Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters











Publication year range
1.
Arthritis Rheumatol ; 74(7): 1132-1138, 2022 07.
Article in English | MEDLINE | ID: mdl-35174669

ABSTRACT

OBJECTIVE: While endothelial dysfunction has been implicated in the widespread thromboinflammatory complications of COVID-19, the upstream mediators of endotheliopathy remain, for the most part, unknown. This study was undertaken to identify circulating factors contributing to endothelial cell activation and dysfunction in COVID-19. METHODS: Human endothelial cells were cultured in the presence of serum or plasma from 244 patients hospitalized with COVID-19 and plasma from 100 patients with non-COVID-19-related sepsis. Cell adhesion molecules (E-selectin, vascular cell adhesion molecule 1, and intercellular adhesion molecule 1 [ICAM-1]) were quantified using in-cell enzyme-linked immunosorbent assay. RESULTS: Serum and plasma from COVID-19 patients increased surface expression of cell adhesion molecules. Furthermore, levels of soluble ICAM-1 and E-selectin were elevated in patient serum and correlated with disease severity. The presence of circulating antiphospholipid antibodies was a strong marker of the ability of COVID-19 serum to activate endothelium. Depletion of total IgG from antiphospholipid antibody-positive serum markedly reduced the up-regulation of cell adhesion molecules. Conversely, supplementation of control serum with patient IgG was sufficient to trigger endothelial activation. CONCLUSION: These data are the first to indicate that some COVID-19 patients have potentially diverse antibodies that drive endotheliopathy, providing important context regarding thromboinflammatory effects of autoantibodies in severe COVID-19.


Subject(s)
Antibodies, Antiphospholipid , COVID-19 , Endothelial Cells , Antibodies, Antiphospholipid/immunology , COVID-19/immunology , Cell Adhesion Molecules/metabolism , E-Selectin , Endothelial Cells/metabolism , Endothelium, Vascular , Humans , Immunoglobulin G/metabolism , Intercellular Adhesion Molecule-1/metabolism , Vascular Cell Adhesion Molecule-1/metabolism
2.
medRxiv ; 2022 Feb 02.
Article in English | MEDLINE | ID: mdl-33501469

ABSTRACT

OBJECTIVE: While endothelial dysfunction has been implicated in the widespread thrombo-inflammatory complications of coronavirus disease-19 ( COVID-19 ), the upstream mediators of endotheliopathy remain for the most part cryptic. Our aim was to identify circulating factors contributing to endothelial cell activation and dysfunction in COVID-19. METHODS: Human endothelial cells were cultured in the presence of serum or plasma from 244 patients hospitalized with COVID-19 and plasma from 100 patients with non-COVID sepsis. Cell adhesion molecules (E-selectin, VCAM-1, and ICAM-1) were quantified by in-cell ELISA. RESULTS: Serum and plasma from patients with COVID-19 increased surface expression of cell adhesion molecules. Furthermore, levels of soluble ICAM-1 and E-selectin were elevated in patient serum and tracked with disease severity. The presence of circulating antiphospholipid antibodies was a strong marker of the ability of COVID-19 serum to activate endothelium. Depletion of total IgG from antiphospholipid antibody-positive serum markedly restrained upregulation of cell adhesion molecules. Conversely, supplementation of control serum with patient IgG was sufficient to trigger endothelial activation. CONCLUSION: These data are the first to suggest that some patients with COVID-19 have potentially diverse antibodies that drive endotheliopathy, adding important context regarding thrombo-inflammatory effects of autoantibodies in severe COVID-19.

3.
Arthritis Rheumatol ; 74(5): 902-907, 2022 05.
Article in English | MEDLINE | ID: mdl-34725956

ABSTRACT

OBJECTIVE: Defibrotide is a heterogenous mixture of polyanionic oligonucleotides currently approved for treatment of transplant-associated venoocclusive disease. While defibrotide has a known role in limiting endothelial cell activation, some studies have also demonstrated anti-leukocyte properties. In a recent study, we found that neutrophil extracellular traps (NETs) play a role in the thrombotic complications of antiphospholipid syndrome (APS). In the present study, we investigated the hypothesis that defibrotide might act to mitigate APS-relevant NET formation in vitro and in mouse models. METHODS: We used in vitro assays and a mouse model to determine the mechanisms by which defibrotide inhibits NET formation and venous thrombosis in APS. RESULTS: At doses ranging from 1 to 10 µg/ml, defibrotide significantly suppressed NET formation from control neutrophils stimulated with IgG isolated from patients with APS. Defibrotide increased levels of intracellular cyclic AMP in neutrophils, and its suppressive effects on NET formation were mitigated by blocking adenosine A2A receptor or by inhibiting the cyclic AMP-dependent kinase protein kinase A. Defibrotide at doses ranging from 15 to 150 mg/kg/day inhibited NET formation and venous thrombosis in a model of antiphospholipid antibody-accelerated thrombosis-an effect that was reduced in adenosine A2A receptor-knockout mice. CONCLUSION: This study is the first to demonstrate mechanisms by which defibrotide counteracts neutrophil-mediated thrombotic inflammation inherent to APS.


