Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
J Lipid Res ; 61(11): 1365-1376, 2020 11.
Article in English | MEDLINE | ID: mdl-32948662

ABSTRACT

Some cases of chylomicronemia are caused by autoantibodies against glycosylphosphatidylinositol-anchored HDL binding protein 1 (GPIHBP1), an endothelial cell protein that shuttles LPL to the capillary lumen. GPIHBP1 autoantibodies prevent binding and transport of LPL by GPIHBP1, thereby disrupting the lipolytic processing of triglyceride-rich lipoproteins. Here, we review the "GPIHBP1 autoantibody syndrome" and summarize clinical and laboratory findings in 22 patients. All patients had GPIHBP1 autoantibodies and chylomicronemia, but we did not find a correlation between triglyceride levels and autoantibody levels. Many of the patients had a history of pancreatitis, and most had clinical and/or serological evidence of autoimmune disease. IgA autoantibodies were present in all patients, and IgG4 autoantibodies were present in 19 of 22 patients. Patients with GPIHBP1 autoantibodies had low plasma LPL levels, consistent with impaired delivery of LPL into capillaries. Plasma levels of GPIHBP1, measured with a monoclonal antibody-based ELISA, were very low in 17 patients, reflecting the inability of the ELISA to detect GPIHBP1 in the presence of autoantibodies (immunoassay interference). However, GPIHBP1 levels were very high in five patients, indicating little capacity of their autoantibodies to interfere with the ELISA. Recently, several GPIHBP1 autoantibody syndrome patients were treated successfully with rituximab, resulting in the disappearance of GPIHBP1 autoantibodies and normalization of both plasma triglyceride and LPL levels. The GPIHBP1 autoantibody syndrome should be considered in any patient with newly acquired and unexplained chylomicronemia.


Subject(s)
Autoantibodies/immunology , Hypertriglyceridemia/immunology , Receptors, Lipoprotein/immunology , Humans
4.
J Clin Lipidol ; 14(2): 197-200, 2020.
Article in English | MEDLINE | ID: mdl-32107180

ABSTRACT

Chylomicronemia caused by a deficiency in lipoprotein lipase (LPL) or GPIHBP1 (the endothelial cell protein that transports LPL to the capillary lumen) is typically diagnosed during childhood and represents a serious, lifelong medical problem. Affected patients have high plasma triglyceride levels (>1500 mg/dL) and a high risk of acute pancreatitis. However, chylomicronemia frequently presents later in life in the absence of an obvious monogenic cause. In these cases, the etiology for the chylomicronemia is presumed to be "multifactorial" (involving diabetes, drugs, alcohol, or polygenic factors), but on a practical level, the underlying cause generally remains a mystery. Here, we describe a 15-year-old female with chylomicronemia caused by GPIHBP1 autoantibodies (which abolish LPL transport to the capillary lumen). Remarkably, chylomicronemia in this patient was intermittent, interspersed between periods when the plasma triglyceride levels were normal. GPIHBP1 autoantibodies were easily detectable during episodes of chylomicronemia but were undetectable during periods of normotriglyceridemia. During the episodes of chylomicronemia (when GPIHBP1 autoantibodies were present), plasma LPL levels were low, consistent with impaired LPL transport into capillaries. During periods of normotriglyceridemia, when GPIHBP1 autoantibodies were absent, plasma LPL levels normalized. Because the chylomicronemia in this patient was accompanied by debilitating episodes of acute pancreatitis, the patient was ultimately treated with immunosuppressive drugs, which resulted in disappearance of GPIHBP1 autoantibodies and normalization of plasma triglyceride levels. GPIHBP1 autoantibodies need to be considered in patients who present with unexplained acquired cases of chylomicronemia.


Subject(s)
Autoantibodies/immunology , Hyperlipoproteinemia Type I/immunology , Receptors, Lipoprotein/immunology , Adolescent , Autoantibodies/blood , Female , Humans
5.
Talanta ; 211: 120660, 2020 May 01.
Article in English | MEDLINE | ID: mdl-32070626

ABSTRACT

An electrochemiluminescence and photothermal immunosensor based on a dual-modality integrated probe was proposed for sensitive and reliable detection of lipolysis stimulated lipoprotein receptor (LSR), a new biomarker of ovarian cancer. Black phosphorous quantum dots (BPQDs) possess fascinating electrochemical property and unique photothermal effect, which could not only enhance ECL signal of N-(4-aminobutyl)-N-ethylisoluminol (ABEI) through accelerating dissolved O2 evolution but also realize temperature signal output by converting laser energy into heat. Furthermore, NiFe2O4 nanotubes (NiFe2O4 NTs) have large specific surface area and favorable adsorption ability, which could increase the immobilized amount of ABEI and BPQDs, further strengthening ECL and temperature signal. As a result, a dual-mode immunosensor was constructed and realized ECL and temperature dual signal to detect LSR, making the results more reliable. This work provided a new thought for the development of sensitive and accurate sensors and was expected to employ for determination of other biomarkers.


Subject(s)
Biomarkers, Tumor/analysis , Biosensing Techniques , Ferric Compounds/chemistry , Nanotubes/chemistry , Nickel/chemistry , Ovarian Neoplasms , Phosphorus/chemistry , Quantum Dots/chemistry , Receptors, Lipoprotein/analysis , Antibodies/chemistry , Biomarkers, Tumor/immunology , Electrochemical Techniques , Female , Humans , Immunoassay , Light , Luminescent Measurements , Luminol/analogs & derivatives , Luminol/chemistry , Receptors, Lipoprotein/immunology , Temperature
6.
Transfus Clin Biol ; 26(4): 346-351, 2019 Nov.
Article in English | MEDLINE | ID: mdl-30797678

ABSTRACT

Presentation of exogenous antigens loaded on major histocompatibility complex class I molecules by antigen presenting cells, termed cross-presentation, is essential for the induction of CD8+ T cells and is performed mainly by specialized dendritic cell subsets. Research into this field has described two main mechanisms of cross-presentation, the cytosolic pathway and the vacuolar pathway. As the first step in cross-presentation, surface receptors relating to cross-presentation are required in the recognition and uptake of Ags, which include C-type lectin receptors, immunoglobulin γ Fc region receptor, chemokine receptor, scavenger receptor etc. After uptake by the cells, there are also many molecules that enable Ags to participate in cross-presentation pathways. By this approach, exogenous Ags can induce CD8+ T cells into cytotoxic T lymphocytes, which is of great significance to induce antitumor and antiviral immune responses, and the molecular mechanism would facilitate the development of related adjuvants. However, the detailed mechanisms of cross-presentation still remain unknown. In this paper, some latest researches, including two major pathways, DC surface receptors and application prospects are summarized.


Subject(s)
Antigen Presentation , Antigens, Heterophile/immunology , Cross-Priming , Isoantigens/immunology , Adjuvants, Immunologic/pharmacology , Animals , Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/immunology , Cytosol/immunology , Dendritic Cells/immunology , Histocompatibility Antigens Class I/immunology , Humans , Lipoproteins, HDL/immunology , Mice , Models, Immunological , Receptors, Fc/immunology , Receptors, Immunologic/immunology , Receptors, Lipoprotein/immunology , Vacuoles/immunology
7.
J Clin Lipidol ; 13(1): 62-69, 2019.
Article in English | MEDLINE | ID: mdl-30514621

ABSTRACT

BACKGROUND: Autoantibodies against glycosylphosphatidylinositol-anchored high-density lipoprotein-binding protein 1 (GPIHBP1) cause chylomicronemia by blocking the ability of GPIHBP1 to bind lipoprotein lipase (LPL) and transport the enzyme to its site of action in the capillary lumen. OBJECTIVE: A patient with multiple sclerosis developed chylomicronemia during interferon (IFN) ß1a therapy. The chylomicronemia resolved when the IFN ß1a therapy was discontinued. Here, we sought to determine whether the drug-induced chylomicronemia was caused by GPIHBP1 autoantibodies. METHODS: We tested plasma samples collected during and after IFN ß1a therapy for GPIHBP1 autoantibodies (by western blotting and with enzyme-linked immunosorbent assays). We also tested whether the patient's plasma blocked the binding of LPL to GPIHBP1 on GPIHBP1-expressing cells. RESULTS: During IFN ß1a therapy, the plasma contained GPIHBP1 autoantibodies, and those autoantibodies blocked GPIHBP1's ability to bind LPL. Thus, the chylomicronemia was because of the GPIHBP1 autoantibody syndrome. Consistent with that diagnosis, the plasma levels of GPIHBP1 and LPL were very low. After IFN ß1a therapy was stopped, the plasma triglyceride levels returned to normal, and GPIHBP1 autoantibodies were undetectable. CONCLUSION: The appearance of GPIHBP1 autoantibodies during IFN ß1a therapy caused chylomicronemia. The GPIHBP1 autoantibodies disappeared when the IFN ß1a therapy was stopped, and the plasma triglyceride levels fell within the normal range.


Subject(s)
Autoimmune Diseases/immunology , Drug-Related Side Effects and Adverse Reactions/immunology , Hyperlipoproteinemia Type I/immunology , Interferon-beta/adverse effects , Multiple Sclerosis/therapy , Receptors, Lipoprotein/immunology , Adult , Autoantibodies/blood , Autoimmune Diseases/etiology , Cells, Cultured , Female , Humans , Hyperlipoproteinemia Type I/etiology , Interferon-beta/therapeutic use , Multiple Sclerosis/complications , Protein Binding , Syndrome , Triglycerides/blood , Withholding Treatment
8.
Exp Cell Res ; 371(1): 31-41, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30044945

ABSTRACT

Guanylate-binding protein-1 (GBP-1) is an interferon-inducible large GTPase involved in the epithelial barrier at tight junctions. To investigate the role of GBP-1 in the epithelial barrier, primary human salivary gland duct epithelial cells were treated with the the proinflammatory cytokines IFNγ, IL-1ß, TNFα and the growth factor TGF-ß. Treatment with IFNγ, IL-1ß, or TNFα markedly enhanced GBP-1 and the epithelial barrier function, and induced not only CLDN-7 but also the tricellular tight junction molecule lipolysis-stimulated lipoprotein receptor (LSR). Knockdown of GBP-1 by its siRNA induced endocytosis of tight junction molecules, and prevented the increases of CLDN-7 and LSR with the upregulation of the epithelial barrier function induced by treatment with IFNγ or TNFα. Treatment with a PKCα inhibitor induced expression of GBP-1, CLDN-7 and LSR and enhanced the epithelial barrier function. In almost intact salivary gland ducts from patients with IgG4-related disease (IgG4-RD) indicated significant infiltration of IgG-positive plasma cells, expression of GBP-1, CLDN-7 and LSR was increased. These findings indicated that GBP-1 might play a crucial role in barrier function of normal human salivary gland duct epithelium and perform a preventive role in the duct epithelium of IgG4-RD disease.


Subject(s)
Claudins/genetics , Epithelial Cells/metabolism , GTP-Binding Proteins/genetics , Immunoglobulin G4-Related Disease/genetics , Immunoglobulin G/genetics , Receptors, Lipoprotein/genetics , Tight Junctions/metabolism , Biological Transport , Claudins/immunology , Endocytosis , Epithelial Cells/drug effects , Epithelial Cells/immunology , Epithelial Cells/pathology , Epithelium/drug effects , Epithelium/immunology , Epithelium/pathology , Epithelium/surgery , GTP-Binding Proteins/antagonists & inhibitors , GTP-Binding Proteins/immunology , Gene Expression Regulation , Humans , Immunoglobulin G/metabolism , Immunoglobulin G4-Related Disease/immunology , Immunoglobulin G4-Related Disease/pathology , Immunoglobulin G4-Related Disease/surgery , Interferon-gamma/pharmacology , Occludin/genetics , Occludin/immunology , Permeability/drug effects , Plasma Cells/immunology , Plasma Cells/pathology , Primary Cell Culture , RNA, Small Interfering/genetics , RNA, Small Interfering/immunology , Receptors, Lipoprotein/immunology , Salivary Ducts/immunology , Salivary Ducts/pathology , Salivary Ducts/surgery , Signal Transduction , Tight Junctions/drug effects , Tight Junctions/immunology , Tight Junctions/ultrastructure , Transcription Factors , Tumor Necrosis Factor-alpha/pharmacology
10.
Cancer Res ; 78(2): 516-527, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29187404

ABSTRACT

Epithelial ovarian cancer (EOC) is the most lethal gynecologic malignancy, but it still lacks effective treatment options. In this study, we utilized proteomic technology to identify lipolysis-stimulated lipoprotein receptor (LSR) as a new tumor antigen of EOC. Immunohistochemical analysis of EOC tissues in conjunction with survival analysis of EOC patients showed that high expression of LSR is associated with poor prognosis. High LSR expression also occurred in tumor metastases including to the lymph node and omentum. To evaluate the possible benefits of blocking this antigen in EOC, we raised a new monoclonal antibody (mAb) to human LSR (hLSR). In mouse xenograft models of hLSR+ EOC (cell lines or patient-derived tumors), we found that administration of anti-hLSR mAb inhibited tumor growth in a manner independent of both antibody-dependent cellular cytotoxicity and complement-dependent cytotoxicity. Mechanistic investigations showed that hLSR expression increased incorporation of very-low-density lipoprotein (VLDL) into EOC cells and that anti-hLSR mAb inhibited lipid uptake in vitro and in vivo Moreover, VLDL promoted cell proliferation in hLSR-positive EOC cells in vitro, and this effect was inhibited by anti-hLSR mAb. While the anti-hLSR mAb studied cross reacted with the mouse antigen, we observed no adverse effects on normal organs and lipid metabolism in murine hosts. Our findings suggest that hLSR plays a key functional role in EOC development and that this antigen can be therapeutically targeted by specific mAb to improve EOC treatment.Significance: These findings offer preclinical evidence of the therapeutic efficacy of a novel targeted antibody therapy against deadly epithelial ovarian cancers. Cancer Res; 78(2); 516-27. ©2017 AACR.


Subject(s)
Antibodies, Monoclonal/pharmacology , Biomarkers, Tumor/metabolism , Lipids/pharmacokinetics , Neoplasm Recurrence, Local/therapy , Neoplasms, Glandular and Epithelial/therapy , Ovarian Neoplasms/therapy , Receptors, Lipoprotein/immunology , Adult , Aged , Animals , Antibody-Dependent Cell Cytotoxicity , Apoptosis , Carcinoma, Ovarian Epithelial , Case-Control Studies , Cell Proliferation , Female , Follow-Up Studies , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Middle Aged , Neoplasm Recurrence, Local/immunology , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/pathology , Neoplasms, Glandular and Epithelial/immunology , Neoplasms, Glandular and Epithelial/metabolism , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/immunology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Prognosis , Proteomics , Receptors, Lipoprotein/metabolism , Survival Rate , Tissue Distribution , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
11.
J Clin Lipidol ; 12(1): 203-210.e1, 2018.
Article in English | MEDLINE | ID: mdl-29246728

ABSTRACT

BACKGROUND: Glycosylphosphatidylinositol-anchored high-density lipoprotein-binding protein 1 (GPIHBP1), a glycosylphosphatidylinositol (GPI)-anchored protein of capillary endothelial cells, transports lipoprotein lipase to the capillary lumen and is essential for the lipolytic processing of triglyceride-rich lipoproteins. OBJECTIVE: Because some GPI-anchored proteins have been detected in plasma, we tested whether GPIHBP1 is present in human blood and whether GPIHBP1 deficiency or a history of cardiovascular disease affected GPIHBP1 circulating levels. METHODS: We developed 2 monoclonal antibodies against GPIHBP1 and used the antibodies to establish a sandwich enzyme-linked immunosorbent assay (ELISA) to measure GPIHBP1 levels in human blood. RESULTS: The GPIHBP1 ELISA was linear in the 8 to 500 pg/mL range and allowed the quantification of GPIHBP1 in serum and in pre- and post-heparin plasma (including lipemic samples). GPIHBP1 was undetectable in the plasma of subjects with null mutations in GPIHBP1. Serum GPIHBP1 median levels were 849 pg/mL (range: 740-1014) in healthy volunteers (n = 28) and 1087 pg/mL (range: 877-1371) in patients with a history of cardiovascular or metabolic disease (n = 415). There was an extremely small inverse correlation between GPIHBP1 and triglyceride levels (r = 0.109; P < .0275). GPIHBP1 levels tended to be slightly higher in patients who had a major cardiovascular event after revascularization. CONCLUSION: We developed an ELISA for quantifying GPIHBP1 in human blood. This assay will be useful to identify patients with GPIHBP1 deficiency and patients with GPIHBP1 autoantibodies. The potential of plasma GPIHBP1 as a biomarker for metabolic or cardiovascular disease is yet questionable but needs additional testing.


Subject(s)
Enzyme-Linked Immunosorbent Assay , Receptors, Lipoprotein/blood , Aged , Antibodies, Monoclonal/immunology , Cardiovascular Diseases/pathology , Case-Control Studies , Female , Humans , Male , Metabolic Diseases/pathology , Middle Aged , Receptors, Lipoprotein/immunology , Triglycerides/blood
12.
J Clin Lipidol ; 11(4): 964-971, 2017.
Article in English | MEDLINE | ID: mdl-28666713

ABSTRACT

BACKGROUND: GPIHBP1, a glycolipid-anchored protein of capillary endothelial cells, binds lipoprotein lipase (LPL) in the interstitial spaces and transports it to the capillary lumen. GPIHBP1 deficiency prevents LPL from reaching the capillary lumen, resulting in low intravascular LPL levels, impaired intravascular triglyceride processing, and severe hypertriglyceridemia (chylomicronemia). A recent study showed that some cases of hypertriglyceridemia are caused by autoantibodies against GPIHBP1 ("GPIHBP1 autoantibody syndrome"). OBJECTIVE: Our objective was to gain additional insights into the frequency of the GPIHBP1 autoantibody syndrome in patients with unexplained chylomicronemia. METHODS: We used enzyme-linked immunosorbent assays to screen for GPIHBP1 autoantibodies in 33 patients with unexplained chylomicronemia and then used Western blots and immunocytochemistry studies to characterize the GPIHBP1 autoantibodies. RESULTS: The plasma of 1 patient, a 36-year-old man with severe hypertriglyceridemia, contained GPIHBP1 autoantibodies. The autoantibodies, which were easily detectable by Western blot, blocked the ability of GPIHBP1 to bind LPL. The plasma levels of LPL mass and activity were low. The patient had no history of autoimmune disease, but his plasma was positive for antinuclear antibodies. CONCLUSIONS: One of 33 patients with unexplained chylomicronemia had the GPIHBP1 autoantibody syndrome. Additional studies in large lipid clinics will be helpful for better defining the frequency of this syndrome and for exploring the best strategies for treatment.


Subject(s)
Autoantibodies/blood , Autoantibodies/immunology , Hyperlipoproteinemia Type I/blood , Hyperlipoproteinemia Type I/immunology , Receptors, Lipoprotein/immunology , Adult , Animals , Cell Line , Humans , Hyperlipoproteinemia Type I/complications , Hyperlipoproteinemia Type I/genetics , Hypertriglyceridemia/complications , Male , Mutation
13.
N Engl J Med ; 376(17): 1647-1658, 2017 04 27.
Article in English | MEDLINE | ID: mdl-28402248

ABSTRACT

BACKGROUND: A protein that is expressed on capillary endothelial cells, called GPIHBP1 (glycosylphosphatidylinositol-anchored high-density lipoprotein binding protein 1), binds lipoprotein lipase and shuttles it to its site of action in the capillary lumen. A deficiency in GPIHBP1 prevents lipoprotein lipase from reaching the capillary lumen. Patients with GPIHBP1 deficiency have low plasma levels of lipoprotein lipase, impaired intravascular hydrolysis of triglycerides, and severe hypertriglyceridemia (chylomicronemia). During the characterization of a monoclonal antibody-based immunoassay for GPIHBP1, we encountered two plasma samples (both from patients with chylomicronemia) that contained an interfering substance that made it impossible to measure GPIHBP1. That finding raised the possibility that those samples might contain GPIHBP1 autoantibodies. METHODS: Using a combination of immunoassays, Western blot analyses, and immunocytochemical studies, we tested the two plasma samples (as well as samples from other patients with chylomicronemia) for the presence of GPIHBP1 autoantibodies. We also tested the ability of GPIHBP1 autoantibodies to block the binding of lipoprotein lipase to GPIHBP1. RESULTS: We identified GPIHBP1 autoantibodies in six patients with chylomicronemia and found that these autoantibodies blocked the binding of lipoprotein lipase to GPIHBP1. As in patients with GPIHBP1 deficiency, those with GPIHBP1 autoantibodies had low plasma levels of lipoprotein lipase. Three of the six patients had systemic lupus erythematosus. One of these patients who had GPIHBP1 autoantibodies delivered a baby with plasma containing maternal GPIHBP1 autoantibodies; the infant had severe but transient chylomicronemia. Two of the patients with chylomicronemia and GPIHBP1 autoantibodies had a response to treatment with immunosuppressive agents. CONCLUSIONS: In six patients with chylomicronemia, GPIHBP1 autoantibodies blocked the ability of GPIHBP1 to bind and transport lipoprotein lipase, thereby interfering with lipoprotein lipase-mediated processing of triglyceride-rich lipoproteins and causing severe hypertriglyceridemia. (Funded by the National Heart, Lung, and Blood Institute and the Leducq Foundation.).


Subject(s)
Autoantibodies/blood , Hyperlipoproteinemia Type I/immunology , Lipoprotein Lipase/metabolism , Receptors, Lipoprotein/immunology , Adult , Autoantibodies/physiology , Female , Humans , Hyperlipoproteinemia Type I/blood , Immunoassay , Lipolysis , Lipoprotein Lipase/blood , Male , Middle Aged , Protein Binding , Protein Transport , Receptors, Lipoprotein/metabolism
14.
J Lipid Res ; 58(1): 208-215, 2017 01.
Article in English | MEDLINE | ID: mdl-27875259

ABSTRACT

GPIHBP1, an endothelial cell protein, binds LPL in the interstitial spaces and shuttles it to its site of action inside blood vessels. For years, studies of human GPIHBP1 have been hampered by an absence of useful antibodies. We reasoned that monoclonal antibodies (mAbs) against human GPIHBP1 would be useful for 1) defining the functional relevance of GPIHBP1's Ly6 and acidic domains to the binding of LPL; 2) ascertaining whether human GPIHBP1 is expressed exclusively in capillary endothelial cells; and 3) testing whether GPIHBP1 is detectable in human plasma. Here, we report the development of a panel of human GPIHBP1-specific mAbs. Two mAbs against GPIHBP1's Ly6 domain, RE3 and RG3, abolished LPL binding, whereas an antibody against the acidic domain, RF4, did not. Also, mAbs RE3 and RG3 bound with reduced affinity to a mutant GPIHBP1 containing an Ly6 domain mutation (W109S) that abolishes LPL binding. Immunohistochemistry studies with the GPIHBP1 mAbs revealed that human GPIHBP1 is expressed only in capillary endothelial cells. Finally, we created an ELISA that detects GPIHBP1 in human plasma. That ELISA should make it possible for clinical lipidologists to determine whether plasma GPIHBP1 levels are a useful biomarker of metabolic or vascular disease.


Subject(s)
Antibodies, Monoclonal/immunology , Lipoprotein Lipase/immunology , Receptors, Lipoprotein/immunology , Triglycerides/metabolism , Animals , Binding Sites/immunology , Cell Line , Drosophila , Endothelial Cells/enzymology , Endothelial Cells/immunology , Humans , Lipoprotein Lipase/antagonists & inhibitors , Lipoprotein Lipase/isolation & purification , Mice , Receptors, Lipoprotein/genetics , Triglycerides/immunology
15.
Curr Opin Endocrinol Diabetes Obes ; 21(2): 121-8, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24569553

ABSTRACT

PURPOSE OF REVIEW: To summarize the recent findings about the roles of scavenger receptor class B type I (SR-BI) in immunity and discuss the underlying mechanisms by which SR-BI prevents immune dysfunctions. RECENT FINDINGS: SR-BI is well known as a high-density lipoprotein (HDL) receptor playing key roles in HDL metabolism and in protection against atherosclerosis. Recent studies have indicated that SR-BI is also an essential modulator in immunity. SR-BI deficiency in mice causes immune dysfunctions, including increased atherosclerosis, elevated susceptibility to sepsis, impaired lymphocyte homeostasis, and autoimmune disorders. SR-BI exerts its protective roles through a variety of HDL-dependent and HDL-independent mechanisms. SR-BI is also involved in hepatitis C virus cell entry. A deficiency of SR-BI in humanized mice has been shown to decrease hepatitis C virus infectivity. SUMMARY: SR-BI regulates immunity via multiple mechanisms and its deficiency causes numerous diseases. A comprehensive understanding of the roles of SR-BI in protection against immune dysfunctions may provide a therapeutic target for intervention against its associated diseases.


Subject(s)
Adaptive Immunity , Atherosclerosis/immunology , Autoimmune Diseases/immunology , Autoimmune Diseases/prevention & control , Oxidative Stress/immunology , Scavenger Receptors, Class B/immunology , Sepsis/immunology , Animals , Atherosclerosis/metabolism , Autoimmune Diseases/metabolism , Cell Proliferation , Disease Models, Animal , Humans , Lipoproteins, HDL/immunology , Mice , Mice, Knockout , Receptors, Lipoprotein/immunology , Scavenger Receptors, Class B/deficiency , Sepsis/metabolism
16.
Curr Neurol Neurosci Rep ; 14(1): 421, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24272275

ABSTRACT

The majority of patients with myasthenia gravis (MG) initially present with ocular symptoms. An unresolved question is whether there are clinical features at onset to guide clinicians to predict an individual patient's conversion risk from ocular MG (OMG) to generalized disease, or "secondary generalized MG" (SGMG), that is, a prognostic model. In light of the emerging theory that early corticosteroids may have a risk-modifying effect, the factors associated with secondary SGMG previously reported should be revisited. Studies showing potential risk-modifying effects of corticosteroids are useful, though flawed, owing to the heterogeneous retrospective studies and methods of reporting. Updates on other potential immunosuppressive agents are also discussed. Thymectomy in OMG has been recently reported in a few studies to be useful. MG associated with antibodies to muscle-specific kinase, usually associated with severe generalized MG, can cause a pure OMG syndrome. Recent serological developments in seronegative patients have also revealed antibodies to clustered anti-acetylcholine receptor and lipoprotein receptor-related protein-4.


Subject(s)
Antibodies/blood , Myasthenia Gravis , Receptors, Cholinergic/immunology , Receptors, Lipoprotein/immunology , Humans , Immunosuppressive Agents/therapeutic use , Myasthenia Gravis/blood , Myasthenia Gravis/immunology , Myasthenia Gravis/therapy , Thymectomy
17.
Patol Fiziol Eksp Ter ; (2): 62-9, 2012.
Article in Russian | MEDLINE | ID: mdl-22708412

ABSTRACT

Current knowledge of immunocellular and lipoprotein mechanisms of the liver-induced anti-endotoxin tolerance has been summarized. The role of T regulatory cells, different macrophage phenotypes, high density lipoproteins, oxidized low density lipoproteins and their receptors as the key players in mechanism of tolerance to endotoxin has been discussed.


