Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 278
Filter
1.
Clin Sci (Lond) ; 135(11): 1389-1408, 2021 06 11.
Article in English | MEDLINE | ID: mdl-34086048

ABSTRACT

The immune system protects the body against harm by inducing inflammation. During the immune response, cells of the immune system get activated, divided and differentiated in order to eliminate the danger signal. This process relies on the metabolic reprogramming of both catabolic and anabolic pathways not only to produce energy in the form of ATP but also to generate metabolites that exert key functions in controlling the response. Equally important to mounting an appropriate effector response is the process of immune resolution, as uncontrolled inflammation is implicated in the pathogenesis of many human diseases, including allergy, chronic inflammation and cancer. In this review, we aim to introduce the reader to the field of cholesterol immunometabolism and discuss how both metabolites arising from the pathway and cholesterol homeostasis are able to impact innate and adaptive immune cells, staging cholesterol homeostasis at the centre of an adequate immune response. We also review evidence that demonstrates the clear impact that cholesterol metabolism has in both the induction and the resolution of the inflammatory response. Finally, we propose that emerging data in this field not only increase our understanding of immunometabolism but also provide new tools for monitoring and intervening in human diseases, where controlling and/or modifying inflammation is desirable.


Subject(s)
Cholesterol/metabolism , Immune System/immunology , Inflammation/metabolism , Lipid Metabolism/immunology , Animals , Cholesterol/immunology , Energy Metabolism/immunology , Energy Metabolism/physiology , Humans , Inflammation/immunology , Signal Transduction/immunology , Signal Transduction/physiology
2.
Proc Natl Acad Sci U S A ; 118(21)2021 05 25.
Article in English | MEDLINE | ID: mdl-34006637

ABSTRACT

The liver X receptor (LXR) is a key transcriptional regulator of cholesterol, fatty acid, and phospholipid metabolism. Dynamic remodeling of immunometabolic pathways, including lipid metabolism, is a crucial step in T cell activation. Here, we explored the role of LXR-regulated metabolic processes in primary human CD4+ T cells and their role in controlling plasma membrane lipids (glycosphingolipids and cholesterol), which strongly influence T cell immune signaling and function. Crucially, we identified the glycosphingolipid biosynthesis enzyme glucosylceramide synthase as a direct transcriptional LXR target. LXR activation by agonist GW3965 or endogenous oxysterol ligands significantly altered the glycosphingolipid:cholesterol balance in the plasma membrane by increasing glycosphingolipid levels and reducing cholesterol. Consequently, LXR activation lowered plasma membrane lipid order (stability), and an LXR antagonist could block this effect. LXR stimulation also reduced lipid order at the immune synapse and accelerated activation of proximal T cell signaling molecules. Ultimately, LXR activation dampened proinflammatory T cell function. Finally, compared with responder T cells, regulatory T cells had a distinct pattern of LXR target gene expression corresponding to reduced lipid order. This suggests LXR-driven lipid metabolism could contribute to functional specialization of these T cell subsets. Overall, we report a mode of action for LXR in T cells involving the regulation of glycosphingolipid and cholesterol metabolism and demonstrate its relevance in modulating T cell function.


Subject(s)
Cholesterol/genetics , Glycosphingolipids/genetics , Liver X Receptors/immunology , T-Lymphocytes/immunology , Adolescent , Adult , Benzoates/pharmacology , Benzylamines/pharmacology , Cell Membrane , Cholesterol/immunology , Female , Glucosyltransferases/genetics , Glycosphingolipids/biosynthesis , Glycosphingolipids/immunology , Humans , Immunological Synapses/drug effects , Immunological Synapses/genetics , Ligands , Lipid Metabolism/genetics , Lipid Metabolism/immunology , Liver X Receptors/agonists , Liver X Receptors/antagonists & inhibitors , Liver X Receptors/genetics , Male , Metabolic Networks and Pathways/immunology , Middle Aged , Oxysterols/pharmacology , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , T-Lymphocytes/metabolism , Young Adult
3.
Sci Rep ; 11(1): 4923, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33649397

ABSTRACT

In atherosclerotic lesions, blood-derived monocytes differentiate into distinct macrophage subpopulations, and further into cholesterol-filled foam cells under a complex milieu of cytokines, which also contains macrophage-colony stimulating factor (M-CSF) and granulocyte-macrophage-colony stimulating factor (GM-CSF). Here we generated human macrophages in the presence of either M-CSF or GM-CSF to obtain M-MØ and GM-MØ, respectively. The macrophages were converted into cholesterol-loaded foam cells by incubating them with acetyl-LDL, and their atheroinflammatory gene expression profiles were then assessed. Compared with GM-MØ, the M-MØ expressed higher levels of CD36, SRA1, and ACAT1, and also exhibited a greater ability to take up acetyl-LDL, esterify cholesterol, and become converted to foam cells. M-MØ foam cells expressed higher levels of ABCA1 and ABCG1, and, correspondingly, exhibited higher rates of cholesterol efflux to apoA-I and HDL2. Cholesterol loading of M-MØ strongly suppressed the high baseline expression of CCL2, whereas in GM-MØ the low baseline expression CCL2 remained unchanged during cholesterol loading. The expression of TNFA, IL1B, and CXCL8 were reduced in LPS-activated macrophage foam cells of either subtype. In summary, cholesterol loading converged the CSF-dependent expression of key genes related to intracellular cholesterol balance and inflammation. These findings suggest that transformation of CSF-polarized macrophages into foam cells may reduce their atheroinflammatory potential in atherogenesis.


Subject(s)
Cholesterol/immunology , Colony-Stimulating Factors/immunology , Macrophages , Monocytes , T-Lymphocytes , Atherosclerosis/immunology , Cells, Cultured , Humans , Inflammation/immunology , Macrophages/cytology , Macrophages/immunology , Monocytes/cytology , Monocytes/immunology , Primary Cell Culture , T-Lymphocytes/cytology , T-Lymphocytes/immunology
4.
Angew Chem Int Ed Engl ; 60(17): 9467-9473, 2021 04 19.
Article in English | MEDLINE | ID: mdl-33464672

ABSTRACT

The search for vaccines that protect from severe morbidity and mortality because of infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the virus that causes coronavirus disease 2019 (COVID-19) is a race against the clock and the virus. Here we describe an amphiphilic imidazoquinoline (IMDQ-PEG-CHOL) TLR7/8 adjuvant, consisting of an imidazoquinoline conjugated to the chain end of a cholesterol-poly(ethylene glycol) macromolecular amphiphile. It is water-soluble and exhibits massive translocation to lymph nodes upon local administration through binding to albumin, affording localized innate immune activation and reduction in systemic inflammation. The adjuvanticity of IMDQ-PEG-CHOL was validated in a licensed vaccine setting (quadrivalent influenza vaccine) and an experimental trimeric recombinant SARS-CoV-2 spike protein vaccine, showing robust IgG2a and IgG1 antibody titers in mice that could neutralize viral infection in vitro and in vivo in a mouse model.


Subject(s)
Adjuvants, Immunologic/therapeutic use , COVID-19 Vaccines/therapeutic use , COVID-19/prevention & control , Imidazoles/therapeutic use , Immunity, Innate/drug effects , Quinolines/therapeutic use , Animals , COVID-19 Vaccines/immunology , Cholesterol/analogs & derivatives , Cholesterol/immunology , Cholesterol/therapeutic use , Female , Humans , Imidazoles/immunology , Influenza A Virus, H1N1 Subtype/drug effects , Influenza Vaccines/immunology , Influenza Vaccines/therapeutic use , Influenza, Human/prevention & control , Membrane Glycoproteins/agonists , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Polyethylene Glycols/therapeutic use , Quinolines/immunology , Recombinant Proteins/immunology , SARS-CoV-2/drug effects , Spike Glycoprotein, Coronavirus/immunology , Surface-Active Agents/therapeutic use , Toll-Like Receptor 7/agonists , Toll-Like Receptor 8/agonists
5.
Front Immunol ; 12: 658895, 2021.
Article in English | MEDLINE | ID: mdl-35126343

ABSTRACT

The innate immune system senses viral and bacterial ribonucleic acid (RNA) via pattern recognition receptors (PRR) leading to subsequent activation of the immune system. One group of RNA sensors is formed by endosomal/lysosomal Toll-like receptors (TLR) such as TLR7 and TLR8. During viral or bacterial infection, immunostimulatory RNA is part of the pathogen reaching the endosomal/lysosomal compartment after cellular uptake. Synthetic single-stranded or double-stranded oligoribonucleotides (ORN) can mimic RNA from pathogens and are widely used as activating ligands for TLR7 and TLR8. However, one limitation in the use of synthetic ORN driven immune stimulation is the need for transfection reagents for RNA delivery into cells. Here we demonstrate that the conjugation of cholesterol to a double-stranded version of immunostimulatory RNA40 strongly enhanced RNA uptake into monocytes and plasmacytoid dendritic cells when compared to naked RNA. Cholesterol-conjugated RNA (RNA-chol) formed nanoparticles that were superior to RNA-liposomes complexes in regard to induction of type I interferon from human and murine plasmacytoid dendritic cells as well as proinflammatory cytokine production (e.g. TNF-α, IL12p70 or IL-6) in human monocytes. Furthermore, the RNA40-chol induced cytokines in human monocyte cultures supported TH1 and TFH cell differentiation underscoring a strong adjuvant function of RNA-chol nanoparticles for adaptive immune responses. In summary, cholesterol-conjugated immunostimulatory RNA forms nanoparticles and functions as a potent immune adjuvant in human and murine immune cells. It further simplifies the use of immunostimulatory RNA by avoiding the need for liposomal transfection reagents.


Subject(s)
Cholesterol/immunology , Immunity, Innate/immunology , Membrane Glycoproteins/immunology , Nanoparticles/administration & dosage , RNA/immunology , Toll-Like Receptor 7/immunology , Toll-Like Receptor 8/immunology , Adjuvants, Immunologic/pharmacology , Animals , Cells, Cultured , Cytokines/immunology , Dendritic Cells/immunology , Humans , Leukocytes, Mononuclear/immunology , Mice , Mice, Inbred C57BL , Monocytes/immunology , Oligoribonucleotides/immunology
6.
Front Immunol ; 11: 584303, 2020.
Article in English | MEDLINE | ID: mdl-33224146

ABSTRACT

The interplay between cellular stress and immune response can be variable and sometimes contradictory. The mechanisms by which stress-activated pathways regulate the inflammatory response to a pathogen, in autoimmunity or during cancer progression remain unclear in many aspects, despite our recent knowledge of the signalling and transcriptional pathways involved in these diseases. In this context, over the last decade many studies demonstrated that cholesterol metabolism is an important checkpoint for immune homeostasis and cancer progression. Indeed, cholesterol is actively metabolized and can regulate, through its mobilization and/or production of active derivatives, many aspects of immunity and inflammation. Moreover, accumulation of cholesterol has been described in cancer cells, indicating metabolic addiction. The nuclear receptors liver-X-receptors (LXRs) are important regulators of intracellular cholesterol and lipids homeostasis. They have also key regulatory roles in immune response, as they can regulate inflammation, innate and adaptive immunity. Moreover, activation of LXRs has been reported to affect the proliferation and survival of different cancer cell types that show altered metabolic pathways and accumulation of cholesterol. In this minireview we will give an overview of the recent understandings about the mechanisms through which LXRs regulate inflammation, autoimmunity, and cancer, and the therapeutic potential for future treatment of these diseases through modulation of cholesterol metabolism.


Subject(s)
Autoimmunity/immunology , Cholesterol/metabolism , Inflammation/metabolism , Liver X Receptors/metabolism , Neoplasms/metabolism , Adaptive Immunity/immunology , Animals , Cholesterol/immunology , Humans , Immunity, Innate/immunology , Inflammation/immunology , Lipid Metabolism/immunology , Metabolic Networks and Pathways/immunology , Signal Transduction/immunology
7.
Front Immunol ; 11: 1675, 2020.
Article in English | MEDLINE | ID: mdl-32849582

ABSTRACT

Clostridium difficile, an obligate anaerobic gram-positive bacillus, generates spores and is commonly found colonizing the human gut. Patients with C. difficile infection (CDI) often exhibit clinical manifestations of pseudomembranous colitis or antibiotic-associated diarrhea. Surface layer proteins (SLPs) are the most abundant proteins in the C. difficile cell wall, suggesting that they might involve in immune recognition. Our previous results demonstrated that C. difficile triggers inflammasome activation. Here, we found SLPs as well as C. difficile induced inflammasome activation, and in a dose-dependent manner. In addition, the cholesterol-rich microdomains on the cell membrane (also referred to as lipid rafts) are thought to be crucial for bacterial adhesion and signal transduction. We demonstrated that lipid rafts participated in C. difficile SLPs binding to the cell membrane. Fluorescence microscopy showed that membrane cholesterol depletion by methyl-ß-cyclodextrin (MßCD) reduced the association of SLPs with the cell surface. The coalescence of SLPs in the cholesterol-rich microdomains was confirmed in C. difficile-infected cells. Furthermore, the inflammasome activations induced by SLPs or C. difficile were abrogated by MßCD. Our results demonstrate that SLPs recruit the lipid rafts, which may be a key step for C. difficile colonization and inducing inflammasome activation.


Subject(s)
Cholesterol/metabolism , Clostridium Infections/metabolism , Inflammasomes/immunology , Membrane Glycoproteins/metabolism , Membrane Microdomains/metabolism , Cholesterol/immunology , Clostridioides difficile/immunology , Clostridioides difficile/pathogenicity , Clostridium Infections/immunology , Humans , Inflammasomes/metabolism , Membrane Lipids/immunology , Membrane Lipids/metabolism , Membrane Microdomains/immunology , Protein Binding , THP-1 Cells
8.
Exp Cell Res ; 395(1): 112169, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32653410

ABSTRACT

It has been reported that cholesterol-lowing agents can ameliorate severity of myocarditis. However, the beneficial effect of the agents has been claimed to be independent of cholesterol reduction as there is no significant change in the plasma cholesterol level in myocarditis. In the present study, using experimental autoimmune myocarditis (EAM) rats as an animal model, we demonstrated that EAM induced elevation of cholesterol level and impaired cholesterol efflux capacity in the cardiac tissue. Moreover, serum high-density lipoprotein (HDL) content was reduced and HDL function associated protein Paraoxonase 1 (PON1) activity was decreased. Besides, the major structural protein within HDL, Apolipoprotein A1 (ApoA1) expression in the cardiac tissues was significantly reduced while the level of serum ApoA1 was not significantly altered. Importantly, cholesterol depleting agent methyl-ß-cyclodextrin (MßCD) alleviated the development of EAM, as monitored by decreased ratio of heart weight to body weight (HW/BW), decreased infiltration of inflammatory cells and collagen deposition, improved cardiac function, reduced expression of apoptosis-related protein Bax, Fas, FasL and caspase-3 and increased level of anti-apoptotic protein Bcl-2. These results suggest that reduction of cholesterol level in cardiac tissue could suppress EAM-induced cardiac apoptosis through both intrinsic and extrinsic apoptotic pathways.


Subject(s)
Apoptosis/physiology , Autoimmune Diseases/immunology , Cholesterol/metabolism , Hypercholesterolemia/metabolism , Myocarditis/metabolism , Animals , Caspase 3/metabolism , Cholesterol/immunology , Cytokines/immunology , Cytokines/metabolism , Disease Models, Animal , Male , Myocarditis/immunology , Myocardium/metabolism , Rats
9.
J Clin Invest ; 130(4): 1683-1698, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32091410

ABSTRACT

Protein arginine methyltransferase 5 (PRMT5) catalyzes symmetric dimethylation (SDM) of arginine, a posttranslational modification involved in oncogenesis and embryonic development. However, the role and mechanisms by which PRMT5 modulates Th cell polarization and autoimmune disease have not yet been elucidated. Here, we found that PRMT5 promoted SREBP1 SDM and the induction of cholesterol biosynthetic pathway enzymes that produce retinoid-related orphan receptor (ROR) agonists that activate RORγt. Specific loss of PRMT5 in the CD4+ Th cell compartment suppressed Th17 differentiation and protected mice from developing experimental autoimmune encephalomyelitis (EAE). We also found that PRMT5 controlled thymic and peripheral homeostasis in the CD4+ Th cell life cycle and invariant NK (iNK) T cell development and CD8+ T cell maintenance. This work demonstrates that PRMT5 expression in recently activated T cells is necessary for the cholesterol biosynthesis metabolic gene expression program that generates RORγt agonistic activity and promotes Th17 differentiation and EAE. These results point to Th PRMT5 and its downstream cholesterol biosynthesis pathway as promising therapeutic targets in Th17-mediated diseases.


Subject(s)
Autoimmunity , Cell Differentiation/immunology , Cholesterol/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Protein-Arginine N-Methyltransferases/immunology , Th17 Cells/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Cell Differentiation/genetics , Cholesterol/genetics , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Mice , Mice, Transgenic , Natural Killer T-Cells/immunology , Natural Killer T-Cells/pathology , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology , Protein-Arginine N-Methyltransferases/genetics , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 1/immunology , Th17 Cells/pathology
10.
Int J Mol Sci ; 20(18)2019 Sep 12.
Article in English | MEDLINE | ID: mdl-31547302

ABSTRACT

Cholesterol is a member of the sterol family that plays essential roles in biological processes, including cell membrane stability and myelin formation. Cholesterol can be metabolized into several molecules including bile acids, hormones, and oxysterols. Studies from the last few decades have demonstrated that oxysterols are not only active metabolites but are further involved in the modulation of immune responses. Liver X Receptors (LXRs), nuclear receptors for oxysterols, are important for cholesterol homeostasis and regulation of inflammatory response but are still poorly characterized during autoimmune diseases. Here we review the current knowledge about the role of oxysterols during autoimmune conditions and focus on the implication of LXR-dependent and LXR-independent pathways. We further highlight the importance of these pathways in particular during central nervous system (CNS) autoimmunity and inflammatory bowel diseases (IBD) in both experimental models and human studies. Finally, we discuss our vision about future applications and research on oxysterols related to autoimmunity.


Subject(s)
Autoimmune Diseases/immunology , Autoimmunity , Oxysterols/immunology , Animals , Central Nervous System Diseases/immunology , Cholesterol/immunology , Humans , Inflammatory Bowel Diseases/immunology , Liver X Receptors/immunology
11.
Int J Mol Sci ; 20(18)2019 Sep 14.
Article in English | MEDLINE | ID: mdl-31540125

ABSTRACT

Natural killer T (NKT) cells are a distinct subset of lymphocytes that bridge the innate and adaptive immune response and can be divided into type I invariant NKT cells (iNKT) and type II NKT cells. The objective of this study is to examine the effects of NKT cell on lipid metabolism and the initiation and progression of atherosclerosis in LDL receptor deficient (LDLR-/-) mice. Mice were fed an atherogenic diet for 4 or 8 weeks and plasma lipids, lipoproteins, and atherosclerosis were measured. The selective absence of iNKT cells in Jα18-/-LDLR-/- mice led to an increase in plasma cholesterol levels in female mice. Transgenic Vα14tg/LDLR-/- mice with elevated numbers of iNKT cells had increased late atherosclerosis of the innominate artery, though absence of either iNKT cells or all NKT cells and other CD1d expressing cells had varying effects on atherosclerotic lesion burden in the ascending aortic arch and aortic root. These studies not only highlight the potential modulatory role played by NKT cells in atherosclerosis and lipid metabolism, but also raise the possibility that divergent roles may be played by iNKT and CD1d restricted cells such as type II NKT cells or other CD1d expressing cells.


Subject(s)
Antigens, CD1d/genetics , Atherosclerosis/immunology , Lipid Metabolism/immunology , Natural Killer T-Cells/immunology , Adaptive Immunity , Animals , Antigens, CD1d/immunology , Antigens, CD1d/metabolism , Aorta/pathology , Atherosclerosis/pathology , Cholesterol/blood , Cholesterol/immunology , Female , Lipoproteins/blood , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Natural Killer T-Cells/metabolism , Receptors, LDL/genetics
12.
Front Immunol ; 10: 1404, 2019.
Article in English | MEDLINE | ID: mdl-31316501

ABSTRACT

Background: The mechanisms connecting dietary intake of processed foods with systemic inflammatory markers and cardiovascular risk remain poorly defined. We sought to compare the abundance of pro-inflammatory stimulants of innate immune receptors in processed foods with those produced by the murine ileal and caecal microbiota, and to explore the impact of their ingestion on systemic inflammation and lipid metabolism in vivo. Methods and results: Calibrated receptor-dependent reporter assays revealed that many processed foods, particularly those based on minced meats, contain pro-inflammatory stimulants of Toll-like receptor (TLR)-2 and TLR4 at concentrations which greatly exceed those produced by the endogenous murine ileal microbiota. Chronic dietary supplementation with these stimulants, at concentrations relevant to those measured in the Western diet, promoted hepatic inflammation and reduced several markers of reverse cholesterol transport (RCT) in mice. Hepatocytes were found to be insensitive to TLR2- and TLR4-stimulants directly, but their secretion of functional cholesterol acceptors was impaired by interleukin (IL)-1ß released by TLR-responsive hepatic macrophages. Hepatic macrophage priming by high-fat diet enhanced the impairment of RCT by ingested endotoxin, and this was reversed by macrophage depletion via clodronate liposome treatment, or genetic deficiency in the IL-1 receptor. Conclusion: These findings reveal an unexpected mechanism connecting processed food consumption with cardiovascular risk factors, and introduce the food microbiota as a potential target for therapeutic regulation of lipid metabolism.


Subject(s)
Cholesterol/immunology , Inflammation/immunology , Interleukin-1/immunology , Liver/immunology , Macrophages/immunology , Toll-Like Receptors/immunology , Adult , Animals , Biological Transport , Cells, Cultured , Cholesterol/metabolism , Diet , Gastrointestinal Microbiome/immunology , Gastrointestinal Microbiome/physiology , HEK293 Cells , Hep G2 Cells , Humans , Inflammation/metabolism , Interleukin-1/biosynthesis , Liver/metabolism , Liver/pathology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pathogen-Associated Molecular Pattern Molecules/immunology , Pathogen-Associated Molecular Pattern Molecules/metabolism , RAW 264.7 Cells , Toll-Like Receptors/metabolism
13.
Vaccine ; 37(35): 4975-4986, 2019 08 14.
Article in English | MEDLINE | ID: mdl-31320217

ABSTRACT

Soyasaponins from soybean (Glycine max) represent promising new potent adjuvants for vaccine research because of their immunostimulating properties and weak hemolytic activity. In the present study, saponin microstructures of soyasaponins (soyasaponin Bb, soyasaponin Ab) with lipid components (cholesterol, DPPC (dipalmitoylphosphatidylcholine)) were designed by the lipid film method. In interaction studies between soyasaponins (soyasaponin Ab/Bb) and Langmuir monolayers (model membranes), composed of cholesterol and DPPC, marked interactions between soyasaponins and a pure cholesterol monolayer were observed. No interaction was detected for soyasaponins with a pure DPPC monolayer. The intercalation of soyasaponins in a mixed DPPC/cholesterol (3:1, w/w) monolayer was only observed for the monodesmosidic soyasaponin Bb whereas the second sugar chain of the bidesmosidic soyasaponin Ab impaired the access to the monolayer. Transmission electron microscopy was used for visualizing particle formation of soyasaponins and lipid components. Pseudo-binary systems (soyasaponin Ab/Bb, cholesterol) formed colloidal associations built up from ring-like subunits in the nanometer size range. In pseudo-ternary systems (soyasaponin, cholesterol, DPPC) soyasaponin Bb attacked the liposomal membrane by forming colloidal associations. Colloidal associations in pseudo-ternary systems with soyasaponin Ab, cholesterol and a phospholipid were only observed in the presence of PE (phosphatidylethanolamine) instead of DPPC. In an MTT assay with a HaCaT cell line (keratinocyte cell line) the cell viability was neither affected by the soyasaponins nor by the corresponding formulations. Both the pure soyasaponin solution and the saponin formulations may be promising adjuvant systems for the intradermal vaccine application. Furthermore, interaction studies between the model antigen ovalbumin and colloidal associations of saponins and cholesterol using MST (Microscale Thermophoresis) gave first indications of an antigen binding to colloidal associations. Ex vivo T-cell proliferation in the presence of soyasaponin Ab was confirmed.


Subject(s)
1,2-Dipalmitoylphosphatidylcholine/immunology , Adjuvants, Immunologic/pharmacology , Cholesterol/immunology , Dendritic Cells/drug effects , Saponins/immunology , Vaccines/immunology , Cell Line , Cell Survival/drug effects , Colloids , Dendritic Cells/immunology , Drug Compounding , Humans , Keratinocytes/drug effects , Lymphocyte Activation/drug effects
14.
J Immunol ; 203(4): 853-863, 2019 08 15.
Article in English | MEDLINE | ID: mdl-31270150

ABSTRACT

Cholesterol crystals (CC) are strong activators of complement and could potentially be involved in thromboinflammation through complement-coagulation cross-talk. To explore the coagulation-inducing potential of CC, we performed studies in lepirudin-based human whole blood and plasma models. In addition, immunohistological examinations of brain thrombi and vulnerable plaque material from patients with advanced carotid atherosclerosis were performed using polarization filter reflected light microscopy to identify CC. In whole blood, CC exposure induced a time- and concentration-dependent generation of prothrombin fragment 1+2 (PTF1.2), tissue factor (TF) mRNA synthesis, and monocyte TF expression. Blocking Abs against TF abolished CC-mediated coagulation, thus indicating involvement of the TF-dependent pathway. Blockade of FXII by corn trypsin inhibitor had a significant inhibitory effect on CC-induced PTF1.2 in platelet-free plasma, although the overall activation potential was low. CC exposure did not induce platelet aggregation, TF microparticle induction, or TF on granulocytes or eosinophils. Inhibition of complement C3 by CP40 (compstatin), C5 by eculizumab, or C5aR1 by PMX53 blocked CC-induced PTF1.2 by 90% and reduced TF+ monocytes from 18-20 to 1-2%. The physiologic relevance was supported by birefringent CC structures adjacent to monocytes (CD14), TF, and activated complement iC3b and C5b-9 in a human brain thrombus. Furthermore, monocyte influx and TF induction in close proximity to CC-rich regions with activated complement were found in a vulnerable plaque. In conclusion, CC could be active, releasable contributors to thrombosis by inducing monocyte TF secondary to complement C5aR1 signaling.


Subject(s)
Blood Coagulation/immunology , Cholesterol/immunology , Complement Activation/immunology , Receptor, Anaphylatoxin C5a/metabolism , Thromboplastin/biosynthesis , Carotid Artery Diseases/immunology , Carotid Artery Diseases/metabolism , Humans , Monocytes/immunology , Monocytes/metabolism , Thromboplastin/immunology , Thrombosis/immunology , Thrombosis/metabolism
15.
Cardiovasc Res ; 115(9): 1393-1407, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31095280

ABSTRACT

Inflammation represents the driving feature of many diseases, including atherosclerosis, cancer, autoimmunity and infections. It is now established that metabolic processes shape a proper immune response and within this context the alteration in cellular cholesterol homeostasis has emerged as a culprit of many metabolic abnormalities observed in chronic inflammatory diseases. Cholesterol accumulation supports the inflammatory response of myeloid cells (i.e. augmentation of toll-like receptor signalling, inflammasome activation, and production of monocytes and neutrophils) which is beneficial in the response to infections, but worsens diseases associated with chronic metabolic inflammation including atherosclerosis. In addition to the innate immune system, cells of adaptive immunity, upon activation, have also been shown to undergo a reprogramming of cellular cholesterol metabolism, which results in the amplification of inflammatory responses. Aim of this review is to discuss (i) the molecular mechanisms linking cellular cholesterol metabolism to specific immune functions; (ii) how cellular cholesterol accumulation sustains chronic inflammatory diseases such as atherosclerosis; (iii) the immunometabolic profile of patients with defects of genes affecting cholesterol metabolism including familial hypercholesterolaemia, cholesteryl ester storage disease, Niemann-Pick type C, and immunoglobulin D syndrome/mevalonate kinase deficiency. Available data indicate that cholesterol immunometabolism plays a key role in directing immune cells function and set the stage for investigating the repurposing of existing 'metabolic' drugs to modulate the immune response.


Subject(s)
Cardiovascular Diseases/immunology , Cardiovascular System/immunology , Cholesterol/immunology , Energy Metabolism/immunology , Immune System/immunology , Immunomodulation , Inflammation Mediators/immunology , Inflammation/immunology , Animals , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/physiopathology , Cardiovascular System/metabolism , Cardiovascular System/physiopathology , Cholesterol/metabolism , Humans , Immune System/metabolism , Immune System/physiopathology , Inflammation/metabolism , Inflammation/physiopathology , Inflammation Mediators/metabolism , Signal Transduction
16.
J Steroid Biochem Mol Biol ; 191: 105364, 2019 07.
Article in English | MEDLINE | ID: mdl-31002862

ABSTRACT

Cholesterol is essential for maintaining membrane fluidity in eukaryotes. Additionally, the synthetic cascade of cholesterol results in precursor molecules important for cellular function such as lipid raft formation and protein prenylation. As such, cholesterol homeostasis is tightly regulated. Interestingly, it is now known that some cholesterol precursors and many metabolites serve as active signaling molecules, binding to different classes of receptors including the nuclear receptors. Furthermore, many cholesterol metabolites or their nuclear receptors have been implicated in the regulation of the immune system in normal physiology and disease. Therefore, in this focused review, cholesterol homeostasis and nuclear receptors involved in this regulation will be discussed, with particular emphasis on how these cascades influence the immune system.


Subject(s)
Cholesterol/immunology , Immunity , Receptors, Cytoplasmic and Nuclear/immunology , Animals , Homeostasis , Humans , Immune System/immunology
17.
J Clin Invest ; 129(4): 1785-1800, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30753167

ABSTRACT

Understanding the tumor immune microenvironment (TIME) promises to be key for optimal cancer therapy, especially in triple-negative breast cancer (TNBC). Integrating spatial resolution of immune cells with laser capture microdissection gene expression profiles, we defined distinct TIME stratification in TNBC, with implications for current therapies including immune checkpoint blockade. TNBCs with an immunoreactive microenvironment exhibited tumoral infiltration of granzyme B+CD8+ T cells (GzmB+CD8+ T cells), a type 1 IFN signature, and elevated expression of multiple immune inhibitory molecules including indoleamine 2,3-dioxygenase (IDO) and programmed cell death ligand 1 (PD-L1), and resulted in good outcomes. An "immune-cold" microenvironment with an absence of tumoral CD8+ T cells was defined by elevated expression of the immunosuppressive marker B7-H4, signatures of fibrotic stroma, and poor outcomes. A distinct poor-outcome immunomodulatory microenvironment, hitherto poorly characterized, exhibited stromal restriction of CD8+ T cells, stromal expression of PD-L1, and enrichment for signatures of cholesterol biosynthesis. Metasignatures defining these TIME subtypes allowed us to stratify TNBCs, predict outcomes, and identify potential therapeutic targets for TNBC.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Triple Negative Breast Neoplasms/immunology , Tumor Microenvironment/immunology , B7-H1 Antigen/immunology , CD8-Positive T-Lymphocytes/pathology , Cholesterol/immunology , Female , Granzymes/immunology , Humans , Interferon Type I/immunology , Triple Negative Breast Neoplasms/pathology
18.
J Biochem Mol Toxicol ; 33(5): e22290, 2019 May.
Article in English | MEDLINE | ID: mdl-30664314

ABSTRACT

In this study, we examined the relative immune response of T-lymphocytes and its intracellular cholesterol homeostasis, in a mouse model system, after treatment with immunogen, mitogen, and carcinogen. We studied the T-lymphocyte percentage, their LDL-receptor expression, along with the levels of serum interleukins (IL-2, IFNγ, IL-4, and IL-10) and intracellular cholesterol concentration (cytoplasmic and nuclear). The mitogen was found to be a better stimulator of T-cell marker expressions than the immunogen; though the immunogen was more effective on immunogenic response as was marked from interleukin levels. The chemical carcinogen benzo-α-pyrene at low concentration acted potentially like a mitogen but a reduced immune response was apparent at a carcinogenic dose. The findings in our study focus on the effect of carcinogenic dose of benzo-α-pyrene (BaP) on T-cell immunity. Benzo-α-pyrene causes immunosuppression through restriction of the T-cell population by targeting intracellular cholesterol.


Subject(s)
Benzo(a)pyrene/toxicity , Carcinogens/toxicity , Cholesterol/immunology , Immunity, Cellular/drug effects , Mitogens/toxicity , T-Lymphocytes/immunology , Animals , Cytokines/immunology , Female , Mice , T-Lymphocytes/pathology
19.
J Cell Physiol ; 234(5): 5436-5450, 2019 05.
Article in English | MEDLINE | ID: mdl-30370619

ABSTRACT

Inflammasome mechanisms are involved as some of the pathways of sterile inflammation. Inflammasomes are large multiprotein complexes in the cytosol and are a key system for the production of the pivotal inflammatory cytokines, interleukin (IL)-1ß and IL-18, and inflammatory cell death called pyroptosis. Although a number of inflammasomes have been described, the nucleotide-binding oligomerization domain-, leucine-rich repeat-, and pyrin domain-containing 3 (NLRP3) is the most extensively investigated inflammasome. Exogenous pathogen-associated molecular patterns released during infection and endogenous crystalline danger/damage-associated molecular patterns (DAMPs) are well-known activators of NLRP3 inflammasomes. In addition, nanoparticle-associated molecular patterns (NAMPs), which are mediated by synthetic materials, including nanomaterials and nanoparticles, are proposed to be new danger signals of NLRP3 inflammasomes. Importantly, NAMP- and DAMP-triggered inflammation, a defining characteristic in inflammatory diseases, is termed as sterile inflammation because it occurs in the absence of foreign pathogens. This review focuses on the role of inflammasomes in exogenous NAMP- and endogenous crystalline DAMP-mediated sterile inflammation. Moreover, many regulatory mechanisms have been identified to attenuate NLRP3 inflammasomes. Therefore, we also summarize endogenous negative regulators of NLRP3 inflammasome activation, particularly induced by NAMPs or crystalline DAMPs.


Subject(s)
Alarmins/immunology , Inflammasomes/drug effects , Inflammation/chemically induced , Lipids/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/drug effects , Nanoparticles/adverse effects , Uric Acid/immunology , Alarmins/metabolism , Animals , Calcium Phosphates/immunology , Calcium Phosphates/metabolism , Cholesterol/immunology , Cholesterol/metabolism , Crystallization , Fatty Acids/immunology , Fatty Acids/metabolism , Humans , Inflammasomes/immunology , Inflammasomes/metabolism , Inflammation/immunology , Inflammation/metabolism , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Lipoproteins, LDL/immunology , Lipoproteins, LDL/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Signal Transduction , Uric Acid/metabolism
20.
Iran J Allergy Asthma Immunol ; 18(5): 554-566, 2019 Oct 23.
Article in English | MEDLINE | ID: mdl-32245299

ABSTRACT

Many observations showed that hypercholesterolemia can disrupt immune response. Statin drugs that were used for the treatment of hypercholesterolemia patients can interfere in the regulation of the immune response and cytokine secretion. The primary aim of the current study was to investigate the immune response among treatment-naïve patients with hypercholesterolemia and healthy subjects. The secondary goal of the study was to determine whether atorvastatin can reverse the detrimental effect of hypercholesterolemia on the immune system. Peripheral blood mononuclear cells (PBMCs) were isolated from 50 patients afflicted with hypercholesterolemia who were treatment-naïve along with 50 sex/age-matched hypercholesterolemia patients receiving atorvastatin, and 50 sex/age-matched healthy subjects. Quantitative PCR and ELISA methods were used for gene and protein expression analysis of T helper 1 (Th1) and Th2 related cytokines. Additionally, the expression of the cluster of differentiation (CD) markers on T, B, and natural killer (NK) cells was measured by flow cytometry method. The results showed that hypercholesterolemia and atorvastatin down-regulated the expression of Th1-related cytokines and elevated the levels of Th2-related cytokines. The expression of cell surface markers, CD25 and CD69, was significantly decreased in the treatment-naïve, and atorvastatin groups. It seems that atorvastatin is not able to repair the deleterious effects of hypercholesterolemia on the immune system. Moreover, elevated levels of cholesterol along with the administration of atorvastatin tilt the Th1/Th2 balance in favor of Th2 and reduce T cell activation.


Subject(s)
Atorvastatin/immunology , Hypercholesterolemia/immunology , Immunologic Factors/immunology , Adult , Antigens, CD/immunology , Cholesterol/immunology , Cytokines/immunology , Female , Humans , Killer Cells, Natural/immunology , Leukocytes, Mononuclear/immunology , Lymphocyte Activation/immunology , Male , Middle Aged , Th1 Cells/immunology , Th2 Cells/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...