Subject(s)
Antiphospholipid Syndrome , Extracellular Traps , Thrombosis , Venous Thrombosis , Animals , Antibodies, Antiphospholipid , Antiphospholipid Syndrome/drug therapy , Antiphospholipid Syndrome/metabolism , Cyclic AMP/metabolism , Disease Models, Animal , Extracellular Traps/metabolism , Humans , Mice , Neutrophils/metabolism , Polydeoxyribonucleotides , Receptor, Adenosine A2A , Thrombosis/etiology , Thrombosis/prevention & control
4.
JCI Insight ; 6(17)2021 09 08.
Article in English | MEDLINE | ID: mdl-34264868

ABSTRACT

Neutrophil-mediated activation and injury of the endothelium play roles in the pathogenesis of diverse disease states ranging from autoimmunity to cancer to COVID-19. Neutralization of cationic proteins (such as neutrophil extracellular trap-derived [NET-derived] histones) with polyanionic compounds has been suggested as a potential strategy for protecting the endothelium from such insults. Here, we report that the US Food and Drug Administration-approved polyanionic agent defibrotide (a pleiotropic mixture of oligonucleotides) directly engages histones and thereby blocks their pathological effects on endothelium. In vitro, defibrotide counteracted endothelial cell activation and pyroptosis-mediated cell death, whether triggered by purified NETs or recombinant histone H4. In vivo, defibrotide stabilized the endothelium and protected against histone-accelerated inferior vena cava thrombosis in mice. Mechanistically, defibrotide demonstrated direct and tight binding to histone H4 as detected by both electrophoretic mobility shift assay and surface plasmon resonance. Taken together, these data provide insights into the potential role of polyanionic compounds in protecting the endothelium from thromboinflammation with potential implications for myriad NET- and histone-accelerated disease states.


Subject(s)
Fibrinolytic Agents/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Polydeoxyribonucleotides/pharmacology , Thrombosis/drug therapy , Animals , Extracellular Traps/drug effects , Extracellular Traps/metabolism , Fibrinolytic Agents/therapeutic use , Histones/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Polydeoxyribonucleotides/therapeutic use , Pyroptosis
5.
Int J Mol Sci ; 22(5)2021 Mar 04.
Article in English | MEDLINE | ID: mdl-33806694

ABSTRACT

Antiphospholipid syndrome (APS) is a leading acquired cause of thrombotic events, with a notable tendency to promote thrombosis in vascular beds of all sizes, including both arterial and venous circuits. While pathogenic antiphospholipid antibodies circulate at relatively stable levels in blood, thrombosis tends to manifest as discrete and acute events, suggesting the requirement for a "second hit." While this two-hit model is generally accepted, much remains to be learned about exactly how antiphospholipid antibodies predispose to thrombosis in vivo and exactly how this predisposition interacts with the second hit. To this end, investigators have turned to animal models. Numerous approaches for modeling APS in animals have been described to date, each with potential advantages and disadvantages. This review will attempt to describe the most common APS models employed so far while discussing some pros and cons of each. Mechanisms of thrombotic APS that have thus far been explored in animal models will also be briefly addressed.


Subject(s)
Antiphospholipid Syndrome/pathology , Thrombosis/pathology , Animals , Antibodies, Antiphospholipid/blood , Antiphospholipid Syndrome/blood , Humans , Models, Animal , Thrombosis/blood
6.
medRxiv ; 2021 Jul 15.
Article in English | MEDLINE | ID: mdl-33655266

ABSTRACT

Neutrophil-mediated activation and injury of the endothelium play a role in the pathogenesis of diverse disease states ranging from autoimmunity to cancer to COVID-19. Neutralization of cationic proteins (such as neutrophil extracellular trap/NET-derived histones) with polyanionic compounds has been suggested as a potential strategy for protecting the endothelium from such insults. Here, we report that the FDA-approved polyanionic agent defibrotide (a pleiotropic mixture of oligonucleotides) directly engages histones and thereby blocks their pathological effects on endothelium. In vitro , defibrotide counteracted endothelial cell activation and pyroptosis-mediated cell death, whether triggered by purified NETs or recombinant histone H4. In vivo , defibrotide stabilized the endothelium and protected against histone-accelerated inferior vena cava thrombosis in mice. Mechanistically, defibrotide demonstrated direct and tight binding to histone H4 as detected by both electrophoretic mobility shift assay and surface plasmon resonance. Taken together, these data provide insights into the potential role of polyanionic compounds in protecting the endothelium from thromboinflammation with potential implications for myriad NET- and histone-accelerated disease states.

