), median responses were 8 and above for all dimensions assessed. In the pilot study, 63 participants were enrolled. Adjusted results show that access, cleanliness, and feeling treated with respect and dignity by nurses and physicians were significantly better in the clinics than in the ED. Conclusion: We developed a questionnaire to assess and compare experience of ambulatory care in different clinical settings.

13.
JCI Insight ; 7(2)2022 01 25.
Article in English | MEDLINE | ID: mdl-35076027

ABSTRACT

Secreted phospholipase A2-IIA (sPLA2-IIA) hydrolyzes phospholipids to liberate lysophospholipids and fatty acids. Given its poor activity toward eukaryotic cell membranes, its role in the generation of proinflammatory lipid mediators is unclear. Conversely, sPLA2-IIA efficiently hydrolyzes bacterial membranes. Here, we show that sPLA2-IIA affects the immune system by acting on the intestinal microbial flora. Using mice overexpressing transgene-driven human sPLA2-IIA, we found that the intestinal microbiota was critical for both induction of an immune phenotype and promotion of inflammatory arthritis. The expression of sPLA2-IIA led to alterations of the intestinal microbiota composition, but housing in a more stringent pathogen-free facility revealed that its expression could affect the immune system in the absence of changes to the composition of this flora. In contrast, untargeted lipidomic analysis focusing on bacteria-derived lipid mediators revealed that sPLA2-IIA could profoundly alter the fecal lipidome. The data suggest that a singular protein, sPLA2-IIA, produces systemic effects on the immune system through its activity on the microbiota and its lipidome.


Subject(s)
Arthritis , Bacterial Physiological Phenomena/immunology , Gastrointestinal Microbiome/physiology , Group II Phospholipases A2/metabolism , Lipid Metabolism/immunology , Animals , Animals, Genetically Modified , Arthritis/immunology , Arthritis/microbiology , Humans , Immune System Phenomena , Lipidomics/methods , Mice , Models, Animal , Pathology, Molecular/methods , Transgenes
14.
J Biol Chem ; 285(10): 7208-21, 2010 Mar 05.
Article in English | MEDLINE | ID: mdl-20048166

ABSTRACT

Host cells respond to viral infections by synthesizing and producing antiviral molecules such as type I interferons (IFN). The Kaposi sarcoma-associated herpesvirus (KSHV) encodes multiple proteins expressed during the lytic replication cycle that alter the antiviral response of the host. Considering that in Kaposi sarcoma lesions and primary effusion lymphoma cells KSHV is latent in the vast majority of cells, we were interested in determining whether latently expressed viral proteins have the ability to modulate IFN synthesis. The latency-associated nuclear antigen (LANA-1) is a large nuclear protein that plays a role in the establishment and maintenance of latent KSHV episome in the nucleus of infected cells. LANA-1 is also described to modulate the cellular transcription. Here, we report that LANA-1 inhibits IFN-beta transcription and synthesis by competing with the binding of interferon regulatory factor-3 (IRF3) to the IFNB promoter. Using mutants of LANA-1, we have identified the central acidic repeated region as the domain essential for interfering with the binding of IRF3 to the positive regulatory domains I-III of the IFNB promoter. In addition, the nuclear localization of LANA-1 proved essential for IFN-beta inhibition. Thus, LANA-1 interferes with the formation of IFN-beta enhanceosome by competing with the fixation of IRF3 and by inhibiting the expression of the CREB-binding protein. The ability of LANA-1 to inhibit IFNB gene expression highlights a new role for this protein in cellular gene modulation and immune evasion strategies.


Subject(s)
Antigens, Viral/metabolism , Herpesvirus 8, Human/metabolism , Interferon Regulatory Factor-3/metabolism , Interferon-beta , Nuclear Proteins/metabolism , Promoter Regions, Genetic , Animals , Antigens, Viral/genetics , CREB-Binding Protein/genetics , CREB-Binding Protein/metabolism , Cell Line , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Gene Expression Regulation , Genes, Reporter , Herpesvirus 8, Human/genetics , Humans , Interferon Regulatory Factor-3/genetics , Interferon-beta/genetics , Interferon-beta/metabolism , Nuclear Proteins/genetics , Protein Binding , Sarcoma, Kaposi/virology , Virus Latency
15.
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
16.
Blood Adv ; 3(3): 256-267, 2019 02 12.
Article in English | MEDLINE | ID: mdl-30696624

ABSTRACT

Antibody ligation of the murine neutrophil surface protein Ly6G disrupts neutrophil migration in some contexts but not others. We tested whether this variability reflected divergent dependence of neutrophil migration on ß2 integrins, adhesion molecules that interact with Ly6G at the neutrophil surface. In integrin-dependent murine arthritis, Ly6G ligation attenuated joint inflammation, even though mice lacking Ly6G altogether developed arthritis normally. By contrast, Ly6G ligation had no impact on integrin-independent neutrophil migration into inflamed lung. In peritoneum, the role of ß2 integrins varied with stimulus, proving dispensable for neutrophil entry in Escherichia coli peritonitis but contributory in interleukin 1 (IL-1)-mediated sterile peritonitis. Correspondingly, Ly6G ligation attenuated only IL-1 peritonitis, disrupting the molecular association between integrins and Ly6G and inducing cell-intrinsic blockade restricted to integrin-dependent migration. Consistent with this observation, Ly6G ligation impaired integrin-mediated postadhesion strengthening for neutrophils arresting on activated cremaster endothelium in vivo. Together, these findings identify selective inhibition of integrin-mediated neutrophil emigration through Ly6G ligation, highlighting the marked site and stimulus specificity of ß2 integrin dependence in neutrophil migration.


Subject(s)
Antigens, Ly/blood , CD18 Antigens/blood , Neutrophils/metabolism , Animals , Cell Movement/physiology , Lung/cytology , Male , Mice , Mice, Inbred C57BL , Neutrophils/cytology , Neutrophils/pathology , Peritonitis/blood , Peritonitis/pathology
17.
Sci Rep ; 9(1): 4530, 2019 03 14.
Article in English | MEDLINE | ID: mdl-30872710

ABSTRACT

Mitochondria are organelles that govern energy supply and control cell death. Mitochondria also express bacterial features, such as the presence of inner membrane cardiolipin and a circular genome rich in hypomethylated CpG motifs. While mitochondrial extrusion by damaged organs or activated cells is thought to trigger innate immunity, it is unclear whether extracellular mitochondria also stimulate an adaptive immune response. We describe the development of novel assays to detect autoantibodies specific to two distinct components of the mitochondrion: the mitochondrial outer membrane and mitochondrial DNA. Antibodies to these two mitochondrial constituents were increased in both human and murine systemic lupus erythematosus (SLE), compared to controls, and were present at higher levels than in patients with antiphospholipid syndrome or primary biliary cirrhosis. In both bi- and multi-variate regression models, antibodies to mitochondrial DNA, but not whole mitochondria, were associated with increased anti-dsDNA antibodies and lupus nephritis. This study describes new and optimized methods for the assessment of anti-mitochondrial antibodies, and demonstrates their presence in both human and murine SLE. These findings suggest that different mitochondrial components are immunogenic in SLE, and support the concept that extracellular mitochondria may provide an important source of circulating autoantigens in SLE.


Subject(s)
Autoantibodies/immunology , Lupus Erythematosus, Systemic/immunology , Mitochondria/immunology , Adult , Aged , Animals , Antibodies, Antinuclear/blood , Antibodies, Antinuclear/immunology , Autoantibodies/blood , DNA, Mitochondrial/immunology , Disease Models, Animal , Female , Hep G2 Cells , Humans , Lupus Erythematosus, Systemic/pathology , Male , Mice , Middle Aged , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Proteins/immunology , Odds Ratio , Young Adult
18.
Sci Rep ; 7(1): 5000, 2017 07 10.
Article in English | MEDLINE | ID: mdl-28694505

ABSTRACT

Beta-cell (ß-cell) injury is the hallmark of autoimmune diabetes. However, the mechanisms by which autoreactive responses are generated in susceptible individuals are not well understood. Extracellular vesicles (EV) are produced by mammalian cells under normal and stressed physiological states. They are an important part of cellular communication, and may serve a role in antigen processing and presentation. We hypothesized that isolated human islets in culture produce EV that contain diabetes autoantigens (DAA) from these otherwise normal, non-diabetic donors. Here we report the caspase-independent production of EV by human islets in culture, and the characterization of DAA glutamic acid decarboxylase 65 (GAD65) and zinc transporter 8 (ZnT8), as well as the ß-cell resident glucose transporter 2 (Glut2), present within the EV.


Subject(s)
Autoantigens/metabolism , Diabetes Mellitus, Type 1/immunology , Extracellular Vesicles/immunology , Islets of Langerhans/cytology , Antigen Presentation , Cells, Cultured , Glucose Transporter Type 2/metabolism , Glutamate Decarboxylase/metabolism , Humans , Islets of Langerhans/immunology , Proteomics/methods , Zinc Transporter 8/metabolism
19.
Sci Rep ; 6: 35928, 2016 10 27.
Article in English | MEDLINE | ID: mdl-27786276

ABSTRACT

Extracellular vesicles (EV) are small membrane vesicles produced by cells upon activation and apoptosis. EVs are heterogeneous according to their origin, mode of release, membrane composition, organelle and biochemical content, and other factors. Whereas it is apparent that EVs are implicated in intercellular communication, they can also be used as biomarkers. Continuous improvements in pre-analytical parameters and flow cytometry permit more efficient assessment of EVs; however, methods to more objectively distinguish EVs from cells and background, and to interpret multiple single-EV parameters are lacking. We used spanning-tree progression analysis of density-normalized events (SPADE) as a computational approach for the organization of EV subpopulations released by platelets and erythrocytes. SPADE distinguished EVs, and logically organized EVs detected by high-sensitivity flow cytofluorometry based on size estimation, granularity, mitochondrial content, and phosphatidylserine and protein receptor surface expression. Plasma EVs were organized by hierarchy, permitting appreciation of their heterogeneity. Furthermore, SPADE was used to analyze EVs present in the synovial fluid of patients with inflammatory arthritis. Its algorithm efficiently revealed subtypes of arthritic patients based on EV heterogeneity patterns. Our study reveals that computational algorithms are useful for the analysis of high-dimensional single EV data, thereby facilitating comprehension of EV functions and biomarker development.


Subject(s)
Extracellular Vesicles/classification , Flow Cytometry/methods , Algorithms , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Biomarkers/metabolism , Blood Platelets/drug effects , Blood Platelets/metabolism , Blood Platelets/ultrastructure , Erythrocytes/metabolism , Erythrocytes/ultrastructure , Extracellular Vesicles/metabolism , Extracellular Vesicles/ultrastructure , Flow Cytometry/statistics & numerical data , Humans , In Vitro Techniques , Particle Size , Synovial Fluid/cytology , Synovial Fluid/metabolism , Thrombin/pharmacology
20.
J Rheumatol ; 43(11): 2019-2025, 2016 11.
Article in English | MEDLINE | ID: mdl-27585687

ABSTRACT

OBJECTIVE: Microparticles (MP) are small extracellular vesicles present in body fluids. MP originate from different cellular lineages, principally from platelets in blood, and may expose phosphatidylserine (PS). In systemic lupus erythematosus (SLE), MP harbor immunoglobulin G (IgG), thereby forming MP-containing immune complexes (mpIC). We aimed to verify an association between SLE disease activity, damage, and surrogate markers of atherosclerosis and MP harboring IgG, taking into account the platelet origin and PS exposure of MP. METHODS: MP expressing surface IgG, platelet antigen (CD41+), and PS were quantified using flow cytometry in plasma of 191 women with SLE. Carotid ultrasounds (US) were available in 113 patients. Spearman correlation analysis was used to analyze whether levels of MP were associated with the following outcomes: SLE Disease Activity Index 2000 (SLEDAI-2K), Systemic Lupus International Collaborating Clinics/American College of Rheumatology Damage Index (SDI), and carotid US plaques and intima-media thickness (CIMT) as surrogates for vascular damage. RESULTS: We found CD41+ MP harboring IgG present in SLE. A positive correlation was found between SLEDAI-2K and levels of CD41+ MP harboring IgG and exposing (p = 0.027) and non-exposing PS (p = 0.001). Conversely, SDI (p = 0.024) and CIMT (p = 0.016) correlated with concentrations of CD41- MP harboring IgG and exposing PS. Associations were independent of low-density lipoprotein cholesterol level, body mass index, and antimalarial drug use. CONCLUSION: Different subtypes of mpIC are produced in SLE and are associated with distinct clinical characteristics such as disease activity and vascular damage. The assessment of MP subtypes might serve for the design of predictive markers of disease activity and vascular damage in patients.


Subject(s)
Carotid Intima-Media Thickness , Cell-Derived Microparticles/immunology , Immunoglobulin G/immunology , Lupus Erythematosus, Systemic/immunology , Plaque, Atherosclerotic/diagnostic imaging , Adult , Carotid Arteries/diagnostic imaging , Female , Humans , Lupus Erythematosus, Systemic/diagnostic imaging , Male , Middle Aged , Severity of Illness Index , Ultrasonography
SELECTION OF CITATIONS
SEARCH DETAIL