Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 190
Filter
2.
J Lipid Res ; 65(1): 100483, 2024 01.
Article in English | MEDLINE | ID: mdl-38101620

ABSTRACT

Apolipoprotein H (APOH) downregulation can cause hepatic steatosis and gut microbiota dysbiosis. However, the mechanism by which APOH-regulated lipid metabolism contributes to metabolic dysfunction-associated steatotic liver disease (MASLD) remains undetermined. Herein, we aim to explore the regulatory effect of APOH, mediated through various pathways, on metabolic homeostasis and MASLD pathogenesis. We analyzed serum marker levels, liver histopathology, and cholesterol metabolism-related gene expression in global ApoH-/- C57BL/6 male mice. We used RNA sequencing and metabolomic techniques to investigate the association between liver metabolism and bacterial composition. Fifty-two differentially expressed genes were identified between ApoH-/- and WT mice. The mRNA levels of de novo lipogenesis genes were highly upregulated in ApoH-/- mice than in WT mice. Fatty acid, glycerophospholipid, sterol lipid, and triglyceride levels were elevated, while hyodeoxycholic acid levels were significantly reduced in the liver tissues of ApoH-/- mice than in those of WT mice. Microbial beta diversity was lower in ApoH-/- mice than in WT mice, and gut microbiota metabolic functions were activated in ApoH-/- mice. Moreover, ApoH transcripts were downregulated in patients with MASLD, and APOH-related differential genes were enriched in lipid metabolism. Open-source transcript-level data from human metabolic dysfunction-associated steatohepatitis livers reinforced a significant association between metabolic dysfunction-associated steatohepatitis and APOH downregulation. In conclusion, our studies demonstrated that APOH downregulation aggravates fatty liver and induces gut microbiota dysbiosis by dysregulating bile acids. Our findings offer a novel perspective on APOH-mediated lipid metabolic dysbiosis and provide a valuable framework for deciphering the role of APOH in fatty liver disease.


Subject(s)
Gastrointestinal Microbiome , Non-alcoholic Fatty Liver Disease , Humans , Male , Mice , Animals , Lipid Metabolism/genetics , beta 2-Glycoprotein I/genetics , beta 2-Glycoprotein I/metabolism , beta 2-Glycoprotein I/pharmacology , Down-Regulation , Dysbiosis/metabolism , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/metabolism , Liver/metabolism , Fatty Acids/metabolism
3.
Anticancer Res ; 43(8): 3455-3462, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37500148

ABSTRACT

BACKGROUND/AIM: Beta2-glycoprotein I (ß2-GPI) is a plasma glycoprotein, which has been implicated in a variety of physiological functions. However, the connection between ß2-GPI and breast cancer is mostly unknown. Breast cancer is a malignant tumor that severely impairs women's health worldwide. The aim of the study was to investigate the role of ß2-GPI in tumor cells of breast cancer patients and its correlation with tumor prognosis. MATERIALS AND METHODS: A total of 125 female patients diagnosed with breast cancer were enrolled in the study. The expression of ß2-GPI in resected breast tissues was determined by immunohistochemistry (IHC) and correlated with clinicopathological variables by the Chi-squared test. The prognostic value of ß2-GPI for overall survival (OS) and disease-free survival (DFS) was determined by Kaplan-Meier estimates and the significance of differences was evaluated by the log-rank test. RESULTS: ß2-GPI staining was predominantly observed in tumor cells of breast cancer patients and significantly correlated with tumor stage and lymph node metastasis of breast cancer. High ß2-GPI expression was significantly correlated with better OS and DFS. Moreover, DFS was found to be significantly better in patients with higher ß2-GPI expression, especially those in the early tumor stage groups. CONCLUSION: High ß2-GPI expression levels in tumor cells of breast cancer patients were independent factors predicting a better OS and DFS. ß2-GPI activation in high-risk patients may be a potential strategy for reducing breast cancer progression.


Subject(s)
Breast Neoplasms , Mammary Neoplasms, Animal , Humans , Female , Animals , Breast Neoplasms/pathology , beta 2-Glycoprotein I/metabolism , Neoplasm Staging , Biomarkers, Tumor/metabolism , Prognosis , Mammary Neoplasms, Animal/pathology , Carrier Proteins , Disease-Free Survival , Kaplan-Meier Estimate
4.
J Chin Med Assoc ; 85(10): 993-999, 2022 10 01.
Article in English | MEDLINE | ID: mdl-36206526

ABSTRACT

BACKGROUND: Although the immune systems of patients with systemic lupus erythematosus (SLE) are affected by both personal characteristics and environmental factors, the effects of parabens on patients with SLE have not been well studied. We investigated the indirect effects of four parabens-methylparaben (MP), ethylparaben (EP), propylparaben (n-PrP), and butylparaben (n-BuP)-on several immunological markers. METHODS: We assessed the serum levels of MP, EP, n-PrP, and n-BuP in 25 SLE patients and correlated the concentration of each paraben with available clinical and laboratory markers, including intracellular markers of antiviral immunity and apoptosis. RESULTS: The expression of aryl hydrocarbon receptor (AhR) was significantly negatively correlated with n-PrP levels (p = 0.03, r = -0.434). In monocytes, APO2.7 was significantly positively correlated with n-BuP levels (p = 0.019, r = 0.467). Glutathione levels were significantly negatively correlated with n-BuP levels (p = 0.019, r = -0.518). Anti- ß2 glycoprotein I IgM was significantly positively correlated with both MP (p = 0.011, r = 0.585) and EP levels (p = 0.032, r = 0.506). Anti-cardiolipin IgA was significantly positively correlated with both MP (p = 0.038, r = 0.493) and n-PrP levels (p = 0.031, r = 0.508). On CD8 T cells, the early apoptotic marker annexin V was significantly negatively correlated with both MP (p < 0.05, r = -0.541) and n-BuP levels (p = 0.02, r = -0.616), and L-selectin was significantly positively correlated with both MP (p < 0.05, r = 0.47) and n-PrP levels (p = 0.02, r = 0.556). CONCLUSION: Our findings suggest that higher parabens levels were associated with lower AhR expression in leukocytes, increased monocyte apoptosis, lower serum glutathione levels, reduced annexin V expression on CD8 T cells, and higher L-selectin levels on leukocytes.


Subject(s)
Lupus Erythematosus, Systemic , Parabens , Annexin A5 , Antiviral Agents , Biomarkers , Glutathione/metabolism , Humans , Immunoglobulin A/metabolism , Immunoglobulin M , L-Selectin/metabolism , Parabens/analysis , Parabens/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Taiwan , beta 2-Glycoprotein I/metabolism
5.
Front Immunol ; 13: 1076167, 2022.
Article in English | MEDLINE | ID: mdl-36700193

ABSTRACT

Antiphospholipid syndrome (APS) is a systemic autoimmune disorder characterized by recurrent vascular thrombosis and miscarriages in the absence of known causes. Antibodies against phospholipid-binding proteins (aPL) are pathogenic players in both clotting and pregnancy APS manifestations. There is sound evidence that antibodies specific for beta2 glycoprotein I (ß2GPI) trigger thrombotic and pregnancy complications by interacting with the molecule on the membranes of different cell types of the coagulation cascade, and in placenta tissues. In addition to the humoral response against ß2GPI, both peripheral and tissue CD4+ ß2GPI-specific T cells have been reported in primary APS as well as in systemic lupus erythematosus (SLE)-associated APS. While adaptive immunity plays a clear role in APS, it is still debated whether innate immunity is involved as well. Acute systemic inflammation does not seem to be present in the syndrome, however, there is sound evidence that complement activation is crucial in animal models and can be found also in patients. Furthermore, neutrophil extracellular traps (NETs) have been documented in arterial and venous thrombi with different etiology, including clots in APS models. Keeping in mind that ß2GPI is a pleiotropic glycoprotein, acting as scavenger molecule for infectious agents and apoptotic/damaged body constituents and that self-molecules externalized through NETs formation may become immunogenic autoantigens, we demonstrated ß2GPI on NETs, and its ability to stimulate CD4+ß2GPI-specific T cells. The aim of this review is to elucidate the role of ß2GPI in the cross-talk between the innate and adaptive immunity in APS.


Subject(s)
Antiphospholipid Syndrome , Extracellular Traps , Thrombosis , beta 2-Glycoprotein I , Animals , Female , Pregnancy , Adaptive Immunity , Antibodies, Antiphospholipid , beta 2-Glycoprotein I/metabolism , Extracellular Traps/metabolism , Thrombosis/complications , Immunity, Innate
6.
Anal Bioanal Chem ; 413(30): 7441-7449, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34686894

ABSTRACT

Apo-H is a plasma glycoprotein. Nearly 19% of the molecular weight of this protein is composed of glycans. Up- and down-regulation and structural changes in protein glycans provide diagnostic value for disease detection. Here, an efficient, sensitive, and optimized method is developed for Apo-H N-glycans analysis by MALDI-TOF-MS in positive mode. This bioanalytical method includes sample preparation, sample purification, and detection. An Apo-H enrichment method is developed using standard proteins by anti-Apo-H beads followed by enrichment from plasma samples. SDS-PAGE confirms the Apo-H protein enrichment, which is further verified by LC-MS/MS analysis. The lower ionization efficiency of sialylated glycan hampers their analysis by MALDI-MS. For this, stabilization of sialic acids is done by selective derivatization of carboxyl groups to differentiate between α(2,3)- and α(2,6)-linked sialic acids. Glycans are further purified by HILIC-SPE and analyzed by MALDI-MS. Several branched bi- and tri-antennary glycans with fucosylation and sialylation are identified. The reproducibility of the developed method is tested by analyzing multiple replicates of human plasma, where the same glycans are consistently identified. This method could be applied for the Apo-H glycan profiling of large clinical cohorts for diagnostic purposes.


Subject(s)
N-Acetylneuraminic Acid/chemistry , Polysaccharides/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , beta 2-Glycoprotein I/metabolism , Chromatography, Liquid/methods , Cohort Studies , Electrophoresis, Polyacrylamide Gel , Humans , Reproducibility of Results
7.
J Biol Chem ; 297(2): 100890, 2021 08.
Article in English | MEDLINE | ID: mdl-34197876

ABSTRACT

ß2-glycoprotein I (ß2GPI) is an abundant multidomain plasma protein that plays various roles in the clotting and complement cascades. It is also the main target of antiphospholipid antibodies (aPL) in the acquired coagulopathy known as antiphospholipid syndrome (APS). Previous studies have shown that ß2GPI adopts two interconvertible biochemical conformations, oxidized and reduced, depending on the integrity of the disulfide bonds. However, the precise contribution of the disulfide bonds to ß2GPI structure and function is unknown. Here, we substituted cysteine residues with serine to investigate how the disulfide bonds C32-C60 in domain I (DI) and C288-C326 in domain V (DV) regulate ß2GPI's structure and function. Results of our biophysical and biochemical studies support the hypothesis that the C32-C60 disulfide bond plays a structural role, whereas the disulfide bond C288-C326 is allosteric. We demonstrate that absence of the C288-C326 bond, unlike absence of the C32-C60 bond, diminishes membrane binding without affecting the thermodynamic stability and overall structure of the protein, which remains elongated in solution. We also document that, while absence of the C32-C60 bond directly impairs recognition of ß2GPI by pathogenic anti-DI antibodies, absence of the C288-C326 disulfide bond is sufficient to abolish complex formation in the presence of anionic phospholipids. We conclude that the disulfide bond C288-C326 operates as a molecular switch capable of regulating ß2GPI's physiological functions in a redox-dependent manner. We propose that in APS patients with anti-DI antibodies, selective rupture of the C288-C326 disulfide bond may be a valid strategy to lower the pathogenic potential of aPL.


Subject(s)
Antibodies, Antiphospholipid/immunology , Antiphospholipid Syndrome/immunology , Autoantibodies/immunology , Recombinant Proteins/metabolism , beta 2-Glycoprotein I/metabolism , Allosteric Regulation , Antibodies, Antiphospholipid/blood , Antiphospholipid Syndrome/pathology , Autoantibodies/blood , Cell Line , Crystallography, X-Ray/methods , Humans , Oxidation-Reduction , Protein Domains , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , beta 2-Glycoprotein I/chemistry , beta 2-Glycoprotein I/immunology , beta 2-Glycoprotein I/isolation & purification
8.
J Autoimmun ; 122: 102675, 2021 08.
Article in English | MEDLINE | ID: mdl-34098405

ABSTRACT

Β2-Glycoprotein I (ß2GPI) is an important anti-thrombotic protein and is the major auto-antigen in the antiphospholipid syndrome (APS). The clinical relevance of nitrosative stress in post translational modification of ß2GPI was examined.The effects of nitrated (n)ß2GPI on its anti-thrombotic properties and its plasma levels in primary and secondary APS were determined with appropriate clinical control groups. ß2-glycoprotein I was nitrated at tyrosines 218, 275 and 309. ß2-glycoprotein I binds to lipid peroxidation modified products through Domains IV and V. Nitrated ß2GPI loses this binding (p < 0.05) and had diminished activity in inhibiting platelet adhesion to vWF under high shear flow (p < 0.01). Levels of nß2GPI were increased in patients with primary APS compared to patients with either secondary APS (p < 0.05), autoimmune disease without APS (p < 0.05) or non-autoimmune patients with arterial thrombosis (p < 0.01) and healthy individuals (p < 0.05).In conclusion tyrosine nitration of plasma ß2GPI is demonstrated and has important implications with regards to the pathophysiology of platelet mediated thrombosis in APS. Elevated plasma levels of nß2GPI in primary APS may be a risk factor for thrombosis warranting further investigation.


Subject(s)
Antiphospholipid Syndrome/complications , Thrombosis/immunology , beta 2-Glycoprotein I/immunology , Antiphospholipid Syndrome/blood , Antiphospholipid Syndrome/immunology , Case-Control Studies , Healthy Volunteers , Humans , Lipid Peroxidation , Nitrates/metabolism , Platelet Aggregation/immunology , Protein Processing, Post-Translational/immunology , Risk Factors , Thrombosis/blood , beta 2-Glycoprotein I/blood , beta 2-Glycoprotein I/metabolism
9.
Front Immunol ; 12: 604222, 2021.
Article in English | MEDLINE | ID: mdl-34040601

ABSTRACT

Background: Antiphospholipid syndrome (APS) is a systemic autoimmune disease that can lead to thrombosis and/or pregnancy complications. Exosomes, membrane-encapsulated vesicles that are released into the extracellular environment by many types of cells, can carry signals to recipient cells to affect angiogenesis, apoptosis, and inflammation. There is increasing evidence suggesting that exosomes play critical roles in pregnancy. However, the contribution of exosomes to APS is still unknown. Methods: Peripheral plasma was collected from healthy early pregnancy patients (NC-exos) and early pregnancy patients with APS (APS-exos) for exosome extraction and characterization. The effect of exosomes from different sources on pregnancy outcomes was determined by establishing a mouse pregnancy model. Following the coincubation of exosomes and human umbilical vein endothelial cells (HUVECs), functional tests examined the features of APS-exos. The APS-exos and NC-exos were analyzed by quantitative proteomics of whole protein tandem mass tag (TMT) markers to explore the different compositions and identify key proteins. After incubation with HUVECs, functional tests investigated the characteristics of key exosomal proteins. Western blot analysis was used to identify the key pathways. Results: In the mouse model, APS-exos caused an APS-like birth outcome. In vitro experiments showed that APS-exos inhibited the migration and tube formation of HUVECs. Quantitative proteomics analysis identified 27 upregulated proteins and 9 downregulated proteins in APS-exos versus NC-exos. We hypothesized that apolipoprotein H (APOH) may be a core protein, and the analysis of clinical samples was consistent with finding from the proteomic TMT analysis. APOH-exos led to APS-like birth outcomes. APOH-exos directly enter HUVECs and may play a role through the phospho-extracellular signal-regulated kinase pathway. Conclusions: Our study suggests that both APS-exos and APOH-exos impair vascular development and lead to pregnancy complications. APOH-exos may be key actors in the pathogenesis of APS. This study provides new insights into the pathogenesis of APS and potential new targets for therapeutic intervention.


Subject(s)
Antiphospholipid Syndrome/etiology , Antiphospholipid Syndrome/metabolism , Disease Susceptibility , Exosomes/metabolism , beta 2-Glycoprotein I/metabolism , Animals , Antiphospholipid Syndrome/pathology , Biological Transport , Biomarkers , Cells, Cultured , Disease Models, Animal , Female , Gene Expression Profiling , Gene Expression Regulation , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mice , Phenotype , Phosphorylation , Pregnancy , Proteome , Proteomics/methods
10.
Clin Appl Thromb Hemost ; 27: 10760296211010969, 2021.
Article in English | MEDLINE | ID: mdl-33882699

ABSTRACT

Portal venous system thrombosis (PVST), a common complication of liver cirrhosis, is closely associated with thrombophilia. To explore the association of homocysteine (Hcy), anticardiolipin antibody (aCL), and anti-ß2 glycoprotein I antibody (aß2GPI), which are possible thrombophilic factors, with PVST in liver cirrhosis. Overall, 654 non-malignant patients (219 with and 435 without liver cirrhosis) admitted between January 2016 and June 2020 were retrospectively evaluated. Presence of PVST, degree of main portal vein (MPV) thrombosis, and clinically significant PVST were identified. Hcy level, hyperhomocysteinemia (HHcy), aCL positivity, and aß2GPI positivity were compared according to the presence of liver cirrhosis and PVST. Positive aß2GPI was significantly more frequent in patients with liver cirrhosis than those without, but Hcy level and proportions of HHcy and positive aCL were not significantly different between them. PVST could be evaluated in 136 cirrhotic patients. Hcy level [10.57 µmol/L (2.71-56.82) versus 9.97 µmol/L (2.05-53.44); P = 0.796] and proportions of HHcy [4/44 (9.1%) versus 13/81 (16.0%); P = 0.413] and positive aCL [1/23 (4.3%) versus 10/52 (19.2%); P = 0.185] and aß2GPI [9/23 (39.1%) versus 21/52 (40.4%); P = 0.919] were not significantly different between cirrhotic patients with and without PVST. There was still no significant association of Hcy level, HHcy, aCL, or aß2GPI with PVST based on Child-Pugh classification, MPV thrombosis >50%, and clinically significant PVST. Hcy, aCL, and aß2GPI may not be associated with PVST in liver cirrhosis, suggesting that routine screening for Hcy, aCL, and aß2GPI should be unnecessary in such patients.


Subject(s)
Antibodies, Anticardiolipin/metabolism , Homocysteine/metabolism , Liver Cirrhosis/blood , Venous Thrombosis/blood , beta 2-Glycoprotein I/metabolism , Adult , Aged , Aged, 80 and over , Humans , Middle Aged , Young Adult
11.
Sci Rep ; 11(1): 4542, 2021 02 25.
Article in English | MEDLINE | ID: mdl-33633190

ABSTRACT

Beta-2-glycoprotein I (ß2GPI) is a blood protein and the major antigen in the autoimmune disorder antiphospholipid syndrome (APS). ß2GPI exists mainly in closed or open conformations and comprises of 11 disulfides distributed across five domains. The terminal Cys288/Cys326 disulfide bond at domain V has been associated with different cysteine redox states. The role of this disulfide bond in conformational dynamics of this protein has not been investigated so far. Here, we report on the enzymatic driven reduction by thioredoxin-1 (recycled by Tris(2-carboxyethyl)phosphine; TCEP) of ß2GPI. Specific reduction was demonstrated by Western blot and mass spectrometry analyses confirming majority targeting to the fifth domain of ß2GPI. Atomic force microscopy images suggested that reduced ß2GPI shows a slightly higher proportion of open conformation and is more flexible compared to the untreated protein as confirmed by modelling studies. We have determined a strong increase in the binding of pathogenic APS autoantibodies to reduced ß2GPI as demonstrated by ELISA. Our study is relevant for understanding the effect of ß2GPI reduction on the protein structure and its implications for antibody binding in APS patients.


Subject(s)
Autoantibodies/chemistry , Protein Conformation , Protein Folding , Protein Interaction Domains and Motifs , beta 2-Glycoprotein I/chemistry , Autoantibodies/immunology , Cysteine/chemistry , Cysteine/metabolism , Disulfides/chemistry , Immunoglobulin G/chemistry , Immunoglobulin G/immunology , Microscopy, Atomic Force , Models, Molecular , Protein Binding/immunology , Structure-Activity Relationship , beta 2-Glycoprotein I/immunology , beta 2-Glycoprotein I/metabolism
12.
Lupus ; 30(5): 775-784, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33554716

ABSTRACT

INTRODUCTION: Antiphospholipid syndrome (APS) is an autoimmune disorder manifested by thromboembolic events, recurrent spontaneous abortions and elevated titers of circulating antiphospholipid antibodies. In addition, the presence of antiphospholipid antibodies seems to confer a fivefold higher risk for stroke or transient ischemic attack. Although the major antigen of APS is ß2 glycoprotein I, it is now well established that antiphospholipid antibodies are heterogeneous and bind to various targets. Recently, antibodies to Annexin A2 (ANXA2) have been reported in APS. This is of special interest since data indicated ANXA2 as a key player in fibrinolysis. Therefore, in the present study we assessed whether anti-ANXA2 antibodies play a pathological role in thrombosis associated disease. MATERIALS AND METHODS: Mice were induced to produce anti-ANXA2 antibodies by immunization with ANXA2 (iANXA2) and control mice were immunized with adjuvant only. A middle cerebral artery occlusion stroke model was applied to the mice. The outcome of stroke severity was assessed and compared between the two groups. RESULTS: Our results indicate that antibodies to ANXA2 lead to a more severe stroke as demonstrated by a significant larger stroke infarct volume (iANXA2 133.9 ± 3.3 mm3 and control 113.7 ± 7.4 mm3; p = 0.017) and a more severe neurological outcome (iANXA2 2.2 ± 0.2, and control 1.5 ± 0.18; p = 0.03). CONCLUSIONS: This study supports the hypothesis that auto-antibodies to ANXA2 are an independent risk factor for cerebral thrombosis. Consequently, we propose screening for anti-ANXA2 antibodies should be more widely used and patients that exhibit the manifestations of APS should be closely monitored by physicians.


Subject(s)
Annexin A2/immunology , Antiphospholipid Syndrome/immunology , Autoantibodies/immunology , Intracranial Thrombosis/metabolism , Adult , Animals , Annexin A2/administration & dosage , Annexin A2/metabolism , Antibodies, Antiphospholipid/blood , Antiphospholipid Syndrome/complications , Antiphospholipid Syndrome/metabolism , Autoantibodies/metabolism , Autoimmunity/immunology , Disease Models, Animal , Female , Fibrinolysis/immunology , Humans , Infarction, Middle Cerebral Artery/immunology , Infarction, Middle Cerebral Artery/physiopathology , Injections, Subcutaneous , Intracranial Thrombosis/etiology , Ischemic Attack, Transient/immunology , Mice , Mice, Inbred BALB C/immunology , Middle Aged , Risk Factors , Severity of Illness Index , Stroke/immunology , beta 2-Glycoprotein I/metabolism
13.
J Cereb Blood Flow Metab ; 41(8): 2038-2053, 2021 08.
Article in English | MEDLINE | ID: mdl-33444093

ABSTRACT

Beta-2 Glycoprotein I (ß2-GPI) is the main target of anti-phospholipid antibodies (aPL) in the autoimmune anti-phospholipid syndrome, characterized by increased risk of stroke. We here investigated the antibody independent role of ß2-GPI after ischemia/reperfusion, modeled in vivo by transient middle cerebral artery occlusion (tMCAo) in male C57Bl/6J mice; in vitro by subjecting immortalized human brain microvascular endothelial cells (ihBMEC) to 16 h hypoxia and 4 h re-oxygenation. ApoH (coding for ß2-GPI) was upregulated selectively in the liver at 48 h after tMCAo. At the same time ß2-GPI circulating levels increased. ß2-GPI was detectable in brain parenchyma and endothelium at all time points after tMCAo. Parenchymal ß2-GPI recognized apoptotic neurons (positive for annexin V, C3 and TUNEL) cleared by CD68+ brain macrophages. Hypoxic ihBMEC showed increased release of IL-6, over-expression of thrombomodulin and IL-1α after re-oxygenation with ß2-GPI alone. ß2-GPI interacted with mannose-binding lectin in mouse plasma and ihBMEC medium, potentially involved in formation of thrombi. We show for the first time that brain ischemia triggers the hepatic production of ß2-GPI. ß2-GPI is present in the ischemic endothelium, enhancing vascular inflammation, and extravasates binding stressed neurons before their clearance by phagocytosis. Thus ß2-GPI may be a new mediator of brain injury following ischemic stroke.


Subject(s)
Brain Ischemia/pathology , Neurons/metabolism , Vascular System Injuries/pathology , beta 2-Glycoprotein I/metabolism , Animals , Brain/metabolism , Brain/pathology , Brain Ischemia/etiology , Complement System Proteins/metabolism , Disease Models, Animal , Endothelial Cells/cytology , Endothelial Cells/metabolism , Humans , Interleukin-6/metabolism , Liver/metabolism , Liver/pathology , Macrophages/cytology , Macrophages/immunology , Macrophages/metabolism , Male , Mannose-Binding Lectin/metabolism , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Neurons/cytology , Phagocytosis , Protein Binding , Vascular System Injuries/complications , beta 2-Glycoprotein I/blood
14.
J Immunol Methods ; 487: 112877, 2020 12.
Article in English | MEDLINE | ID: mdl-33031793

ABSTRACT

One of the most abundant coagulation proteins is ß2-glycoprotein I (ß2GPI) that is present in humans at a concentration of around 200 mg/L. Its physiological role is only partially understood, but it adopts several different structural forms the majority of which are the open and closed forms. We isolated native (circular) ß2GPI and converted it into an open conformation. The effectiveness of these procedures was assessed by Western blot and negative-staining electron microscopy. We found that in coagulation assays the open form of ß2GPI had a significant prolonging effect on fibrin formation in a dilute prothrombin time test (p < 0.001). In the dilute activated partial thromboplastin time test, both conformations had a significant prolonging effect (p < 0.001) but the open conformation was more effective. In a fluorescent thrombin generation assay both conformations slightly delayed thrombin generation with no significant effect on the quantity of formed thrombin. By using surface plasmon resonance assays, the equilibrium dissociation constants of both the open and closed conformations of ß2GPI showed a similar and strong affinity to isolated anti-ß2GPI autoantibodies (Kd closed ß2GPI = 5.17 × 10-8 M, Kd open ß2GPI = 5.56 × 10-8 M) and the open form had one order of magnitude stronger affinity to heparin (Kd = 0.30 × 10-6 M) compared to the closed conformation (Kd = 3.50 × 10-6 M). The two different forms of ß2GPI have distinct effects in functional tests and in ligand binding, which may considerably affect the intravascular events related to this abundant plasma protein in health and disease.


Subject(s)
Blood Coagulation , beta 2-Glycoprotein I/metabolism , Anticoagulants/pharmacology , Autoantibodies/metabolism , Binding Sites, Antibody , Blood Coagulation/drug effects , Fibrin/metabolism , Heparin/pharmacology , Humans , Ligands , Partial Thromboplastin Time , Protein Conformation , Prothrombin Time , Structure-Activity Relationship , Thrombin/metabolism , beta 2-Glycoprotein I/antagonists & inhibitors , beta 2-Glycoprotein I/chemistry , beta 2-Glycoprotein I/immunology
15.
Cancer Lett ; 495: 66-75, 2020 12 28.
Article in English | MEDLINE | ID: mdl-32891714

ABSTRACT

Melanoma, a form of skin cancer, is one of the most common cancers in young men and women. Tumors require angiogenesis to provide oxygen and nutrients for growth. Pro-angiogenic molecules such as VEGF and anti-angiogenic molecules such as sFlt-1 control angiogenesis. In addition, the serum protein, Beta2 Glycoprotein I (ß2-GPI) induces or inhibits angiogenesis depending on conformation and concentration. ß2-GPI binds to proteins and negatively charged phospholipids on hypoxic endothelial cells present in the tumor microenvironment. We hypothesized that peptides derived from the binding domain of ß2-GPI would regulate angiogenesis and melanoma growth. In vitro analyses determined the peptides reduced endothelial cell migration and sFlt-1 secretion. In a syngeneic, immunocompetent mouse melanoma model, ß2-GPI-derived peptides also reduced melanoma growth in a dose-dependent response with increased sFlt-1 and attenuated vascular markers compared to negative controls. Importantly, administration of peptide with sFlt-1 antibody resulted in tumor growth. These data demonstrate the therapeutic potential of novel ß2-GPI-derived peptides to attenuate tumor growth and endothelial migration is sFlt-1 dependent.


Subject(s)
Endothelial Cells/cytology , Melanoma, Experimental/drug therapy , Peptides/administration & dosage , Vascular Endothelial Growth Factor Receptor-1/metabolism , beta 2-Glycoprotein I/chemistry , Animals , Cell Line, Tumor , Cell Movement/drug effects , Coculture Techniques , Dose-Response Relationship, Drug , Female , Melanoma, Experimental/blood supply , Melanoma, Experimental/metabolism , Mice , Peptides/pharmacology , Phospholipids/metabolism , Protein Domains , Tumor Microenvironment/drug effects , Xenograft Model Antitumor Assays , beta 2-Glycoprotein I/metabolism
16.
J Immunol ; 205(5): 1385-1392, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32759297

ABSTRACT

ß2-Glycoprotein I (ß2-GPI) is an abundant plasma glycoprotein with unknown physiological function and is currently recognized as the main target of antiphospholipid Abs responsible for complement activation and vascular thrombosis in patients with antiphospholipid syndrome (APS). In this study, we provide evidence that mannose-binding lectin (MBL) binds to ß2-GPI in Ca++ and a dose-dependent manner and that this interaction activates complement and promotes complement-dependent thrombin generation. Surprisingly, a significant binding was observed between MBL and isolated domains II and IV of ß2-GPI, whereas the carbohydrate chains, domain I and domain V, were not involved in the interaction, documenting a noncanonical binding mode between MBL and ß2-GPI. Importantly, this interaction may occur on endothelial cells because binding of MBL to ß2-GPI was detected on the surface of HUVECs, and colocalization of MBL with ß2-GPI was observed on the endothelium of a biopsy specimen of a femoral artery from an APS patient. Because ß2-GPI-mediated MBL-dependent thrombin generation was increased after priming the endothelium with TNF-α, our data suggests that this mechanism could play an important yet unrecognized role under physiological conditions and may be upregulated in pathological situations. Moreover, the complement activation and the procoagulant effects of the ß2-GPI/MBL complex may contribute to amplify similar activities of anti-ß2-GPI Abs in APS and possibly act independently of Abs, raising the issue of developing appropriate therapies to avoid recurrences and disability in patients at risk for these clinical conditions.


Subject(s)
Complement Activation/immunology , Mannose-Binding Lectin/metabolism , Thrombin/metabolism , beta 2-Glycoprotein I/metabolism , Antiphospholipid Syndrome/immunology , Antiphospholipid Syndrome/metabolism , Calcium/metabolism , Cell Line , Endothelial Cells/immunology , Endothelial Cells/metabolism , Endothelium/immunology , Endothelium/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Mannose-Binding Lectin/immunology , Protein Binding/immunology , Thrombin/immunology , Thrombosis/immunology , Thrombosis/metabolism , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism , beta 2-Glycoprotein I/immunology
17.
J Autoimmun ; 115: 102527, 2020 12.
Article in English | MEDLINE | ID: mdl-32709480

ABSTRACT

OBJECTIVE: Antiphospholipid syndrome (APS) is an acquired thrombophilia characterized by recurrent thrombosis and/or pregnancy morbidity, in the presence of antibodies to ß2 glycoprotein-I (ß2GPI), prothrombin or Lupus anticoagulant (LA). Anti-ß2GPI antibodies recognize complexes of ß2GPI dimers with CXCL4 chemokine and activate platelets. Thrombospondin 1 (TSP-1) is secreted by platelets and exhibits prothrombotic and proinflammatory properties. Therefore, we investigated its implication in APS. METHODS: Plasma from APS patients (n = 100), Systemic Lupus Erythematosus (SLE) (n = 27) and healthy donors (HD) (n = 50) was analyzed for TSP-1, IL-1ß, IL-17A and free active TGF-ß1 by ELISA. Human Umbilical Vein Endothelial Cells (HUVECs) and HD monocytes were treated with total HD-IgG or anti-ß2GPI, ß2GPI and CXCL4 and CD4+ T-cells were stimulated by monocyte supernatants. TSP-1, IL-1ß, IL-17A TGF-ß1 levels were quantified by ELISA and Real-Time PCR. RESULTS: Higher plasma levels of TSP-1 and TGF-ß1, which positively correlated each other, were observed in APS but not HDs or SLE patients. Patients with arterial thrombotic events or those undergoing a clinical event had the highest TSP-1 levels. These patients also had detectable IL-1ß, IL-17A in their plasma. HD-derived monocytes and HUVECs stimulated with anti-ß2GPI-IgG-ß2GPI-CXCL4 secreted the highest TSP-1 and IL-1ß levels. Supernatants from anti-ß2GPI-ß2GPI-CXCL4 treated monocytes induced IL-17A expression from CD4+ T-cells. Transcript levels followed a similar pattern. CONCLUSIONS: TSP-1 is probably implicated in the pathogenesis of APS. In vitro cell treatments along with high TSP-1 levels in plasma of APS patients suggest that high TSP-1 levels could mark a prothrombotic state and an underlying inflammatory process.


Subject(s)
Antiphospholipid Syndrome/complications , Lupus Erythematosus, Systemic/immunology , Pregnancy Complications, Hematologic/immunology , Thrombosis/immunology , Thrombospondin 1/metabolism , Antiphospholipid Syndrome/blood , Antiphospholipid Syndrome/immunology , Autoantibodies/blood , Autoantibodies/immunology , Blood Platelets/metabolism , Case-Control Studies , Female , Healthy Volunteers , Human Umbilical Vein Endothelial Cells , Humans , Lupus Erythematosus, Systemic/blood , Lupus Erythematosus, Systemic/complications , Male , Platelet Activation/immunology , Platelet Factor 4/metabolism , Pregnancy , Pregnancy Complications, Hematologic/blood , Pregnancy Complications, Hematologic/epidemiology , Thrombosis/blood , Thrombosis/epidemiology , beta 2-Glycoprotein I/immunology , beta 2-Glycoprotein I/metabolism
18.
J Biol Chem ; 295(31): 10794-10806, 2020 07 31.
Article in English | MEDLINE | ID: mdl-32518155

ABSTRACT

ß2-Glycoprotein I (ß2GPI) is an abundant plasma protein displaying phospholipid-binding properties. Because it binds phospholipids, it is a target of antiphospholipid antibodies (aPLs) in antiphospholipid syndrome (APS), a life-threatening autoimmune thrombotic disease. Indeed, aPLs prefer membrane-bound ß2GPI to that in solution. ß2GPI exists in two almost equally populated redox states: oxidized, in which all the disulfide bonds are formed, and reduced, in which one or more disulfide bonds are broken. Furthermore, ß2GPI can adopt multiple conformations (i.e. J-elongated, S-twisted, and O-circular). While strong evidence indicates that the J-form is the structure bound to aPLs, which conformation exists and predominates in solution remains controversial, and so is the conformational pathway leading to the bound state. Here, we report that human recombinant ß2GPI purified under native conditions is oxidized. Moreover, under physiological pH and salt concentrations, this oxidized form adopts a J-elongated, flexible conformation, not circular or twisted, in which the N-terminal domain I (DI) and the C-terminal domain V (DV) are exposed to the solvent. Consistent with this model, binding kinetics and mutagenesis experiments revealed that in solution the J-form interacts with negatively charged liposomes and with MBB2, a monoclonal anti-DI antibody that recapitulates most of the features of pathogenic aPLs. We conclude that the preferential binding of aPLs to phospholipid-bound ß2GPI arises from the ability of its preexisting J-form to accumulate on the membranes, thereby offering an ideal environment for aPL binding. We propose that targeting the J-form of ß2GPI provides a strategy to block pathogenic aPLs in APS.