7.
JCI Insight ; 6(3)2021 02 08.
Article in English | MEDLINE | ID: mdl-33373329

ABSTRACT

Ginger is known to have antiinflammatory and antioxidative effects and has traditionally been used as an herbal supplement in the treatment of various chronic diseases. Here, we report antineutrophil properties of 6-gingerol, the most abundant bioactive compound of ginger root, in models of lupus and antiphospholipid syndrome (APS). Specifically, we demonstrate that 6-gingerol attenuates neutrophil extracellular trap (NET) release in response to lupus- and APS-relevant stimuli through a mechanism that is at least partially dependent on inhibition of phosphodiesterases. At the same time, administration of 6-gingerol to mice reduces NET release in various models of lupus and APS, while also improving other disease-relevant endpoints, such as autoantibody formation and large-vein thrombosis. In summary, this study is the first to our knowledge to demonstrate a protective role for ginger-derived compounds in the context of lupus. Importantly, it provides a potential mechanism for these effects via phosphodiesterase inhibition and attenuation of neutrophil hyperactivity.


Subject(s)
Catechols/pharmacology , Fatty Alcohols/pharmacology , Lupus Erythematosus, Systemic/drug therapy , Lupus Erythematosus, Systemic/immunology , Neutrophils/drug effects , Neutrophils/immunology , Animals , Antibodies, Antiphospholipid/biosynthesis , Antiphospholipid Syndrome/drug therapy , Antiphospholipid Syndrome/immunology , Antiphospholipid Syndrome/metabolism , Catechols/blood , Catechols/pharmacokinetics , Disease Models, Animal , Extracellular Traps/drug effects , Extracellular Traps/immunology , Fatty Alcohols/blood , Fatty Alcohols/pharmacokinetics , Female , Humans , In Vitro Techniques , Lupus Erythematosus, Systemic/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neutrophils/metabolism , Phosphodiesterase 4 Inhibitors/pharmacology , Phosphodiesterase Inhibitors/blood , Phosphodiesterase Inhibitors/pharmacokinetics , Phosphodiesterase Inhibitors/pharmacology , Phytotherapy , Reactive Oxygen Species/metabolism , Venous Thrombosis/drug therapy , Venous Thrombosis/pathology
8.
Sci Transl Med ; 12(570)2020 11 18.
Article in English | MEDLINE | ID: mdl-33139519

ABSTRACT

Patients with COVID-19 are at high risk for thrombotic arterial and venous occlusions. Lung histopathology often reveals fibrin-based blockages in the small blood vessels of patients who succumb to the disease. Antiphospholipid syndrome is an acquired and potentially life-threatening thrombophilia in which patients develop pathogenic autoantibodies targeting phospholipids and phospholipid-binding proteins (aPL antibodies). Case series have recently detected aPL antibodies in patients with COVID-19. Here, we measured eight types of aPL antibodies in serum samples from 172 patients hospitalized with COVID-19. These aPL antibodies included anticardiolipin IgG, IgM, and IgA; anti-ß2 glycoprotein I IgG, IgM, and IgA; and anti-phosphatidylserine/prothrombin (aPS/PT) IgG and IgM. We detected aPS/PT IgG in 24% of serum samples, anticardiolipin IgM in 23% of samples, and aPS/PT IgM in 18% of samples. Antiphospholipid autoantibodies were present in 52% of serum samples using the manufacturer's threshold and in 30% using a more stringent cutoff (≥40 ELISA-specific units). Higher titers of aPL antibodies were associated with neutrophil hyperactivity, including the release of neutrophil extracellular traps (NETs), higher platelet counts, more severe respiratory disease, and lower clinical estimated glomerular filtration rate. Similar to IgG from patients with antiphospholipid syndrome, IgG fractions isolated from patients with COVID-19 promoted NET release from neutrophils isolated from healthy individuals. Furthermore, injection of IgG purified from COVID-19 patient serum into mice accelerated venous thrombosis in two mouse models. These findings suggest that half of patients hospitalized with COVID-19 become at least transiently positive for aPL antibodies and that these autoantibodies are potentially pathogenic.


Subject(s)
Antibodies, Antiphospholipid/blood , COVID-19/immunology , Venous Thrombosis/etiology , Adult , Aged , Aged, 80 and over , Animals , Antibodies, Antiphospholipid/administration & dosage , Antiphospholipid Syndrome/blood , Antiphospholipid Syndrome/etiology , Antiphospholipid Syndrome/immunology , COVID-19/blood , COVID-19/complications , Cohort Studies , Cross-Sectional Studies , Disease Models, Animal , Extracellular Traps/metabolism , Female , Humans , Immunoglobulin G/administration & dosage , Immunoglobulin G/blood , Male , Mice , Mice, Inbred C57BL , Middle Aged , Neutrophils/immunology , Neutrophils/metabolism , Thrombophilia/blood , Thrombophilia/etiology , Thrombophilia/immunology , Translational Research, Biomedical , Venous Thrombosis/blood , Venous Thrombosis/immunology
9.
medRxiv ; 2020 Sep 15.
Article in English | MEDLINE | ID: mdl-32587992

ABSTRACT

Patients with coronavirus disease 19 (COVID-19) are at high risk for thrombotic arterial and venous occlusions. At the same time, lung histopathology often reveals fibrin-based occlusion in the small vessels of patients who succumb to the disease. Antiphospholipid syndrome (APS) is an acquired and potentially life-threatening thrombophilia in which patients develop pathogenic autoantibodies (aPL) targeting phospholipids and phospholipid-binding proteins. Case series have recently detected aPL in patients with COVID-19. Here, we measured eight types of aPL [anticardiolipin IgG/IgM/IgA, anti-beta-2 glycoprotein I IgG/IgM/IgA, and anti- phosphatidylserine/prothrombin (aPS/PT) IgG/IgM] in the sera of 172 patients hospitalized with COVID-19. We detected aPS/PT IgG in 24%, anticardiolipin IgM in 23%, and aPS/PT IgM in 18%. Any aPL was present in 52% of patients using the manufacturer's threshold and in 30% using a more stringent cutoff (≥40 units). Higher levels of aPL were associated with neutrophil hyperactivity (including the release of neutrophil extracellular traps/NETs), higher platelet count, more severe respiratory disease, and lower glomerular filtration rate. Similar to patients with longstanding APS, IgG fractions isolated from patients with COVID-19 promoted NET release from control neutrophils. Furthermore, injection of these COVID-19 IgG fractions into mice accelerated venous thrombosis. Taken together, these studies suggest that a significant percentage of patients with COVID-19 become at least transiently positive for aPL and that these aPL are potentially pathogenic.

10.
Nat Commun ; 10(1): 1916, 2019 04 23.
Article in English | MEDLINE | ID: mdl-31015489

ABSTRACT

Potentiation of neutrophil extracellular trap (NET) release is one mechanism by which antiphospholipid antibodies (aPL Abs) effect thrombotic events in patients with antiphospholipid syndrome (APS). Surface adenosine receptors trigger cyclic AMP (cAMP) formation in neutrophils, and this mechanism has been proposed to regulate NETosis in some contexts. Here we report that selective agonism of the adenosine A2A receptor (CGS21680) suppresses aPL Ab-mediated NETosis in protein kinase A-dependent fashion. CGS21680 also reduces thrombosis in the inferior vena cavae of both control mice and mice administered aPL Abs. The antithrombotic medication dipyridamole is known to potentiate adenosine signaling by increasing extracellular concentrations of adenosine and interfering with the breakdown of cAMP. Like CGS21680, dipyridamole suppresses aPL Ab-mediated NETosis via the adenosine A2A receptor and mitigates venous thrombosis in mice. In summary, these data suggest an anti-inflammatory therapeutic paradigm in APS, which may extend to thrombotic disease in the general population.


Subject(s)
Adenosine A2 Receptor Agonists/pharmacology , Adenosine/analogs & derivatives , Antiphospholipid Syndrome/drug therapy , Extracellular Traps/drug effects , Neutrophils/drug effects , Phenethylamines/pharmacology , Venous Thrombosis/drug therapy , Adenosine/immunology , Adenosine/metabolism , Adenosine/pharmacology , Animals , Antibodies, Antiphospholipid/blood , Antiphospholipid Syndrome/genetics , Antiphospholipid Syndrome/immunology , Antiphospholipid Syndrome/pathology , Cyclic AMP/immunology , Cyclic AMP/metabolism , Dipyridamole/pharmacology , Disease Models, Animal , Extracellular Traps/immunology , Extracellular Traps/metabolism , Fibrinolytic Agents/pharmacology , Gene Expression Regulation , Humans , Immunoglobulin G/blood , Male , Mice , Mice, Inbred C57BL , Neutrophils/immunology , Receptor, Adenosine A2A/genetics , Receptor, Adenosine A2A/immunology , Signal Transduction , Vena Cava, Inferior/drug effects , Vena Cava, Inferior/immunology , Vena Cava, Inferior/metabolism , Venous Thrombosis/genetics , Venous Thrombosis/immunology , Venous Thrombosis/pathology
11.
JCI Insight ; 2(18)2017 09 21.
Article in English | MEDLINE | ID: mdl-28931754

ABSTRACT

Antiphospholipid antibodies, present in one-third of lupus patients, increase the risk of thrombosis. We recently reported a key role for neutrophils - neutrophil extracellular traps (NETs), in particular - in the thrombotic events that define antiphospholipid syndrome (APS). To further elucidate the role of neutrophils in APS, we performed a comprehensive transcriptome analysis of neutrophils isolated from patients with primary APS. Moreover, APS-associated venous thrombosis was modeled by treating mice with IgG prepared from APS patients, followed by partial restriction of blood flow through the inferior vena cava. In patients, APS neutrophils demonstrated a proinflammatory signature with overexpression of genes relevant to IFN signaling, cellular defense, and intercellular adhesion. For in vivo studies, we focused on P-selectin glycoprotein ligand-1 (PSGL-1), a key adhesion molecule overexpressed in APS neutrophils. The introduction of APS IgG (as compared with control IgG) markedly potentiated thrombosis in WT mice, but not PSGL-1-KOs. PSGL-1 deficiency was also associated with reduced leukocyte vessel wall adhesion and NET formation. The thrombosis phenotype was restored in PSGL-1-deficient mice by infusion of WT neutrophils, while an anti-PSGL-1 monoclonal antibody inhibited APS IgG-mediated thrombosis in WT mice. PSGL-1 represents a potential therapeutic target in APS.


Subject(s)
Antiphospholipid Syndrome/immunology , Extracellular Traps/immunology , Neutrophils/immunology , Animals , Antibodies, Antiphospholipid/immunology , Antiphospholipid Syndrome/genetics , Humans , Immunoglobulin G/administration & dosage , Immunoglobulin G/immunology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mice , Mice, Knockout , Transcriptome , Up-Regulation
12.
Ann Rheum Dis ; 76(2): 450-457, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27432357

ABSTRACT

OBJECTIVES: Patients with antiphospholipid syndrome (APS) are at risk for subclinical endothelial injury, as well as accelerated atherosclerosis. In the related disease systemic lupus erythematosus, there is a well-established defect in circulating endothelial progenitors, which leads to an accrual of endothelial damage over time. This defect has been at least partially attributed to exaggerated expression of type I interferons (IFNs). We sought to determine whether these pathways are important in APS. METHODS: We studied 68 patients with primary APS. Endothelial progenitors were assessed by flow cytometry and functional assay. Type I IFN activity was determined by a well-accepted bioassay, while peripheral blood mononuclear cells were scored for expression of IFN-responsive genes. RESULTS: Endothelial progenitors from patients with APS demonstrated a marked defect in the ability to differentiate into endothelial cells, a phenotype which could be mimicked by treating control progenitors with APS sera. Elevated type I IFN activity was detected in the circulation of patients with APS (a finding that was then replicated in an independent cohort). While IgG depletion from APS sera did not rescue endothelial progenitor function, the dysfunction was successfully reversed by a type I IFN receptor-neutralising antibody. CONCLUSIONS: We describe, for the first time to our knowledge, an IFN signature in primary APS and show that this promotes impaired endothelial progenitor function. This work opens the door to novel approaches that may mitigate vascular damage in APS, such as anti-IFN drugs.


Subject(s)
Antiphospholipid Syndrome/immunology , Cell Differentiation/physiology , Endothelial Progenitor Cells/physiology , Interferon-alpha/immunology , Leukocytes, Mononuclear/immunology , Adult , Aged , Antibodies, Neutralizing/pharmacology , Antiphospholipid Syndrome/physiopathology , Case-Control Studies , Cell Differentiation/drug effects , Endothelial Progenitor Cells/drug effects , Female , Flow Cytometry , Humans , Interferon Type I/immunology , Male , Middle Aged , Receptor, Interferon alpha-beta/antagonists & inhibitors , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL