Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Front Immunol ; 14: 1111629, 2023.
Article in English | MEDLINE | ID: mdl-36761759

ABSTRACT

Background: The emergence of novel SARS-CoV-2 variants that resist neutralizing antibodies drew the attention to cellular immunity and calls for the development of alternative vaccination strategies to combat the pandemic. Here, we have assessed the kinetics of T cell responses and protective efficacy against severe COVID-19 in pre- and post-exposure settings, elicited by PolyPEPI-SCoV-2, a peptide based T cell vaccine. Methods: 75 Syrian hamsters were immunized subcutaneously with PolyPEPI-SCoV-2 on D0 and D14. On D42, hamsters were intranasally challenged with 102 TCID50 of the virus. To analyze immunogenicity by IFN-γ ELISPOT and antibody secretion, lymphoid tissues were collected both before (D0, D14, D28, D42) and after challenge (D44, D46, D49). To measure vaccine efficacy, lung tissue, throat swabs and nasal turbinate samples were assessed for viral load and histopathological changes. Further, body weight was monitored on D0, D28, D42 and every day after challenge. Results: The vaccine induced robust activation of T cells against all SARS-CoV-2 structural proteins that were rapidly boosted after virus challenge compared to control animals (~4-fold, p<0.05). A single dose of PolyPEPI-SCoV-2 administered one day after challenge also resulted in elevated T cell response (p<0.01). The vaccination did not induce virus-specific antibodies and viral load reduction. Still, peptide vaccination significantly reduced body weight loss (p<0.001), relative lung weight (p<0.05) and lung lesions (p<0.05), in both settings. Conclusion: Our study provides first proof of concept data on the contribution of T cell immunity on disease course and provide rationale for the use of T cell-based peptide vaccines against both novel SARS-CoV-2 variants and supports post-exposure prophylaxis as alternative vaccination strategy against COVID-19.


Subject(s)
COVID-19 , Cancer Vaccines , Animals , Cricetinae , T-Lymphocytes , SARS-CoV-2 , COVID-19/prevention & control , Vaccines, Subunit , Mesocricetus , Post-Exposure Prophylaxis , Patient Acuity , Antibodies, Neutralizing
2.
Scand J Immunol ; 97(2): e13241, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36519562

ABSTRACT

Der p 1 is one of the major allergenic molecules of Dermatophagoides pteronyssinus, causing house dust mite (HDM) allergy. The pathological B cells produce allergen-specific IgE antibodies that mediate the hypersensitivity reaction, therefore the selective elimination of these B cells is a legitimate therapeutic goal in allergy. Chimeric molecule Dp51-72 able to cross-link B cell inhibitory complement receptor type 1 and BCR on Der p 1-specific B cells was constructed. The signalling capabilities of this molecule have been tested on human B cells. A humanized mouse model of HDM allergy has been used to test the in vivo effects of the chimeric molecule administration. Administering the chimeric molecule to immunodeficient Rag2- γc- mice transferred with PBMCs from allergic patients resulted in reduction of allergen-specific IgE antibodies in the sera, and reduced infiltration of immune cells in lung histology preparations. Reduced numbers of human CD45+ and CD4+ cells in the lungs as well as inhibition of mast cell degranulation were also observed. The treatment with Dp51-72 chimera significantly decreased the local levels of anti-Dpt IgE antibodies in the bronchoalveolar lavage fluid (BALF). The binding of the chimeric molecule to tonsillar B cells triggers the tyrosine phosphorylation of 30-32 kDa protein, which is most likely involved in the inhibitory process. Administration of constructed chimeric molecules to humanized mice with developed inflammation resulted in specific suppression of disease-associated IgE antibody-producing cells and preserved lung histology. This effective approach could be further developed into a therapeutic agent for treatment of patients with HDM allergy.


Subject(s)
Dust Mite Allergy , Hypersensitivity , Receptors, Antigen, B-Cell , Animals , Humans , Mice , Allergens , Antigens, Dermatophagoides , Disease Models, Animal , DNA-Binding Proteins , Dust Mite Allergy/metabolism , Immunoglobulin E , Nuclear Proteins , B-Lymphocytes/drug effects , Receptors, Antigen, B-Cell/drug effects
3.
Cell Mol Life Sci ; 79(6): 291, 2022 May 11.
Article in English | MEDLINE | ID: mdl-35546365

ABSTRACT

Complement C3 was originally regarded as a serum effector protein, although recent data has emerged suggesting that intracellular C3 can also regulate basic cellular processes. Despite the growing interest in intracellular C3 functions, the mechanism behind its generation has not been demonstrated. In this study we show that C3 can be expressed from an alternative translational start site, resulting in C3 lacking the signal peptide, which is therefore translated in the cytosol. In contrast to the secreted form, alternatively translated cytosolic C3 is not glycosylated, is present mainly in a reduced state, and is turned over by the ubiquitin-proteasome system. C3 can also be retrotranslocated from the endoplasmic reticulum into the cytosol, structurally resembling secreted C3. Finally, we demonstrate that intracellular cytosolic C3 can opsonize invasive Staphylococcus aureus within epithelial cell, slowing vacuolar escape as well as impacting bacterial survival on subsequent exposure to phagocytes. Our work therefore reveals the existence and origin of intracellular, cytosolic C3, and demonstrates functions for cytosolic C3 in intracellular detection of cytoinvasive pathogens.


Subject(s)
Complement C3 , Proteasome Endopeptidase Complex , Bacteria/metabolism , Complement C3/metabolism , Cytosol/metabolism , Endoplasmic Reticulum/metabolism , Proteasome Endopeptidase Complex/metabolism
4.
Front Immunol ; 12: 660852, 2021.
Article in English | MEDLINE | ID: mdl-34671340

ABSTRACT

Besides being a key effector arm of innate immunity, a plethora of non-canonical functions of complement has recently been emerging. Factor H (FH), the main regulator of the alternative pathway of complement activation, has been reported to bind to various immune cells and regulate their functions, beyond its role in modulating complement activation. In this study we investigated the effect of FH, its alternative splice product FH-like protein 1 (FHL-1), the FH-related (FHR) proteins FHR-1 and FHR-5, and the recently developed artificial complement inhibitor mini-FH, on two key innate immune cells, monocytes and neutrophilic granulocytes. We found that, similar to FH, the other factor H family proteins FHL-1, FHR-1 and FHR-5, as well as the recombinant mini-FH, are able to bind to both monocytes and neutrophils. As a functional outcome, immobilized FH and FHR-1 inhibited PMA-induced NET formation, but increased the adherence and IL-8 production of neutrophils. FHL-1 increased only the adherence of the cells, while FHR-5 was ineffective in altering these functions. The adherence of monocytes was increased on FH, recombinant mini-FH and FHL-1 covered surfaces and, except for FHL-1, the same molecules also enhanced secretion of the inflammatory cytokines IL-1ß and TNFα. When monocytes were stimulated with LPS in the presence of immobilized FH family proteins, FH, FHL-1 and mini-FH enhanced whereas FHR-1 and FHR-5 decreased the secretion of TNFα; FHL-1 and mini-FH also enhanced IL-10 release compared to the effect of LPS alone. Our results reveal heterogeneous effects of FH and FH family members on monocytes and neutrophils, altering key features involved in pathogen killing, and also demonstrate that FH-based complement inhibitors, such as mini-FH, may have effects beyond their function of inhibiting complement activation. Thus, our data provide new insight into the non-canonical functions of FH, FHL-1, FHR-1 and FHR-5 that might be exploited during protection against infections and in vaccine development.


Subject(s)
Blood Proteins/metabolism , Complement System Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , LIM Domain Proteins/metabolism , Monocytes/metabolism , Muscle Proteins/metabolism , Neutrophils/metabolism , Animals , Blood Proteins/genetics , Cell Shape , Complement Factor H/genetics , Complement Factor H/metabolism , Complement Inactivating Agents/pharmacology , Complement System Proteins/genetics , Cytokines/metabolism , Extracellular Traps/metabolism , Humans , Inflammation Mediators/metabolism , Intracellular Signaling Peptides and Proteins/genetics , LIM Domain Proteins/genetics , Monocytes/drug effects , Monocytes/immunology , Muscle Proteins/genetics , Neutrophils/drug effects , Neutrophils/immunology , Phenotype , Sf9 Cells , Signal Transduction , Tetradecanoylphorbol Acetate/pharmacology , U937 Cells
5.
Front Immunol ; 10: 493, 2019.
Article in English | MEDLINE | ID: mdl-30941132

ABSTRACT

Beside its classical role as a serum effector system of innate immunity, evidence is accumulating that complement has an intracellular repertoire of components that provides not only immune defense, but also functions to maintain cellular homeostasis. While complement proteins, mainly the central component C3, have been detected in B cells, their exact function and source remain largely unexplored. In this study, we investigated the expression and origin of intracellular C3 in human B cells together with its role in B cell homeostasis. Our data provide evidence that endogenous expression of C3 is very low in human B cells and, in accordance with the recent publication, the main origin of intracellular C3 is the serum. Interestingly, we found that both serum-derived and purified C3 are able to enter the nucleus of viable B cells, suggesting its potential involvement in regulation of gene transcription. ELISA, gel shift assay, confocal microscopy, and chromatin immunoprecipitation proved that C3 and C3a strongly bind to nuclear DNA, and among the interacting genes there are key factors of lymphocyte development and differentiation. The strong interaction of C3 with histone proteins and its potential ability to induce chromatin rearrangement suggest that C3/C3a might regulate DNA transcription via chromatin remodeling. Our data reveal a novel, hitherto undescribed role of C3 in immune cell homeostasis, which further extends the repertoire how complement links innate and adaptive immunity and regulates basic processes of the cells.


Subject(s)
B-Lymphocytes/immunology , Complement C3/immunology , DNA/genetics , Transcription, Genetic/immunology , Cell Differentiation/immunology , Cell Line , Cell Line, Tumor , Chromatin/immunology , HEK293 Cells , Humans , Immunity, Innate/immunology , Jurkat Cells , Lymphocytes/immunology , THP-1 Cells/immunology
6.
Hum Mol Genet ; 27(15): 2678-2688, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29767720

ABSTRACT

Age-related macular degeneration (AMD) is a progressive disease of the central retina and the leading cause of irreversible vision loss in the western world. The involvement of abnormal complement activation in AMD has been suggested by association of variants in genes encoding complement proteins with disease development. A low-frequency variant (p.P167S) in the complement component C9 (C9) gene was recently shown to be highly associated with AMD; however, its functional outcome remains largely unexplored. In this study, we reveal five novel rare genetic variants (p.M45L, p.F62S, p.G126R, p.T170I and p.A529T) in C9 in AMD patients, and evaluate their functional effects in vitro together with the previously identified (p.R118W and p.P167S) C9 variants. Our results demonstrate that the concentration of C9 is significantly elevated in patients' sera carrying the p.M45L, p.F62S, p.P167S and p.A529T variants compared with non-carrier controls. However, no difference can be observed in soluble terminal complement complex levels between the carrier and non-carrier groups. Comparing the polymerization of the C9 variants we reveal that the p.P167S mutant spontaneously aggregates, while the other mutant proteins (except for C9 p.A529T) fail to polymerize in the presence of zinc. Altered polymerization of the p.F62S and p.P167S proteins associated with decreased lysis of sheep erythrocytes and adult retinal pigment epithelial-19 cells by carriers' sera. Our data suggest that the analyzed C9 variants affect only the secretion and polymerization of C9, without influencing its classical lytic activity. Future studies need to be performed to understand the implications of the altered polymerization of C9 in AMD pathology.


Subject(s)
Complement C9/genetics , Complement C9/metabolism , Genetic Variation , Macular Degeneration/genetics , Animals , Case-Control Studies , Complement C9/pharmacology , Erythrocytes/drug effects , HEK293 Cells , Hemolysis/drug effects , Humans , Polymerization , Polymorphism, Single Nucleotide , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Sheep
7.
J Hum Genet ; 63(1): 93-96, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29215086

ABSTRACT

Most cases of hemolytic uremic syndrome (HUS) are caused by infection with enterohemorrhagic Escherichia coli (EHEC). Genetic defects causing uncontrolled complement activation are associated with the more severe atypical HUS (aHUS). Non-EHEC infections can trigger the disease, however, complement defects predisposing to such infections have not yet been studied. We describe a 2-month-old patient infected with different Gram-negative bacterial species resulting in aHUS. Serum analysis revealed slow complement activation kinetics. Rare variant R229C was found in complement inhibitor vitronectin. Recombinant mutated vitronectin showed enhanced complement inhibition in vitro and may have been a predisposing factor for infection. Our work indicates that genetic changes in aHUS can not only result in uncontrolled complement activation but also increase vulnerability to infections contributing to aHUS.


Subject(s)
Atypical Hemolytic Uremic Syndrome/genetics , Enterohemorrhagic Escherichia coli , Escherichia coli Infections/genetics , