Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 184
Filter
1.
J Clin Invest ; 132(3)2022 02 01.
Article in English | MEDLINE | ID: mdl-34784295

ABSTRACT

Bitter taste receptors (taste 2 receptors, TAS2Rs) serve as warning sensors in the lingual system against the ingestion of potentially poisonous food. Here, we investigated the functional role of TAS2Rs in the human gut and focused on their potential to trigger an additional host defense pathway in the intestine. Human jejunal crypts, especially those from individuals with obesity, responded to bitter agonists by inducing the release of antimicrobial peptides (α-defensin 5 and regenerating islet-derived protein 3 α [REG3A]) but also regulated the expression of other innate immune factors (mucins, chemokines) that affected E. coli growth. We found that the effect of aloin on E. coli growth and on the release of the mucus glycoprotein CLCA1, identified via proteomics, was affected by TAS2R43 deletion polymorphisms and thus confirmed a role for TAS2R43. RNA-Seq revealed that denatonium benzoate induced an NRF2-mediated nutrient stress response and an unfolded protein response that increased the expression of the mitokine GDF15 but also ADM2 and LDLR, genes that are involved in anorectic signaling and lipid homeostasis. In conclusion, TAS2Rs in the intestine constitute a promising target for treating diseases that involve disturbances in the innate immune system and body weight control. TAS2R polymorphisms may be valuable genetic markers to predict therapeutic responses.


Subject(s)
Immunity, Innate , Intestinal Mucosa/immunology , Obesity/immunology , Receptors, G-Protein-Coupled/immunology , Growth Differentiation Factor 15/immunology , Humans , Male , Middle Aged , Pancreatitis-Associated Proteins/immunology , Peptide Hormones/immunology , RNA-Seq , Receptors, LDL/immunology
2.
Front Endocrinol (Lausanne) ; 12: 601160, 2021.
Article in English | MEDLINE | ID: mdl-33815271

ABSTRACT

Background: Non-alcoholic fatty liver disease (NAFLD) is a complex multifactorial disorder that is characterised by dysfunctional lipid metabolism and cholesterol homeostasis, and a related chronic inflammatory response. NAFLD has become the most common cause of chronic liver disease in many countries, and its prevalence continues to rise in parallel with increasing rates of obesity. Here, we evaluated the putative NAFLD-attenuating effects of a multicomponent medicine consisting of 24 natural ingredients: Hepar compositum (HC-24). Methods: Ldlr-/-.Leiden mice were fed a high-fat diet (HFD) with a macronutrient composition and cholesterol content comparable to human diets for 24 weeks to induce obesity-associated metabolic dysfunction, including hepatic steatosis and inflammation. HC-24 or vehicle control was administered intraperitoneally 3 times/week (1.5 ml/kg) for the last 18 weeks of the study. Histological analyses of liver and adipose tissue were combined with extensive hepatic transcriptomics analysis. Transcriptomics results were further substantiated with ELISA, immunohistochemical and liver lipid analyses. Results: HFD feeding induced obesity and metabolic dysfunction including adipose tissue inflammation and increased gut permeability. In the liver, HFD-feeding resulted in a disturbance of cholesterol homeostasis and an associated inflammatory response. HC-24 did not affect body weight, metabolic risk factors, adipose tissue inflammation or gut permeability. While HC-24 did not alter total liver steatosis, there was a pronounced reduction in lobular inflammation in HC-24-treated animals, which was associated with modulation of genes and proteins involved in inflammation (e.g., neutrophil chemokine Cxcl1) and cholesterol homeostasis (i.e., predicted effect on 'cholesterol' as an upstream regulator, based on gene expression changes associated with cholesterol handling). These effects were confirmed by CXCL1 ELISA, immunohistochemical staining of neutrophils and biochemical analysis of hepatic free cholesterol content. Intrahepatic free cholesterol levels were found to correlate significantly with the number of inflammatory aggregates in the liver, thereby providing a potential rationale for the observed anti-inflammatory effects of HC-24. Conclusions: Free cholesterol accumulates in the liver of Ldlr-/-.Leiden mice under physiologically translational dietary conditions, and this is associated with the development of hepatic inflammation. The multicomponent medicine HC-24 reduces accumulation of free cholesterol and has molecular and cellular anti-inflammatory effects in the liver.


Subject(s)
Cholesterol/metabolism , Non-alcoholic Fatty Liver Disease/drug therapy , Plant Extracts/administration & dosage , Animals , Chemokine CXCL1/genetics , Chemokine CXCL1/metabolism , Diet, High-Fat/adverse effects , Humans , Lipid Metabolism/drug effects , Liver/drug effects , Liver/immunology , Liver/metabolism , Male , Mice , Mice, Knockout , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/immunology , Non-alcoholic Fatty Liver Disease/metabolism , Receptors, LDL/genetics , Receptors, LDL/immunology
3.
Protein Cell ; 12(4): 240-260, 2021 04.
Article in English | MEDLINE | ID: mdl-33606190

ABSTRACT

Metabolic regulation has been proven to play a critical role in T cell antitumor immunity. However, cholesterol metabolism as a key component of this regulation remains largely unexplored. Herein, we found that the low-density lipoprotein receptor (LDLR), which has been previously identified as a transporter for cholesterol, plays a pivotal role in regulating CD8+ T cell antitumor activity. Besides the involvement of cholesterol uptake which is mediated by LDLR in T cell priming and clonal expansion, we also found a non-canonical function of LDLR in CD8+ T cells: LDLR interacts with the T-cell receptor (TCR) complex and regulates TCR recycling and signaling, thus facilitating the effector function of cytotoxic T-lymphocytes (CTLs). Furthermore, we found that the tumor microenvironment (TME) downregulates CD8+ T cell LDLR level and TCR signaling via tumor cell-derived proprotein convertase subtilisin/kexin type 9 (PCSK9) which binds to LDLR and prevents the recycling of LDLR and TCR to the plasma membrane thus inhibits the effector function of CTLs. Moreover, genetic deletion or pharmacological inhibition of PCSK9 in tumor cells can enhance the antitumor activity of CD8+ T cells by alleviating the suppressive effect on CD8+ T cells and consequently inhibit tumor progression. While previously established as a hypercholesterolemia target, this study highlights PCSK9/LDLR as a potential target for cancer immunotherapy as well.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunity, Cellular , Neoplasms/immunology , Proprotein Convertase 9/immunology , Receptors, Antigen, T-Cell/immunology , Receptors, LDL/immunology , Signal Transduction/immunology , Animals , Cell Membrane/genetics , Cell Membrane/immunology , Humans , Mice , Mice, Knockout , Neoplasms/genetics , Proprotein Convertase 9/genetics , Receptors, Antigen, T-Cell/genetics , Receptors, LDL/genetics , Signal Transduction/genetics
4.
Dis Markers ; 2020: 8974793, 2020.
Article in English | MEDLINE | ID: mdl-32454908

ABSTRACT

A growing body of evidence has indicated that behaviors of cancers are defined by not only intrinsic activities of tumor cells but also tumor-infiltrating immune cells (TIICs) in the tumor microenvironment. However, it still lacks a well-structured and comprehensive analysis of TIICs and its therapeutic value in esophageal cancer (EC). The proportions of 22 TIICs were evaluated between 150 normal tissues and 141 tumor tissues of EC by the CIBERSORT algorithm. Besides, correlation analyses between proportions of TIICs and clinicopathological characters, including age, gender, histologic grade, tumor location, histologic type, LRP1B mutation, TP53 mutation, tumor stage, lymph node stage, and TNM stage, were conducted. We constructed a risk score model to improve prognostic capacity with 5 TIICs by least absolute shrinkage and selection operator (lasso) regression analysis. The risk score = -1.86∗plasma + 2.56∗T cell follicular helper - 1.37∗monocytes - 3.64∗activated dendritic cells - 2.24∗resting mast cells (immune cells in the risk model mean the proportions of immune cell infiltration in EC). Patients in the high-risk group had significantly worse overall survival than these in the low-risk group (HR: 2.146, 95% CI: 1.243-3.705, p = 0.0061). Finally, we identified Semustine and Sirolimus as two candidate compounds for the treatment of EC based on CMap analysis. In conclusion, the proportions of TIICs may be important to the progression, prognosis, and treatment of EC.


Subject(s)
Antineoplastic Agents/therapeutic use , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/immunology , Lymphocytes, Tumor-Infiltrating/pathology , Tumor Microenvironment/immunology , Adult , Algorithms , Cell Count , Databases, Factual , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/pathology , Esophageal Neoplasms/diagnosis , Esophageal Neoplasms/mortality , Female , Gene Expression , Humans , Lymph Nodes/drug effects , Lymph Nodes/immunology , Lymph Nodes/pathology , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Male , Mast Cells/drug effects , Mast Cells/immunology , Mast Cells/pathology , Middle Aged , Monocytes/drug effects , Monocytes/immunology , Monocytes/pathology , Neoplasm Staging , Plasma Cells/drug effects , Plasma Cells/immunology , Plasma Cells/pathology , Prognosis , Receptors, LDL/genetics , Receptors, LDL/immunology , Regression Analysis , Semustine/therapeutic use , Sirolimus/therapeutic use , Survival Analysis , T Follicular Helper Cells/drug effects , T Follicular Helper Cells/immunology , T Follicular Helper Cells/pathology , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/immunology
5.
J Exp Med ; 216(2): 350-368, 2019 02 04.
Article in English | MEDLINE | ID: mdl-30647120

ABSTRACT

Heart failure due to dilated cardiomyopathy is frequently caused by myocarditis. However, the pathogenesis of myocarditis remains incompletely understood. Here, we report the presence of neutrophil extracellular traps (NETs) in cardiac tissue of patients and mice with myocarditis. Inhibition of NET formation in experimental autoimmune myocarditis (EAM) of mice substantially reduces inflammation in the acute phase of the disease. Targeting the cytokine midkine (MK), which mediates NET formation in vitro, not only attenuates NET formation in vivo and the infiltration of polymorphonuclear neutrophils (PMNs) but also reduces fibrosis and preserves systolic function during EAM. Low-density lipoprotein receptor-related protein 1 (LRP1) acts as the functionally relevant receptor for MK-induced PMN recruitment as well as NET formation. In summary, NETosis substantially contributes to the pathogenesis of myocarditis and drives cardiac inflammation, probably via MK, which promotes PMN trafficking and NETosis. Thus, MK as well as NETs may represent novel therapeutic targets for the treatment of cardiac inflammation.


Subject(s)
Autoimmune Diseases/immunology , Cell Movement/immunology , Extracellular Traps/immunology , Midkine/immunology , Myocarditis/immunology , Myocardium/immunology , Neutrophils/immunology , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/pathology , Cell Movement/genetics , Extracellular Traps/genetics , Humans , Low Density Lipoprotein Receptor-Related Protein-1/genetics , Low Density Lipoprotein Receptor-Related Protein-1/immunology , Mice , Mice, Transgenic , Midkine/genetics , Myocarditis/genetics , Myocarditis/pathology , Myocardium/pathology , Neutrophils/pathology , Receptors, LDL/genetics , Receptors, LDL/immunology , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/immunology
6.
Biofactors ; 45(1): 43-48, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30496627

ABSTRACT

Leptin influences inflammation and immune response. Dose-dependent effects of leptin on biomarkers of inflammation have not been studied in vivo, so far. Leptin-deficient low-density lipoprotein receptor (LDLR) knockout (LDLR-/- ;ob/ob) female mice were treated with three different leptin doses or saline for 12 weeks. The effect of leptin on plasma interleukin (IL)-6 and monocyte chemoattractant protein (MCP)-1 concentrations and Il-6 and Mcp-1 mRNA expression in vivo were assessed. Macrophage infiltration in epididymal adipose tissue (epiAT) after leptin treatment was determined by quantitative immunohistochemical analysis. Aortic root atherosclerotic lesions were analyzed by oil red O staining. Mean plasma IL-6 and MCP-1 decreased significantly in the 3.0 mg/kg BW/day group as compared to control mice (both P < 0.01). Messenger RNA expression of Il-6 and Mcp-1 was significantly down-regulated by leptin treatment in different adipose tissues in vivo. Characteristic crown-like structures formed by adipose tissue macrophages were significantly reduced by leptin treatment in epiAT. Recombinant leptin dose-dependently diminished plaque area in the aortic root. Leptin administration within the subphysiological to physiological range diminishes circulating pro-inflammatory IL-6 and MCP-1. Reduction of Il-6 and Mcp-1 gene expression in adipose tissue, as well as decreased adipose tissue macrophage infiltration might contribute. © 2018 BioFactors, 45(1):43-48, 2019.


Subject(s)
Chemokine CCL2/antagonists & inhibitors , Interleukin-6/antagonists & inhibitors , Leptin/genetics , Leptin/pharmacology , Plaque, Atherosclerotic/drug therapy , Adipose Tissue/drug effects , Adipose Tissue/immunology , Adipose Tissue/pathology , Animals , Aorta/drug effects , Aorta/immunology , Aorta/pathology , Cell Movement/drug effects , Chemokine CCL2/blood , Chemokine CCL2/genetics , Chemokine CCL2/immunology , Drug Administration Schedule , Epididymis/drug effects , Epididymis/immunology , Epididymis/pathology , Female , Gene Expression Regulation , Injections, Intraperitoneal , Interleukin-6/blood , Interleukin-6/genetics , Interleukin-6/immunology , Leptin/deficiency , Leptin/immunology , Macrophages/drug effects , Macrophages/immunology , Macrophages/pathology , Male , Mice , Mice, Knockout , Plaque, Atherosclerotic/genetics , Plaque, Atherosclerotic/immunology , Plaque, Atherosclerotic/pathology , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/genetics , RNA, Messenger/immunology , RNA, Messenger/metabolism , Receptors, LDL/deficiency , Receptors, LDL/genetics , Receptors, LDL/immunology , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/pharmacology , Signal Transduction
7.
Dev Comp Immunol ; 92: 29-34, 2019 03.
Article in English | MEDLINE | ID: mdl-30408492

ABSTRACT

C-type lectins (CTLs) are a group of pattern recognition receptors (PRRs) that contain carbohydrate recognition domains and play important roles in innate immunity. CTLs that contain an additional low-density lipoprotein receptor (LDLR) class A domain (LdlrCTL) have been identified in many crustaceans, but their functions in immune responses are mostly unknown. In this study, a novel LdlrCTL was identified from pacific white shrimp Litopenaeus vannamei (LvLdlrCTL), which showed high homology with previously reported crustacean LdlrCTLs. LvLdlrCTL was highly expressed in hemocytes and its expression was up-regulated after immune stimulations. Silencing of LdlrCTL significantly promoted infection of shrimp by Vibrio parahaemolyticus but inhibited infection by white spot syndrome virus (WSSV), suggesting that LdlrCTL could play opposite roles in antibacterial and antiviral responses. LdlrCTL exhibited agglutination activity against bacteria and fungi and could potentiate the phagocytosis of hemocytes. Moreover, the expression of many immune effector genes and signalling pathway components was significantly changed in LdlrCTL-silenced shrimp, indicating that LdlrCTL could be involved in immune regulation.


Subject(s)
Arthropod Proteins/metabolism , DNA Virus Infections/immunology , Hemocytes/physiology , Penaeidae/immunology , Receptors, LDL/metabolism , Vibrio Infections/immunology , Vibrio parahaemolyticus/physiology , White spot syndrome virus 1/physiology , Animals , Arthropod Proteins/genetics , Arthropod Proteins/immunology , Immunity, Innate , Phagocytosis , Protein Domains/genetics , Protein Domains/immunology , RNA, Small Interfering/genetics , Receptors, LDL/genetics , Receptors, LDL/immunology , Receptors, Pattern Recognition/genetics , Sequence Alignment
8.
Cardiovasc Res ; 112(3): 656-668, 2016 12.
Article in English | MEDLINE | ID: mdl-27979811

ABSTRACT

AIMS: The secretion of enzymatically active heparanase (HepA) has been implicated as an essential metabolic adaptation in the heart following diabetes. However, the regulation and function of the enzymatically inactive heparanase (HepL) remain poorly understood. We hypothesized that in response to high glucose (HG) and secretion of HepL from the endothelial cell (EC), HepL uptake and function can protect the cardiomyocyte by modifying its cell death signature. METHODS AND RESULTS: HG promoted both HepL and HepA secretion from microvascular (rat heart micro vessel endothelial cells, RHMEC) and macrovascular (rat aortic endothelial cells, RAOEC) EC. However, only RAOEC were capable of HepL reuptake. This occurred through a low-density lipoprotein receptor-related protein 1 (LRP1) dependent mechanism, as LRP1 inhibition using small interfering RNA (siRNA), receptor-associated protein, or an LRP1 neutralizing antibody significantly reduced uptake. In cardiomyocytes, which have a negligible amount of heparanase gene expression, LRP1 also participated in the uptake of HepL. Exogenous addition of HepL to rat cardiomyocytes produced a dramatically altered expression of apoptosis-related genes, and protection against HG and H2O2 induced cell death. Cardiomyocytes from acutely diabetic rats demonstrated a robust increase in LRP1 expression and levels of heparanase, a pro-survival gene signature, and limited evidence of cell death, observations that were not apparent following chronic and progressive diabetes. CONCLUSION: Our results highlight EC-to-cardiomyocyte transfer of heparanase to modulate the cardiomyocyte cell death signature. This mechanism was observed in the acutely diabetic heart, and its interruption following chronic diabetes may contribute towards the development of diabetic cardiomyopathy.


Subject(s)
Apoptosis , Diabetic Cardiomyopathies/enzymology , Endothelial Cells/enzymology , Glucose/metabolism , Glucuronidase/metabolism , Myocytes, Cardiac/enzymology , Paracrine Communication , Animals , Antibodies, Neutralizing/pharmacology , Apoptosis/drug effects , Cells, Cultured , Diabetes Mellitus, Experimental/chemically induced , Diabetic Cardiomyopathies/chemically induced , Diabetic Cardiomyopathies/genetics , Diabetic Cardiomyopathies/pathology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Gene Expression Regulation , Male , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Paracrine Communication/drug effects , RNA Interference , Rats, Sprague-Dawley , Receptors, LDL/antagonists & inhibitors , Receptors, LDL/genetics , Receptors, LDL/immunology , Receptors, LDL/metabolism , Signal Transduction , Streptozocin , Time Factors , Transcriptome , Transfection
9.
Thromb Haemost ; 116(6): 1122-1130, 2016 Nov 30.
Article in English | MEDLINE | ID: mdl-27580629

ABSTRACT

Our previous studies revealed that the interaction of fibrin with the very low density lipoprotein receptor (VLDLR) promotes transendothelial migration of leukocytes and thereby inflammation, and localised the fibrin-binding site to CR-domains 2-4 of this receptor. In the present study, we tested interaction of three anti-VLDLR monoclonal antibodies, mAb 1H10, 1H5, and 5F3, with recombinant fragments of VLDLR containing various combinations of its CR-domains and found that the epitopes for mAb 1H10 and mAb 1H5 overlap with the fibrin-binding site of VLDLR. Based on these findings, we hypothesised that mAb 1H10 and mAb 1H5 should inhibit fibrin-VLDLR interaction and modulate leukocyte transmigration. To test this hypothesis, we first demonstrated that these monoclonal antibodies both have high affinity to the fibrin-binding fragments of the VLDL receptor and efficiently inhibit interaction between the VLDLR-binding fragment of fibrin and the fibrin-binding fragments of VLDLR. Next, in the in vitro experiments using leukocyte transendothelial migration assay we found that both monoclonal antibodies efficiently inhibit leukocyte transmigration induced by fibrin mimetic NDSK-II. Finally, in vivo experiments using mouse model of peritonitis revealed that mAb 1H10 and mAb 1H5 both significantly reduce infiltration of leukocytes into the peritoneum. Furthermore, our experiments using mouse model of myocardial ischemia-reperfusion injury revealed that both monoclonal antibodies significantly reduce myocardial injury induced by ischaemia-reperfusion. Thus, the results obtained indicate that monoclonal antibodies 1H10 and 1H5 are novel specific inhibitors of fibrin-VLDLR-dependent leukocyte transmigration pathway. They may represent potential therapeutics for treatment of fibrin-dependent inflammation including myocardial ischaemia-reperfusion injury.


Subject(s)
Antibodies, Monoclonal/immunology , Fibrin/metabolism , Leukocytes/cytology , Receptors, LDL/immunology , Transendothelial and Transepithelial Migration , Animals , Epitope Mapping , Human Umbilical Vein Endothelial Cells , Humans , Mice, Inbred C57BL , Receptors, LDL/metabolism
10.
Clin Immunol ; 172: 78-82, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27503146

ABSTRACT

In commemoration of Henry Kunkel's 100th birthday, the effect of his legacy on the investigation of hepatitis C virus is recounted. The delineation of a major cross-idiotype (WA) among patients with essential mixed cryoglobulinemia led to the discovery that HCV was the etiologic agent for this disease. Studies of the cryoglobulins led to the discovery that WA RF reacted specifically with HCV-VLDL like particles that on electronmicroscopy and binding studies appeared to be the virion within a lipid shell. This particle mediates cell entry via LDLr and may serve to avoid the immune response by masking the virion. In addition, the WA B cell may be a prognostic marker for cutaneous vasculitis and B cell malignancy in HCV-infected patients. In commemoration of Henry Kunkel's 100th birthday, this is an account of how his legacy had a role in the investigation of hepatitis C virus (HCV) infection. There is a bit of a historical basis for the legacy of this great immunologist having a role in virology. He began his research career studying hepatitis. Later he worked with HCV in his studies of mixed cryoglobulins although he didn't know it at the time. There may also have been a Kunkel historical basis for why he accepted me as fellow in his laboratory considering that my credentials paled in comparison with those of the fellows and PhD students in his laboratory. Like Henry I was drafted into the Navy following my internship, I had had minimal research experience in medical school and only one minor publication, and I had a passion for clinical investigation. It may have been fortuitous that while on active duty at the Bayonne NJ Naval Base I visited Henry Kunkel in my Navy uniform and told him I was interested in studying SLE. I did not know at the time the dramatic role the Navy had played in his career or that one of his major training goals was to teach MDs to use clinical observation as a focus for delineating disease mechanisms in the laboratory. When I started work in the laboratory on discharge from the Navy, the first thing he told me was that it took five years to make a clinical investigator so I might as well get a Rockefeller University PhD while working in his laboratory. I was sure I would leave the laboratory after two years so I declined his offer. I did not leave until six years later!


Subject(s)
Hepatitis C/immunology , B-Lymphocytes/immunology , Cryoglobulinemia/immunology , Hepacivirus , Humans , Receptors, LDL/immunology , Rheumatoid Factor/immunology , Vasculitis/immunology
11.
J Neurochem ; 136(2): 306-15, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26484803

ABSTRACT

Low-density lipoprotein receptors (LDLRs) mediate the uptake of lipoprotein particles into cells, as studied mainly in peripheral tissues. Here, we show that nerve growth factor (NGF) increases LDLR levels in PC6.3 cells and in cultured septal neurons from embryonic rat brain. Study of the mechanisms showed that NGF enhanced transcription of the LDLR gene, acting mainly via Tropomyosin receptor kinase A receptors. Simvastatin, a cholesterol-lowering drug, also increased the LDLR expression in PC6.3 cells. In addition, pro-NGF and pro-brain-derived neurotrophic factor, acting via the p75 neurotrophin receptor (p75NTR) also increased LDLRs. We further observed that Myosin Regulatory Light Chain-Interacting Protein/Inducible Degrader of the LDLR (Mylip/Idol) was down-regulated by pro-NGF, whereas the other LDLR regulator, proprotein convertase subtilisin kexin 9 (PCSK9) was not significantly changed. On the functional side, NGF and pro-NGF increased lipoprotein uptake by neuronal cells as shown using diacetyl-labeled LDL. The addition of serum-derived lipoprotein particles in conjunction with NGF or simvastatin enhanced neurite outgrowth. Collectively, these results show that NGF and simvastatin are able to stimulate lipoprotein uptake by neurons with a positive effect on neurite outgrowth. Increases in LDLRs and lipoprotein particles in neurons could play a functional role during brain development, in neuroregeneration and after brain injuries. Nerve growth factor (NGF) and pro-NGF induce the expression of low-density lipoprotein receptors (LDLRs) in neuronal cells leading to increased LDLR levels. Pro-NGF also down-regulated myosin regulatory light chain-interacting protein/inducible degrader of the LDLR (Mylip/Idol) that is involved in the degradation of LDLRs. NGF acts mainly via Tropomyosin receptor kinase A (TrkA) receptors, whereas pro-NGF stimulates p75 neurotrophin receptor (p75NTR). Elevated LDLRs upon NGF and pro-NGF treatments enhanced lipoprotein uptake by neurons. Addition of LDL particles further led to the stimulation of neurite outgrowth in PC6.3 cells after NGF or simvastatin treatments, suggesting a stimulatory role of lipoproteins on neuronal differentiation. In contrast, pro-NGF had no effect on neurite outgrowth either in the absence or presence of LDL particles. The precise mechanisms by which increased lipoproteins uptake can affect neurite outgrowth warrant further studies.


Subject(s)
Lipoproteins, LDL/metabolism , Neurites/physiology , Neurons/cytology , Neurons/metabolism , Receptors, LDL/metabolism , Receptors, Nerve Growth Factor/metabolism , Animals , Antibodies/pharmacology , Benzoates/pharmacology , Benzylamines/pharmacology , Carbazoles/pharmacology , Cells, Cultured , Culture Media, Serum-Free/pharmacology , Embryo, Mammalian , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Humans , Indole Alkaloids/pharmacology , Nerve Growth Factor/pharmacology , Nerve Growth Factors/pharmacology , Neurites/drug effects , Neurons/drug effects , Protein Precursors/pharmacology , Rats , Rats, Wistar , Receptors, LDL/immunology , Septum of Brain/cytology , Simvastatin/pharmacology
12.
Cell Rep ; 13(1): 132-144, 2015 Oct 06.
Article in English | MEDLINE | ID: mdl-26411684

ABSTRACT

Enhanced glucose utilization can be visualized in atherosclerotic lesions and may reflect a high glycolytic rate in lesional macrophages, but its causative role in plaque progression remains unclear. We observe that the activity of the carbohydrate-responsive element binding protein ChREBP is rapidly downregulated upon TLR4 activation in macrophages. ChREBP inactivation refocuses cellular metabolism to a high redox state favoring enhanced inflammatory responses after TLR4 activation and increased cell death after TLR4 activation or oxidized LDL loading. Targeted deletion of ChREBP in bone marrow cells resulted in accelerated atherosclerosis progression in Ldlr(-/-) mice with increased monocytosis, lesional macrophage accumulation, and plaque necrosis. Thus, ChREBP-dependent macrophage metabolic reprogramming hinders plaque progression and establishes a causative role for leukocyte glucose metabolism in atherosclerosis.


Subject(s)
Atherosclerosis/immunology , Macrophages/immunology , Nuclear Proteins/immunology , Plaque, Atherosclerotic/immunology , Receptors, LDL/immunology , Transcription Factors/immunology , Animals , Apoptosis/drug effects , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Bone Marrow Cells/drug effects , Bone Marrow Cells/immunology , Bone Marrow Cells/pathology , Female , Gene Expression Regulation , Glucose/metabolism , Glycolysis/genetics , Humans , Inflammation , Lipoproteins, LDL/pharmacology , Macrophage Activation/drug effects , Macrophages/drug effects , Macrophages/pathology , Mice , Mice, Knockout , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oxidation-Reduction , Plaque, Atherosclerotic/genetics , Plaque, Atherosclerotic/metabolism , Plaque, Atherosclerotic/pathology , Primary Cell Culture , Receptors, LDL/deficiency , Receptors, LDL/genetics , Signal Transduction , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology , Toll-Like Receptor 4/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
13.
PLoS One ; 10(8): e0136187, 2015.
Article in English | MEDLINE | ID: mdl-26291618

ABSTRACT

The angiotensin converting enzyme 2-angiotensin-(1-7)-Mas axis (ACE2-Ang-(1-7)-Mas axis) is reported to participate in lipid metabolism in kidney, but its precise effects and underlying mechanisms remain unknown. We hypothesized that Ang-(1-7) reduces lipid accumulation and improves renal injury through the low density lipoprotein receptor-sterol regulatory element binding proteins 2-SREBP cleavage activating protein (LDLr-SREBP2-SCAP) system by suppressing inflammation in high fat diet (HFD)-fed mice. In this study, male C57BL/6 mice were randomized into four groups: STD (standard diet)+saline, HFD+saline, HFD+Ang-(1-7) and STD+Ang-(1-7). After 10 weeks of feeding, mice were administered Ang-(1-7) or saline for two weeks. We found that high inflammation status induced by HFD disrupted the LDLr-SREBP2-SCAP feedback system. Treatment of mice fed a high-fat diet with Ang-(1-7) induced significant improvement in inflammatory status, following the downregulation of LDLr, SREBP2 and SCAP, and then, decreased lipid deposition in kidney and improved renal injury. In conclusion, the anti-inflammatory effect of Ang-(1-7) alleviates renal injury triggered by lipid metabolic disorders through a LDLr- SREBP2-SCAP pathway.


Subject(s)
Acute Kidney Injury/drug therapy , Angiotensin I/therapeutic use , Anti-Inflammatory Agents/therapeutic use , Diet, High-Fat/adverse effects , Peptide Fragments/therapeutic use , Receptors, LDL/immunology , Sterol Regulatory Element Binding Protein 2/immunology , Acute Kidney Injury/etiology , Acute Kidney Injury/immunology , Acute Kidney Injury/pathology , Angiotensin I/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Dyslipidemias/drug therapy , Dyslipidemias/etiology , Dyslipidemias/immunology , Dyslipidemias/pathology , Inflammation/drug therapy , Inflammation/etiology , Inflammation/immunology , Inflammation/pathology , Kidney/drug effects , Kidney/immunology , Kidney/pathology , Lipid Metabolism/drug effects , Male , Mice, Inbred C57BL , Peptide Fragments/pharmacology , Signal Transduction/drug effects
14.
Mem. Inst. Oswaldo Cruz ; 110(4): 534-542, 09/06/2015. graf
Article in English | LILACS | ID: lil-748872

ABSTRACT

Hepatitis C virus (HCV) envelope protein 2 (E2) is involved in viral binding to host cells. The aim of this work was to produce recombinant E2B and E2Y HCV proteins in Escherichia coli and Pichia pastoris, respectively, and to study their interactions with low-density lipoprotein receptor (LDLr) and CD81 in human umbilical vein endothelial cells (HUVEC) and the ECV304 bladder carcinoma cell line. To investigate the effects of human LDL and differences in protein structure (glycosylated or not) on binding efficiency, the recombinant proteins were either associated or not associated with lipoproteins before being assayed. The immunoreactivity of the recombinant proteins was analysed using pooled serum samples that were either positive or negative for hepatitis C. The cells were immunophenotyped by LDLr and CD81 using flow cytometry. Binding and binding inhibition assays were performed in the presence of LDL, foetal bovine serum (FCS) and specific antibodies. The results revealed that binding was reduced in the absence of FCS, but that the addition of human LDL rescued and increased binding capacity. In HUVEC cells, the use of antibodies to block LDLr led to a significant reduction in the binding of E2B and E2Y. CD81 antibodies did not affect E2B and E2Y binding. In ECV304 cells, blocking LDLr and CD81 produced similar effects, but they were not as marked as those that were observed in HUVEC cells. In conclusion, recombinant HCV E2 is dependent on LDL for its ability to bind to LDLr in HUVEC and ECV304 cells. These findings are relevant because E2 acts to anchor HCV to host cells; therefore, high blood levels of LDL could enhance viral infectivity in chronic hepatitis C patients.


Subject(s)
Animals , Cattle , Humans , /physiology , Endothelial Cells/virology , Hepacivirus/immunology , Receptors, LDL/physiology , Viral Envelope Proteins/physiology , /immunology , Cell Line , Escherichia coli , Endothelial Cells/immunology , Flow Cytometry , Membrane Proteins , Pichia , Recombinant Proteins , Receptors, LDL/immunology
15.
Mem Inst Oswaldo Cruz ; 110(4): 534-42, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26018451

ABSTRACT

Hepatitis C virus (HCV) envelope protein 2 (E2) is involved in viral binding to host cells. The aim of this work was to produce recombinant E2B and E2Y HCV proteins in Escherichia coli and Pichia pastoris, respectively, and to study their interactions with low-density lipoprotein receptor (LDLr) and CD81 in human umbilical vein endothelial cells (HUVEC) and the ECV304 bladder carcinoma cell line. To investigate the effects of human LDL and differences in protein structure (glycosylated or not) on binding efficiency, the recombinant proteins were either associated or not associated with lipoproteins before being assayed. The immunoreactivity of the recombinant proteins was analysed using pooled serum samples that were either positive or negative for hepatitis C. The cells were immunophenotyped by LDLr and CD81 using flow cytometry. Binding and binding inhibition assays were performed in the presence of LDL, foetal bovine serum (FCS) and specific antibodies. The results revealed that binding was reduced in the absence of FCS, but that the addition of human LDL rescued and increased binding capacity. In HUVEC cells, the use of antibodies to block LDLr led to a significant reduction in the binding of E2B and E2Y. CD81 antibodies did not affect E2B and E2Y binding. In ECV304 cells, blocking LDLr and CD81 produced similar effects, but they were not as marked as those that were observed in HUVEC cells. In conclusion, recombinant HCV E2 is dependent on LDL for its ability to bind to LDLr in HUVEC and ECV304 cells. These findings are relevant because E2 acts to anchor HCV to host cells; therefore, high blood levels of LDL could enhance viral infectivity in chronic hepatitis C patients.


Subject(s)
Endothelial Cells/virology , Hepacivirus/immunology , Receptors, LDL/physiology , Tetraspanin 28/physiology , Viral Envelope Proteins/physiology , Animals , Cattle , Cell Line , Endothelial Cells/immunology , Escherichia coli , Flow Cytometry , Humans , Membrane Proteins , Pichia , Receptors, LDL/immunology , Recombinant Proteins , Tetraspanin 28/immunology
16.
Oncotarget ; 6(6): 3722-36, 2015 Feb 28.
Article in English | MEDLINE | ID: mdl-25669985

ABSTRACT

Mineral dust-induced gene (mdig, also named Mina53) was first identified from alveolar macrophages of the coal miners with chronic lung inflammation or fibrosis, but how this gene is involved in lung diseases is poorly understood. Here we show that heterozygotic knockout of mdig (mdig+/-) ameliorates silica-induced lung fibrosis by altering the balance between Th17 cells and Treg cells. Relative to the wild type (WT) mice, infiltration of the macrophages and Th17 cells was reduced in lungs from silica-exposed mdig+/- mice. In contrast, an increased infiltration of the T regulatory (Treg) cells to the lung intestitium was observed in the mdig+/- mice treated with silica. Both the number of Th17 cells in the lung lymph nodes and the level of IL-17 in the bronchoalveolar lavage fluids were decreased in the mdig+/- mice in response to silica. Thus, these results suggest that mdig may contribute to silica-induced lung fibrosis by altering the balance between Th17 and Treg cells. Genetic deficiency of mdig impairs Th17 cell infiltration and function, but favors infiltration of the Treg cells, the immune suppressive T cells that are able to limit the inflammatory responses by repressing the Th17 cells and macrophages.


Subject(s)
Pulmonary Fibrosis/immunology , Receptors, LDL/immunology , Silicon Dioxide/toxicity , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Animals , LDL-Receptor Related Proteins , Mice , Mice, Inbred C57BL , Mice, Knockout , Pulmonary Fibrosis/genetics , Receptors, LDL/deficiency , Receptors, LDL/genetics , Silicon Dioxide/immunology
17.
Circulation ; 130(16): 1363-73, 2014 Oct 14.
Article in English | MEDLINE | ID: mdl-25223984

ABSTRACT

BACKGROUND: Plasmacytoid dendritic cells (pDCs) bridge innate and adaptive immune responses and are important regulators of immuno-inflammatory diseases. However, their role in atherosclerosis remains elusive. METHODS AND RESULTS: Here, we used genetic approaches to investigate the role of pDCs in atherosclerosis. Selective pDC deficiency in vivo was achieved using CD11c-Cre × Tcf4(-/flox) bone marrow transplanted into Ldlr(-/-) mice. Compared with control Ldlr(-/-) chimeric mice, CD11c-Cre × Tcf4(-/flox) mice had reduced atherosclerosis levels. To begin to understand the mechanisms by which pDCs regulate atherosclerosis, we studied chimeric Ldlr(-/-) mice with selective MHCII deficiency on pDCs. Significantly, these mice also developed reduced atherosclerosis compared with controls without reductions in pDC numbers or changes in conventional DCs. MHCII-deficient pDCs showed defective stimulation of apolipoprotein B100-specific CD4(+) T cells in response to native low-density lipoprotein, whereas production of interferon-α was not affected. Finally, the atheroprotective effect of selective MHCII deficiency in pDCs was associated with significant reductions of proatherogenic T cell-derived interferon-γ and lesional T cell infiltration, and was abrogated in CD4(+) T cell-depleted animals. CONCLUSIONS: This study supports a proatherogenic role for pDCs in murine atherosclerosis and identifies a critical role for MHCII-restricted antigen presentation by pDCs in driving proatherogenic T cell immunity.


Subject(s)
Antigen-Presenting Cells/immunology , Atherosclerosis/immunology , Atherosclerosis/pathology , CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Histocompatibility Antigens Class II/immunology , Adaptive Immunity/immunology , Animals , Aorta/cytology , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/immunology , CD4-Positive T-Lymphocytes/cytology , Cell Communication/immunology , Cells, Cultured , Dendritic Cells/cytology , Flow Cytometry , Mice, Inbred C57BL , Mice, Knockout , Receptors, LDL/genetics , Receptors, LDL/immunology , Transcription Factor 4
18.
J Immunol ; 193(2): 783-96, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24928988

ABSTRACT

Human ficolin-2 (L-ficolin/p35) is a lectin-complement pathway activator that is present in normal human plasma and is associated with infectious diseases; however, little is known regarding the roles and mechanisms of ficolin-2 during chronic hepatitis C virus (HCV) infection. In this study, we found that ficolin-2 inhibits the entry of HCV at an early stage of viral infection, regardless of the viral genotype. Ficolin-2 neutralized and inhibited the initial attachment and infection of HCV by binding to the HCV envelope surface glycoproteins E1 and E2, blocking HCV attachment to low-density lipoprotein receptor (LDLR) and scavenger receptor B1, and weakly interfering with CD81 receptor attachment. However, no interference with claudin-1 and occludin receptor attachment was observed. The C-terminal fibrinogen domain (201-313 aa) of ficolin-2 was identified as the critical binding region for the HCV-E1-E2 N-glycans, playing a critical role in the anti-HCV activity. More importantly, we found that apolipoprotein E (ApoE)3, which is enriched in the low-density fractions of HCV RNA-containing particles, promotes HCV infection and inhibits ficolin-2-mediated antiviral activity. ApoE3, but not ApoE2 and ApoE4, blocked the interaction between ficolin-2 and HCV-E2. Our data suggest that the HCV entry inhibitor ficolin-2 is a novel and promising antiviral innate immune molecule, whereas ApoE3 blocks the effect of ficolin-2 and mediates an immune escape mechanism during chronic HCV infection. HCV may be neutralized using compounds directed against the lipoprotein moiety of the viral particle, and ApoE3 may be a new target to combat HCV infection.


Subject(s)
Apolipoprotein E3/immunology , Hepacivirus/immunology , Lectins/immunology , Tumor Escape/immunology , Apolipoprotein E3/genetics , Apolipoprotein E3/metabolism , Binding, Competitive/immunology , Blotting, Western , Cell Line, Tumor , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , HeLa Cells , Hepacivirus/genetics , Hepacivirus/physiology , Host-Pathogen Interactions/immunology , Humans , Lectins/genetics , Lectins/metabolism , Mannans/immunology , Mannans/metabolism , Microscopy, Confocal , Polysaccharides/immunology , Polysaccharides/metabolism , Protein Binding/immunology , RNA Interference , Receptors, LDL/genetics , Receptors, LDL/immunology , Receptors, LDL/metabolism , Scavenger Receptors, Class B/genetics , Scavenger Receptors, Class B/immunology , Scavenger Receptors, Class B/metabolism , Tetraspanin 28/genetics , Tetraspanin 28/immunology , Tetraspanin 28/metabolism , Tumor Escape/genetics , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology , Viral Envelope Proteins/metabolism , Ficolins
19.
J Immunol ; 192(10): 4497-509, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24733846

ABSTRACT

The very low density lipoprotein receptor (VLDLR) is a member of the low-density lipoprotein receptor family that binds multiple ligands and plays a key role in brain development. Although the VLDLR mediates pleiotropic biological processes, only a limited amount of information is available regarding its role in adaptive immunity. In this study, we identify an important role for the VLDLR in attenuating house dust mite (HDM)-induced airway inflammation in experimental murine asthma. We show that HDM-challenged Vldlr(-/-) mice have augmented eosinophilic and lymphocytic airway inflammation with increases in Th2 cytokines, C-C chemokines, IgE production, and mucous cell metaplasia. A genome-wide analysis of the lung transcriptome identified that mRNA levels of CD209e (DC-SIGNR4), a murine homolog of DC-SIGN, were increased in the lungs of HDM-challenged Vldlr(-/-) mice, which suggested that the VLDLR might modify dendritic cell (DC) function. Consistent with this, VLDLR expression by human monocyte-derived DCs was increased by HDM stimulation. In addition, 55% of peripheral blood CD11c(+) DCs from individuals with allergy expressed VLDLR under basal conditions. Lastly, the adoptive transfer of HDM-pulsed, CD11c(+) bone marrow-derived DCs (BMDCs) from Vldlr(-/-) mice to the airways of wild type recipient mice induced augmented eosinophilic and lymphocytic airway inflammation upon HDM challenge with increases in Th2 cytokines, C-C chemokines, IgE production, and mucous cell metaplasia, as compared with the adoptive transfer of HDM-pulsed, CD11c(+) BMDCs from wild type mice. Collectively, these results identify a novel role for the VLDLR as a negative regulator of DC-mediated adaptive immune responses in HDM-induced allergic airway inflammation.


Subject(s)
Adaptive Immunity , Dendritic Cells/immunology , Pyroglyphidae , Receptors, LDL/immunology , Respiratory Hypersensitivity/immunology , Animals , CD11c Antigen/genetics , CD11c Antigen/immunology , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/immunology , Cytokines/genetics , Cytokines/immunology , Dendritic Cells/pathology , Eosinophils/immunology , Eosinophils/pathology , Female , Genome-Wide Association Study , Humans , Immunoglobulin E/genetics , Immunoglobulin E/immunology , Lectins, C-Type/genetics , Lectins, C-Type/immunology , Male , Mice , Mice, Knockout , Receptors, Cell Surface/genetics , Receptors, Cell Surface/immunology , Receptors, LDL/genetics , Respiratory Hypersensitivity/genetics , Respiratory Hypersensitivity/pathology , Th2 Cells/immunology , Th2 Cells/pathology
20.
Clin Immunol ; 152(1-2): 90-100, 2014.
Article in English | MEDLINE | ID: mdl-24650778

ABSTRACT

In this study, the capacity for t-PA to affect T cell-brain microvascular endothelial cell adhesion by acting as a cytokine was investigated. Following the treatment of a brain-derived endothelial cell line, bEnd.3, with various concentrations of t-PA, adhesion and transwell migration assays were performed. In the presence of t-PA, enhanced adhesion of T cells to bEnd.3 cells was observed. Using western blot analysis, an increase in ICAM-1 expression was detected for both t-PA-treated bEnd.3 cells and bEnd.3 cells treated with a non-enzymatic form of t-PA. In contrast, when LRP1 was blocked using a specific antibody, upregulation of ICAM-1 was inhibited and cAMP-PKA signaling was affected. Furthermore, using an EAE mouse model, administration of t-PA was associated with an increase in ICAM-1 expression by brain endothelial cells. Taken together, these findings suggest that t-PA can induce ICAM-1 expression in brain microvascular endothelial cells, and this may promote the development of EAE.


Subject(s)
Brain/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Intercellular Adhesion Molecule-1/biosynthesis , Multiple Sclerosis/immunology , Tissue Plasminogen Activator/physiology , Animals , Antibodies, Monoclonal/immunology , Brain/blood supply , Cell Adhesion/drug effects , Cell Line, Transformed , Cell Movement/drug effects , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Cytokines/immunology , Cytokines/metabolism , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/pathology , Endothelial Cells/immunology , Endothelium, Vascular/cytology , Endothelium, Vascular/immunology , Endothelium, Vascular/metabolism , Female , Intercellular Adhesion Molecule-1/metabolism , Low Density Lipoprotein Receptor-Related Protein-1 , Mice , Mice, Inbred C57BL , Multiple Sclerosis/pathology , Receptors, LDL/immunology , Signal Transduction/immunology , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Tissue Plasminogen Activator/administration & dosage , Tumor Suppressor Proteins/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...