Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 209
Filter
1.
J Clin Invest ; 131(1)2021 01 04.
Article in English | MEDLINE | ID: mdl-33393500

ABSTRACT

CD1a-autoreactive T cells contribute to skin disease, but the identity of immunodominant self-lipid antigens and their mode of recognition are not yet solved. In most models, MHC and CD1 proteins serve as display platforms for smaller antigens. Here, we showed that CD1a tetramers without added antigen stained large T cell pools in every subject tested, accounting for approximately 1% of skin T cells. The mechanism of tetramer binding to T cells did not require any defined antigen. Binding occurred with approximately 100 lipid ligands carried by CD1a proteins, but could be tuned upward or downward with certain natural self-lipids. TCR recognition mapped to the outer A' roof of CD1a at sites remote from the antigen exit portal, explaining how TCRs can bind CD1a rather than carried lipids. Thus, a major antigenic target of CD1a T cell autoreactivity in vivo is CD1a itself. Based on their high frequency and prevalence among donors, we conclude that CD1a-specific, lipid-independent T cells are a normal component of the human skin T cell repertoire. Bypassing the need to select antigens and effector molecules, CD1a tetramers represent a simple method to track such CD1a-specific T cells from tissues and in any clinical disease.


Subject(s)
Antigens, CD1/immunology , Membrane Lipids/immunology , Receptors, Antigen, T-Cell/immunology , Skin/immunology , T-Lymphocytes/immunology , HEK293 Cells , Humans , K562 Cells
3.
Front Immunol ; 11: 2187, 2020.
Article in English | MEDLINE | ID: mdl-33013920

ABSTRACT

Microvilli are finger-like membrane protrusions, supported by the actin cytoskeleton, and found on almost all cell types. A growing body of evidence suggests that the dynamic lymphocyte microvilli, with their highly curved membranes, play an important role in signal transduction leading to immune responses. Nevertheless, challenges in modulating local membrane curvature and monitoring the high dynamicity of microvilli hampered the investigation of the curvature-generation mechanism and its functional consequences in signaling. These technical barriers have been partially overcome by recent advancements in adapted super-resolution microscopy. Here, we review the up-to-date progress in understanding the mechanisms and functional consequences of microvillus formation in T cell signaling. We discuss how the deformation of local membranes could potentially affect the organization of signaling proteins and their biochemical activities. We propose that curved membranes, together with the underlying cytoskeleton, shape microvilli into a unique compartment that sense and process signals leading to lymphocyte activation.


Subject(s)
Cell Membrane/immunology , Lymphocyte Activation/physiology , Microvilli/immunology , Signal Transduction/immunology , T-Lymphocytes/ultrastructure , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/ultrastructure , Animals , Carrier Proteins/pharmacology , Cell Line , Cell Membrane/drug effects , Cell Membrane/ultrastructure , Cell Shape , Cyclodextrins/pharmacology , Cytokines/physiology , Glycocalyx/drug effects , Glycocalyx/immunology , Humans , Lymphocyte Activation/drug effects , Membrane Lipids/immunology , Membrane Proteins/immunology , Mice , Microfilament Proteins/pharmacology , Microscopy, Electron, Scanning , Microvilli/drug effects , Microvilli/ultrastructure , Receptors, Antigen, T-Cell/immunology , Signal Transduction/drug effects , Stress, Mechanical , Surface Properties , Synaptosomes/drug effects , Synaptosomes/immunology , Synaptosomes/ultrastructure , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
4.
Cell Rep ; 32(7): 108037, 2020 08 18.
Article in English | MEDLINE | ID: mdl-32814041

ABSTRACT

The contribution of membrane interfacial interactions to recognition of membrane-embedded antigens by antibodies is currently unclear. This report demonstrates the optimization of this type of antibodies via chemical modification of regions near the membrane but not directly involved in the recognition of the epitope. Using the HIV-1 antibody 10E8 as a model, linear and polycyclic synthetic aromatic compounds are introduced at selected sites. Molecular dynamics simulations predict the favorable interactions of these synthetic compounds with the viral lipid membrane, where the epitope of the HIV-1 glycoprotein Env is located. Chemical modification of 10E8 with aromatic acetamides facilitates the productive and specific recognition of the native antigen, partially buried in the crowded environment of the viral membrane, resulting in a dramatic increase of its capacity to block viral infection. These observations support the harnessing of interfacial affinity through site-selective chemical modification to optimize the function of antibodies that target membrane-proximal epitopes.


Subject(s)
Antibodies, Neutralizing/immunology , Membrane Lipids/immunology , Humans
5.
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
6.
Eur J Pharm Biopharm ; 145: 1-6, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31560955

ABSTRACT

Both Gram-positive and Gram-negative bacteria can release nano-sized lipid bilayered structures, known as membrane vesicles (MVs). These MVs play an important role in bacterial survival by orchestrating interactions between bacteria and between bacteria and host. The major constituents of MVs are proteins, lipids and nucleic acids. Due to the immunogenicity of the membrane lipids and/or proteins of the MVs, in combination with adjuvant danger signals and the repeating patterns on the nanosized surface, MVs can effectively stimulate the innate and adaptive immune system. Since they are non-replicating, they are safer than attenuated vaccines. In addition, by genetic engineering of the donor cells, further improvements to their safety profile, immunogenicity and yield can be achieved. To date, one MV-based vaccine against Neisseria meningitidis (N. meningitidis) serogroup B was approved. Other (engineered) MVs in the pipeline study are mostly in the preclinical phase.


Subject(s)
Bacteria/immunology , Lipid Bilayers/immunology , Membrane Lipids/immunology , Membranes/immunology , Vaccines/immunology , Adaptive Immunity/immunology , Adjuvants, Immunologic , Animals , Antibody Formation/immunology , Bacterial Proteins/immunology , Humans
7.
Front Immunol ; 10: 1174, 2019.
Article in English | MEDLINE | ID: mdl-31191542

ABSTRACT

Mast cells (MCs) have long been mainly regarded as effector cells in IgE-associated allergic disorders with potential immunoregulatory roles. Located close to the allergen entry sites in the skin and mucosa, MCs can capture foreign substances such as allergens, toxins, or noxious substances and are exposed to the danger signals produced by epithelial cells. MC reactivity shaped by tissue-specific factors is crucial for allergic responses ranging from local skin reactions to anaphylactic shock. Development of Th2 response leading to allergen-specific IgE production is a prerequisite for MC sensitization and induction of FcεRI-mediated MC degranulation. Up to now, IgE production has been mainly associated with proteins, whereas lipids present in plant pollen grains, mite fecal particles, insect venoms, or food have been largely overlooked regarding their immunostimulatory and immunomodulatory properties. Recent studies, however, have now demonstrated that lipids affect the sensitization process by modulating innate immune responses of epithelial cells, dendritic cells, and NK-T cells and thus crucially contribute to the outcome of sensitization. Whether and how lipids affect also MC effector functions in allergic reactions has not yet been fully clarified. Here, we discuss how lipids can affect MC responses in the context of allergic inflammation. Direct effects of immunomodulatory lipids on MC degranulation, changes in local lipid composition induced by allergens themselves and changes in lipid transport affecting MC reactivity are possible mechanisms by which the function of MC might be modulated.


Subject(s)
Immunomodulation , Lipids/immunology , Mast Cells/immunology , Mast Cells/metabolism , Allergens/chemistry , Allergens/immunology , Animals , Cell Degranulation/immunology , Disease Susceptibility , Humans , Hypersensitivity/etiology , Hypersensitivity/metabolism , Lipid Metabolism , Lipids/chemistry , Membrane Lipids/immunology , Membrane Lipids/metabolism , Structure-Activity Relationship
8.
Molecules ; 23(11)2018 Nov 15.
Article in English | MEDLINE | ID: mdl-30445712

ABSTRACT

Zinc Oxide (ZnO) nanoparticles are suspected to produce toxic effects toward mammalian cells; however, discrepancies in the extent of this effect have been reported between different cell lines. Simultaneously, high levels of ultraviolet (UV-C) radiation can have carcinogenic effects. The mechanism of this effect is also not well understood. Due to similarities in phenotype morphology after cell exposure to ZnO nanoparticles and UV-C irradiation, we emit the hypothesis that the toxicity of both these factors is related to damage of cellular membranes and affect their sterol content. Wild-type Chinese Hamster Ovary (CHO-K1) cells were exposed to ZnO nanoparticles or UV-C radiation. The amount of absorbed ZnO was determined by UV-visible spectroscopy and the changes in sterol profiles were evaluated by gas chromatography. Cell viability after both treatments was determined by microscopy. Comparing morphology results suggested similarities in toxicology events induced by ZnO nanoparticles and UV exposure. UV-C exposure for 360 min disrupts the sterol metabolic pathway by increasing the concentration of cholesterol by 21.6-fold. This increase in cholesterol production supports the hypothesis that UV irradiation has direct consequences in initiating sterol modifications in the cell membrane.


Subject(s)
Cholesterol/chemistry , Cholesterol/pharmacology , Membrane Lipids/chemistry , Membrane Lipids/immunology , Metal Nanoparticles/chemistry , Ultraviolet Rays , Zinc Oxide/chemistry , Animals , CHO Cells , Cell Survival/drug effects , Chromatography, Gas , Cricetulus , Spectrum Analysis , Sterols
9.
EMBO J ; 37(5)2018 03 01.
Article in English | MEDLINE | ID: mdl-29378774

ABSTRACT

Intestinal homeostasis relies on a continuous dialogue between the commensal bacteria and the immune system. Natural killer T (NKT) cells, which recognize CD1d-restricted microbial lipids and self-lipids, contribute to the regulation of mucosal immunity, yet the mechanisms underlying their functions remain poorly understood. Here, we demonstrate that NKT cells respond to intestinal lipids and CD11c+ cells (including dendritic cells (DCs) and macrophages) are essential to mediate lipid presentation within the gut ultimately controlling intestinal NKT cell homeostasis and activation. Conversely, CD1d and NKT cells participate in the control of the intestinal bacteria composition and compartmentalization, in the regulation of the IgA repertoire and in the induction of regulatory T cells within the gut. These changes in intestinal homeostasis require CD1d expression on DC/macrophage populations as mice with conditional deletion of CD1d on CD11c+ cells exhibit dysbiosis and altered immune homeostasis. These results unveil the importance of CD11c+ cells in controlling lipid-dependent immunity in the intestinal compartment and reveal an NKT cell-DC crosstalk as a key mechanism for the regulation of gut homeostasis.


Subject(s)
Intestinal Mucosa/immunology , Membrane Lipids/immunology , Natural Killer T-Cells/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Antigens, CD1d/biosynthesis , Antigens, CD1d/genetics , Antigens, CD1d/immunology , CD11c Antigen/metabolism , Dendritic Cells/immunology , Dysbiosis/genetics , Gastrointestinal Microbiome/immunology , Immunoglobulin A/immunology , Interleukin-4/immunology , Intestinal Mucosa/microbiology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout
10.
Biochimie ; 141: 70-79, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28483690

ABSTRACT

The hepatitis E virus (HEV) is the most common cause of acute hepatitis worldwide. Although HEV is a small, naked RNA virus, HEV particles become associated with lipids in the blood of infected patients and in the supernatant of culture systems. The egress of these particles from cells implies the exocytosis pathway but the question of the role of the resulting HEV RNA containing exosomes and the nature of the lipids they contain has not been fully addressed. We determined the lipid proportions of exosomes from uninfected and HEV-infected cells and their role in HEV spreading. We cultured a suitable HEV strain on HepG2/C3A cells and analyzed the population of exosomes containing HEV RNA using lipidomics methods and electron microscopy. We also quantified HEV infectivity using an infectivity endpoint method based on HEV RNA quantification to calculate the tissue culture infectious dose 50. Exosomes produced by HEV-infected HepG2/C3A cells contained encapsidated HEV RNA. These HEV RNA-containing exosomes were infectious but ten times less than stools. HEV from stools, but not exosome-associated HEV from culture supernatant, was neutralized by anti-HEV antibodies in a dose-dependent manner. HEV infection did not influence the morphology or lipid proportions of the bulk of exosomes. These exosomes contained significantly more cholesterol, phosphatidylserine, sphingomyelin and ceramides than the parent cells, but less phosphoinositides and polyunsaturated fatty acids. Exosomes play a major role in HEV egress but HEV infection does not modify the characteristics of the bulk of exosomes produced by infected cells. PS and cholesterol enriched in these vesicles could then be critical for HEV entry. HEV particles in exosomes are protected from the immune response which could lead to the wide circulation of HEV in its host.