Subject(s)
Endotoxins/immunology , Immune Tolerance , Liver/immunology , Macrophages/immunology , T-Lymphocytes, Regulatory/immunology , Immunity, Cellular , Lipoproteins/immunology , Receptors, Lipoprotein/immunology
18.
Neurosci Res ; 63(4): 251-8, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19167437

ABSTRACT

Reelin plays critical roles in brain formation by binding to apolipoprotein E receptor 2 (ApoER2) and very low-density lipoprotein receptor. Several isoforms and fragments of Reelin are generated by alternative splicing and proteolytic cleavage. In addition, two splice variants of ApoER2 have been recognized, namely, LA1237 and LA12378, that differ in the number of ligand-binding type A (LA) repeats. Here, we quantitatively investigated the affinity between various isoforms/fragments of Reelin and the ApoER2 splice variants. ApoER2-LA1237 bound rather strongly to the Reelin central fragment than to the fragment bearing Reelin repeat 8 (RR8). ApoER2-LA12378 bound comparably to all Reelin fragments without the C-terminal region. These findings suggest that LA8 of ApoER2 and RR8 interfere with the interaction between the Reelin central fragment and ApoER2. Using a monoclonal antibody that only recognizes ApoER2-LA12378, we found that this variant of ApoER2 was expressed in the cerebral cortical wall and in the internal granule cells of the cerebellum during development. Primary-cultured cortical neurons did not express ApoER2-LA12378, and the extent of signal activation by Reelin fragments was well correlated with their affinity for ApoER2-LA1237. Therefore, proteolytic cleavage of Reelin and alternative splicing of ApoER2 may be involved in the fine regulation of Reelin signaling.


Subject(s)
Alternative Splicing , Cell Adhesion Molecules, Neuronal/metabolism , Extracellular Matrix Proteins/metabolism , Nerve Tissue Proteins/metabolism , Receptors, Lipoprotein/genetics , Receptors, Lipoprotein/metabolism , Serine Endopeptidases/metabolism , Animals , Animals, Newborn , Antibodies, Monoclonal/metabolism , Binding Sites , Cells, Cultured , Cerebral Cortex/cytology , Embryo, Mammalian , Gene Expression Regulation, Developmental , Green Fluorescent Proteins/genetics , Humans , Inositol 1,4,5-Trisphosphate Receptors/metabolism , LDL-Receptor Related Proteins , Ligands , Mice , Mice, Inbred ICR , Microtubule-Associated Proteins/metabolism , Models, Biological , Molecular Sequence Data , Neurons , Protein Binding/genetics , Protein Isoforms/genetics , Protein Structure, Tertiary , Receptors, Lipoprotein/immunology , Reelin Protein , Signal Transduction/physiology , Transfection/methods
20.
Clin Exp Immunol ; 142(3): 441-5, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16297155

ABSTRACT

Atherosclerosis is associated with activation of the immune system. Intravenously applied normal polyclonal immunoglobulins (IVIg) have broad therapeutic applications in the treatment of autoimmune and systemic inflammatory diseases. Recently, IVIg have been shown to inhibit atherogenesis in experimental animal models. To investigate the role of the complement system in this process, we used third complement component-deficient (C3(-/-)) and control atherosclerosis-prone apolipoprotein E (ApoE) and low-density lipoprotein receptor (LDLR) double knock-out mice fed a normal diet. IVIg treatment reduced lesion fraction area in the aortic root of complement-sufficient mice whereas the lesion fraction area of C3(-/-) mice was not affected. Thus, complement activation plays a role in the anti-atherosclerotic effects of IVIg, possibly by C3-derived fragments generated through Fc-dependent complement activation.


Subject(s)
Apolipoproteins E/immunology , Arteriosclerosis/immunology , Complement C3/deficiency , Immunoglobulins, Intravenous/immunology , Receptors, Lipoprotein/immunology , Animals , Complement Activation/immunology , Complement C3/immunology , Disease Models, Animal , Immunoglobulins, Intravenous/administration & dosage , Immunohistochemistry/methods , Injections, Intraperitoneal , Macrophages/immunology , Male , Mice , Mice, Knockout , Monocytes/immunology , Receptors, IgG/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...