Subject(s)
Antibodies, Antiphospholipid/chemistry , Antiphospholipid Syndrome , beta 2-Glycoprotein I/chemistry , Animals , Antibodies, Antiphospholipid/metabolism , Cricetinae , HEK293 Cells , Humans , Kinetics , Mutagenesis , Protein Domains , beta 2-Glycoprotein I/metabolism
19.
COPD ; 17(1): 29-33, 2020 02.
Article in English | MEDLINE | ID: mdl-31920121

ABSTRACT

Chronic obstructive pulmonary disease (COPD) is characterised by a progressive pulmonary and systemic inflammation. Acute exacerbations of COPD (AECOPD) are associated with acute inflammation and infection, increase in the rates of morbidity and mortality. Previous proteomic studies have focussed on identifying proteins involved in COPD pathogenesis in samples collected from the lung (e.g. lung tissue biopsies, bronchoalveolar lavage and sputum) but not from blood of patients who experienced AECOPD. In this study, plasma was analysed by two independent quantitative proteomics techniques; isobaric tag for relative and absolute quantitation (iTRAQ) and multiple reaction monitoring (MRM) to identify differential expression of circulating proteins in patients with stable COPD (sCOPD) and AECOPD. Firstly, iTRAQ performed on pooled plasma samples from AECOPD, sCOPD, and healthy non-smoking controls (HC) revealed 15 differentially expressed proteins between the 3 groups. MRM subsequently performed on a separate cohort of AECOPD, sCOPD, and HC patients confirmed 9 proteins to be differentially expressed by AECOPD compared to HC (Afamin, alpha-1-antichymotrypsin, Apolipoprotein E, Beta-2-glycoprotein 1, Complement component C9, Fibronectin, Immunoglobulin lambda like polypeptide 5, Inter-alpha-trypsin inhibitor heavy chain H3, Leucine rich alpha-2-glycoprotein 1). Network analysis demonstrates that most of these proteins are involved in proteolysis regulation, platelet degranulation and cholesterol metabolism. In conclusion, several potential plasma biomarkers for AECOPD were identified in this study. Further validation studies of these proteins may elucidate their roles in the development of AECOPD.


Subject(s)
Blood Platelets/physiology , Cell Degranulation/physiology , Cholesterol/metabolism , Pulmonary Disease, Chronic Obstructive/metabolism , Apolipoproteins E/metabolism , Biomarkers , Carrier Proteins/metabolism , Case-Control Studies , Complement C9/metabolism , Disease Progression , Fibronectins/metabolism , Glycoproteins/metabolism , Humans , Immunoglobulin Light Chains, Surrogate/metabolism , Metabolic Networks and Pathways , Protein Interaction Maps , Protein Precursors/metabolism , Proteolysis , Proteomics , Serum Albumin, Human/metabolism , alpha 1-Antichymotrypsin/metabolism , beta 2-Glycoprotein I/metabolism
20.
Immunol Cell Biol ; 97(9): 799-814, 2019 10.
Article in English | MEDLINE | ID: mdl-31187539

ABSTRACT

Systemic lupus erythematosus (SLE) is characterized by the development of autoantibodies against diverse self-antigens with damage to multiple organs. Immunization with the SLE autoantigen ß2 -glycoprotein I (ß2 GPI) and lipopolysaccharide (LPS), a known trigger of necroptosis, induces a murine model of SLE. We hypothesized that necroptotic cells, like apoptotic cells, provide a "scaffold" of cellular self-antigens, but, unlike apoptotic cells, necroptotic cells do so in a proinflammatory and immunogenic context. We demonstrate that ß2 GPI indeed binds to necroptotic cells and serves as a target for anti-ß2 GPI autoantibodies. We further demonstrate that necroptotic, but not apoptotic, cells promote antigenic presentation of ß2 GPI to CD4 T cells by dendritic cells. Finally, we show that ß2 GPI/LPS-immunized mice deficient in RIPK3 (receptor-interacting serine/threonine-protein kinase 3) or MLKL (mixed lineage kinase domain like), and consequently unable to undergo necroptosis, have reduced SLE autoantibody production and pathology. RIPK3-/- mice had low levels of SLE autoantibodies and no renal pathology, while MLKL-/- mice produced low levels of SLE autoantibodies initially, but later developed levels comparable with wild type (WT) mice and pathology intermediate to that of WT and RIPK3-/- mice. Serum cytokine levels induced by LPS tended to be lower in RIPK3-/- and MLKL-/- mice than in WT mice, suggesting that impaired proinflammatory cytokine production may impact the initiation of autoantibody production in both strains. Our data suggest that self-antigen (i.e. ß2 GPI) presented in the context of necroptosis and proinflammatory signals may be sufficient to overcome immune tolerance and induce SLE.


Subject(s)
Autoantigens/immunology , Lupus Erythematosus, Systemic/immunology , Necroptosis/immunology , beta 2-Glycoprotein I/metabolism , Animals , Antigen Presentation/immunology , Antigen-Presenting Cells/metabolism , Apoptosis , Autoantibodies/immunology , Cell Line , Cytokines/metabolism , Dendritic Cells/metabolism , Histocompatibility Antigens Class II/metabolism , Inflammation Mediators/metabolism , Lipopolysaccharides/pharmacology , Mice, Inbred C57BL , Protein Binding , Protein Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...