Subject(s)
Cell-Derived Microparticles/immunology , Exosomes/immunology , Hepatitis E virus/immunology , Hepatitis E/immunology , Membrane Lipids/immunology , Hep G2 Cells , Hepatitis E/pathology , Humans
11.
J Biol Chem ; 292(13): 5571-5583, 2017 Mar 31.
Article in English | MEDLINE | ID: mdl-28213514

ABSTRACT

The 4E10 antibody displays an extreme breadth of HIV-1 neutralization and therefore constitutes a suitable model system for structure-guided vaccine design and immunotherapeutics against AIDS. In this regard, the relevance of autoreactivity with membrane lipids for the biological function of this antibody is still a subject of controversy. To address this dispute, herein we have compared the membrane partitioning ability of the 4E10 antibody and several of its variants, which were mutated at the region of the paratope surface in contact with the membrane interface. We first employed a physical separation approach (vesicle flotation) and subsequently carried out quantitative fluorescence measurements in an intact system (spectroscopic titration), using 4E10 Fab labeled with a polarity-sensitive fluorescent probe. Moreover, recognition of epitope peptide in membrane was demonstrated by photo-cross-linking assays using a Fab that incorporated the genetically encoded unnatural amino acid p-benzoylphenylalanine. The experimental data ruled out that the proposed stereospecific recognition of viral lipids was necessary for the function of the antibody. In contrast, our data suggest that nonspecific electrostatic interactions between basic residues of 4E10 and acidic phospholipids in the membranes contribute to the observed biological function. Moreover, the energetics of membrane partitioning indicated that 4E10 behaves as a peripheral membrane protein, tightening the binding to the ligand epitope inserted in the viral membrane. The implications of these findings for the natural production and biological function of this antibody are discussed.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Cell Membrane/immunology , HIV Antibodies/immunology , AIDS Vaccines , Antibodies, Monoclonal/adverse effects , Antibodies, Neutralizing/adverse effects , Autoimmunity , Broadly Neutralizing Antibodies , Cell Line , Drug Design , Epitopes , HIV Antibodies/adverse effects , Humans , Membrane Lipids/immunology , Viral Matrix Proteins/immunology
12.
Immunity ; 44(1): 21-31, 2016 Jan 19.
Article in English | MEDLINE | ID: mdl-26777395

ABSTRACT

Numerous studies of the anti-HIV-1 envelope glycoprotein 41 (gp41) broadly neutralizing antibody 4E10 suggest that 4E10 also interacts with membrane lipids, but the antibody regions contacting lipids and its orientation with respect to the viral membrane are unknown. Vaccine immunogens capable of re-eliciting these membrane proximal external region (MPER)-like antibodies may require a lipid component to be successful. We performed a systematic crystallographic study of lipid binding to 4E10 to identify lipids bound by the antibody and the lipid-interacting regions. We identified phosphatidic acid, phosphatidylglycerol, and glycerol phosphate as specific ligands for 4E10 in the crystal structures. 4E10 used its CDRH1 loop to bind the lipid head groups, while its CDRH3 interacted with the hydrophobic lipid tails. Identification of the lipid binding sites on 4E10 may aid design of immunogens for vaccines that include a lipid component in addition to the MPER on gp41 for generation of broadly neutralizing antibodies.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Binding Sites, Antibody/immunology , Epitopes, B-Lymphocyte/immunology , HIV Antibodies/immunology , Membrane Lipids/immunology , Broadly Neutralizing Antibodies , Cell Line , Crystallography, X-Ray , Epitopes, B-Lymphocyte/chemistry , Humans , Protein Binding/immunology , Protein Conformation
13.
J Immunol ; 195(10): 4595-603, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26466957

ABSTRACT

Human T cells are activated by both peptide and nonpeptide Ags produced by Mycobacterium tuberculosis. T cells recognize cell wall lipids bound to CD1 molecules, but effector functions of CD1-reactive T cells have not been systematically assessed in M. tuberculosis-infected humans. It is also not known how these features correlate with T cell responses to secreted protein Ags. We developed a flow cytometric assay to profile CD1-restricted T cells ex vivo and assessed T cell responses to five cell wall lipid Ags in a cross-sectional study of 19 M. tuberculosis-infected and 22 M. tuberculosis-uninfected South African adolescents. We analyzed six T cell functions using a recently developed computational approach for flow cytometry data in high dimensions. We compared these data with T cell responses to five protein Ags in the same cohort. We show that CD1b-restricted T cells producing antimycobacterial cytokines IFN-γ and TNF-α are detectable ex vivo in CD4(+), CD8(+), and CD4(-)CD8(-) T cell subsets. Glucose monomycolate was immunodominant among lipid Ags tested, and polyfunctional CD4 T cells specific for this lipid simultaneously expressed CD40L, IFN-γ, IL-2, and TNF-α. Lipid-reactive CD4(+) T cells were detectable at frequencies of 0.001-0.01%, and this did not differ by M. tuberculosis infection status. Finally, CD4 T cell responses to lipids were poorly correlated with CD4 T cell responses to proteins (Spearman rank correlation -0.01; p = 0.95). These results highlight the functional diversity of CD1-restricted T cells circulating in peripheral blood as well as the complementary nature of T cell responses to mycobacterial lipids and proteins. Our approach enables further population-based studies of lipid-specific T cell responses during natural infection and vaccination.


Subject(s)
Antigens, CD1/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Membrane Lipids/immunology , Mycobacterium tuberculosis/immunology , Tuberculosis, Pulmonary/immunology , Adolescent , Antigens, Bacterial/immunology , CD40 Ligand/biosynthesis , Cell Wall/immunology , Cross-Sectional Studies , Female , Flow Cytometry , Glycolipids/immunology , Humans , Interferon-gamma/biosynthesis , Interleukin-2/biosynthesis , K562 Cells , Lymphocyte Activation/immunology , Male , South Africa/epidemiology , Tuberculosis, Pulmonary/epidemiology , Tuberculosis, Pulmonary/microbiology , Tumor Necrosis Factor-alpha/biosynthesis
14.
J Immunol ; 195(6): 2540-51, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26254340

ABSTRACT

Semi-invariant/type I NKT cells are a well-characterized CD1d-restricted T cell subset. The availability of potent Ags and tetramers for semi-invariant/type I NKT cells allowed this population to be extensively studied and revealed their central roles in infection, autoimmunity, and tumor immunity. In contrast, diverse/type II NKT (dNKT) cells are poorly understood because the lipid Ags that they recognize are largely unknown. We sought to identify dNKT cell lipid Ag(s) by interrogating a panel of dNKT mouse cell hybridomas with lipid extracts from the pathogen Listeria monocytogenes. We identified Listeria phosphatidylglycerol as a microbial Ag that was significantly more potent than a previously characterized dNKT cell Ag, mammalian phosphatidylglycerol. Further, although mammalian phosphatidylglycerol-loaded CD1d tetramers did not stain dNKT cells, the Listeria-derived phosphatidylglycerol-loaded tetramers did. The structure of Listeria phosphatidylglycerol was distinct from mammalian phosphatidylglycerol because it contained shorter, fully-saturated anteiso fatty acid lipid tails. CD1d-binding lipid-displacement studies revealed that the microbial phosphatidylglycerol Ag binds significantly better to CD1d than do counterparts with the same headgroup. These data reveal a highly potent microbial lipid Ag for a subset of dNKT cells and provide an explanation for its increased Ag potency compared with the mammalian counterpart.


Subject(s)
Antigens/immunology , Listeria monocytogenes/immunology , Membrane Lipids/immunology , Natural Killer T-Cells/immunology , Phosphatidylglycerols/immunology , Animals , Antigens, CD1d/immunology , Cell Line , Hybridomas/immunology , Mice , T-Lymphocyte Subsets/immunology
15.
PLoS One ; 10(7): e0132949, 2015.
Article in English | MEDLINE | ID: mdl-26172831

ABSTRACT

In this study, we investigated the impact of the cell membrane composition of E. faecalis on its recognition by the host immune system. To this end, we employed an E. faecalis deletion mutant (ΔbgsA) that does not synthesize the major cell membrane glycolipid diglycosyl-diacylglycerol (DGlcDAG). Proteomic analysis revealed that 13 of a total of 21 upregulated surface-associated proteins of E. faecalis ΔbgsA were lipoproteins. This led to a total lipoprotein content in the cell membrane of 35.8% in ΔbgsA compared to only 9.4% in wild-type bacteria. Increased lipoprotein content strongly affected the recognition of ΔbgsA by mouse macrophages in vitro with an increased stimulation of TNF-α production by heat-fixed bacteria and secreted antigens. Inactivation of the prolipoprotein diacylglycerol transferase (lgt) in ΔbgsA abrogated TNF-α induction by a ΔbgsA_lgt double mutant indicating that lipoproteins mediate increased activation of mouse macrophages by ΔbgsA. Heat-fixed ΔbgsA bacteria, culture supernatant, or cell membrane lipid extract activated transfected HEK cells in a TLR2-dependent fashion; the same was not true of wild-type bacteria. In mice infected intraperitoneally with a sublethal dose of E. faecalis we observed a 70% greater mortality in mice infected with ΔbgsA compared with wild-type-infected mice. Increased mortality due to ΔbgsA infection was associated with elevated plasma levels of the inflammatory cytokines TNF-α, IL-6 and MIP-2. In summary, our results provide evidence that an E. faecalis mutant lacking its major bilayer forming glycolipid DGlcDAG upregulates lipoprotein expression leading to increased activation of the host innate immune system and virulence in vivo.


Subject(s)
Cell Membrane/immunology , Enterococcus faecalis/immunology , Glycolipids/immunology , Host-Pathogen Interactions/immunology , Lipoproteins/immunology , Animals , Bacterial Proteins/immunology , Cell Line , Chemokine CXCL2/blood , Female , HEK293 Cells , Humans , Immunity, Innate/immunology , Interleukin-6/blood , Macrophages , Membrane Lipids/immunology , Membrane Proteins/immunology , Mice , Mice, Inbred BALB C , Toll-Like Receptor 2/immunology , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/immunology , Virulence/immunology
16.
EMBO Mol Med ; 7(4): 488-505, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25750210

ABSTRACT

Group B streptococci (GBS) are Gram-positive bacteria that cause infections in utero and in newborns. We recently showed that the GBS pigment is hemolytic and increased pigment production promotes bacterial penetration of human placenta. However, mechanisms utilized by the hemolytic pigment to induce host cell lysis and the consequence on fetal injury are not known. Here, we show that the GBS pigment induces membrane permeability in artificial lipid bilayers and host cells. Membrane defects induced by the GBS pigment trigger K(+) efflux leading to osmotic lysis of red blood cells or pyroptosis in human macrophages. Macrophages lacking the NLRP3 inflammasome recovered from pigment-induced cell damage. In a murine model of in utero infection, hyperpigmented GBS strains induced fetal injury in both an NLRP3 inflammasome-dependent and NLRP3 inflammasome-independent manner. These results demonstrate that the dual mechanism of action of the bacterial pigment/lipid toxin leading to hemolysis or pyroptosis exacerbates fetal injury and suggest that preventing both activities of the hemolytic lipid is likely critical to reduce GBS fetal injury and preterm birth.


Subject(s)
Bacterial Toxins , Cell Membrane Permeability , Fetal Diseases , Membrane Lipids , Pyroptosis/immunology , Streptococcal Infections , Streptococcus agalactiae , Animals , Bacterial Toxins/immunology , Bacterial Toxins/toxicity , Cell Line, Tumor , Female , Fetal Diseases/immunology , Fetal Diseases/microbiology , Fetal Diseases/pathology , Humans , Male , Membrane Lipids/immunology , Membrane Lipids/toxicity , Mice , Mice, Knockout , Streptococcal Infections/immunology , Streptococcal Infections/pathology , Streptococcus agalactiae/immunology , Streptococcus agalactiae/pathogenicity
17.
Adv Healthc Mater ; 4(7): 1054-65, 2015 May.
Article in English | MEDLINE | ID: mdl-25612325

ABSTRACT

Two complementary coiled-coil peptides CCE/CCK are used to develop a "drug free" therapeutic system, which can specifically kill cancer cells without a drug. CCE is attached to the Fab' fragment of anti-CD20 1F5 antibody (Fab'-CCE), and CCK is conjugated in multiple grafts to poly[N-(2-hydroxypropyl)methacrylamide] (P-(CCK)x ). Two conjugates are consecutively administered: First, Fab'-CCE coats peptide CCE at CD20 antigen of lymphoma cell surface; second, CCE/CCK biorecognition between Fab'-CCE and P-(CCK)x leads to coiled-coil formation, CD20 crosslinking, membrane reorganization, and ultimately cell apoptosis. To prove that two conjugates can assemble at cell surface, multiple fluorescence imaging studies are performed, including 2-channel FMT, 3D confocal microscopy, and 4-color FACS. Confocal microscopy shows colocalization of two fluorescently labeled conjugates on non-Hodgkin's lymphoma (NHL) Raji cell surface, indicating "two-step" targeting specificity. The fluorescent images also reveal that these two conjugates can disrupt normal membrane lipid distribution and form lipid raft clusters, leading to cancer cell apoptosis. This "two-step" biorecognition capacity is further demonstrated in a NHL xenograft model, using fluorescent images at whole-body, tissue and cell levels. It is also found that delaying injection of P-(CCK)x can significantly enhance targeting efficacy. This high-specificity therapeutics provide a safe option to treat NHL and other B cell malignancies.


Subject(s)
Immunoglobulin Fab Fragments/immunology , Lymphoma/drug therapy , Lymphoma/immunology , Peptides/immunology , Peptides/therapeutic use , Acrylamides/immunology , Animals , Antigens, CD20/immunology , Apoptosis/drug effects , Apoptosis/immunology , Cell Line, Tumor , Female , Fluorescence , Humans , Lipids/immunology , Membrane Lipids/immunology , Mice, Nude , Mice, SCID , Multimodal Imaging/methods
18.
Eur J Immunol ; 45(4): 1258-69, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25545687

ABSTRACT

Membrane microdomains play an important role in the regulation of natural killer (NK) cell activities. These cholesterol-rich membrane domains are enriched at the activating immunological synapse and several activating NK-cell receptors are known to localize to membrane microdomains upon receptor engagement. In contrast, inhibitory receptors do not localize in these specialized membrane domains. In addition, the functional competence of educated NK cells correlates with a confinement of activating receptors in membrane microdomains. However, the molecular basis for this confinement is unknown. Here, we investigate the structural requirements for the recruitment of the human-activating NK-cell receptors NKG2D and 2B4 to detergent-resistant membrane fractions in the murine BA/F3 cell line and in the human NK-cell line NKL. This stimulation-dependent recruitment occurred independently of the intracellular domains of the receptors. However, either interfering with the association between NKG2D and DAP10, or mutating the transmembrane region of 2B4 impacted the recruitment of the receptors to detergent-resistant membrane fractions and modulated the function of 2B4 in NK cells. Our data suggest a potential interaction between the transmembrane region of NK-cell receptors and membrane lipids as a molecular mechanism involved in determining the membrane confinement of activating NK-cell receptors.


Subject(s)
Antigens, CD/metabolism , Killer Cells, Natural/immunology , Membrane Microdomains/metabolism , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Receptors, Immunologic/immunology , Receptors, Immunologic/metabolism , Animals , Antigens, CD/genetics , Cell Line , Humans , Lymphocyte Activation/immunology , Membrane Lipids/immunology , Mice , Multiprotein Complexes/immunology , Protein Structure, Tertiary , Receptors, Immunologic/genetics , Signaling Lymphocytic Activation Molecule Family
19.
J Biol Chem ; 289(48): 33098-108, 2014 Nov 28.
Article in English | MEDLINE | ID: mdl-25305015

ABSTRACT

The transporter associated with antigen processing (TAP) constitutes a focal element in the adaptive immune response against infected or malignantly transformed cells. TAP shuttles proteasomal degradation products into the lumen of the endoplasmic reticulum for loading of major histocompatibility complex (MHC) class I molecules. Here, the heterodimeric TAP complex was purified and reconstituted in nanodiscs in defined stoichiometry. We demonstrate that a single heterodimeric core-TAP complex is active in peptide binding, which is tightly coupled to ATP hydrolysis. Notably, with increasing peptide length, the ATP turnover was gradually decreased, revealing that ATP hydrolysis is coupled to the movement of peptide through the ATP-binding cassette transporter. In addition, all-atom molecular dynamics simulations show that the observed 22 lipids are sufficient to form an annular belt surrounding the TAP complex. This lipid belt is essential for high affinity inhibition by the herpesvirus immune evasin ICP47. In conclusion, nanodiscs are a powerful approach to study the important role of lipids as well as the function, interaction, and modulation of the antigen translocation machinery.


Subject(s)
Antigen Presentation , Immediate-Early Proteins/metabolism , Membrane Lipids/metabolism , Multiprotein Complexes/metabolism , Peptides/metabolism , Proteasome Endopeptidase Complex/metabolism , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/immunology , Adenosine Triphosphatases/metabolism , Adenosine Triphosphate/genetics , Adenosine Triphosphate/immunology , Adenosine Triphosphate/metabolism , Cell Line, Tumor , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Humans , Immediate-Early Proteins/genetics , Immediate-Early Proteins/immunology , Membrane Lipids/genetics , Membrane Lipids/immunology , Multiprotein Complexes/genetics , Multiprotein Complexes/immunology , Peptides/genetics , Peptides/immunology , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/immunology , Protein Transport/genetics , Protein Transport/immunology
20.
Article in English | MEDLINE | ID: mdl-24667417

ABSTRACT

Psychological stress is a condition that not only generates behavioral disorders but also disrupts homeostasis and immune activity that can exacerbate or lead to inflammatory diseases. The aim of this work was to study biochemical changes in circulating immune cells from rats under psychological stress by using vibrational spectroscopy. A stress model was used, where exposure to a stressor was repeated for 5 days. Subsequently, circulating lymphocytes were examined for their biomolecular vibrational fingerprints with synchrotron radiation based-Fourier transform infrared microspectroscopy. The results showed an increased absorption at the ester lipid region (1720-1755 cm(-1)) in lymphocytes from stressed rats, suggesting lipid peroxidation. Statistical significant changes in wavenumber peak position and absorbance in the nucleic acid region were also observed (915-950 cm(-1) Z-DNA, 1090-1150 cm(-1) symmetric stretching of P-O-C, 1200-1260 cm(-1) asymmetric PO2 and 1570-1510 cm(-1) methylated nucleotides) which suggest a reduction of transcriptional activity in lymphocytes from stressed rat. These results unravel part of the mechanisms by which psychological stress may affect the immune system leading to systemic consequences.


Subject(s)
Lymphocytes/immunology , Membrane Lipids/immunology , Stress, Psychological/immunology , Transcription, Genetic/immunology , Animals , Lymphocytes/pathology , Rats , Rats, Wistar , Spectroscopy, Fourier Transform Infrared , Stress, Psychological/pathology , Synchrotrons
SELECTION OF CITATIONS
SEARCH DETAIL
...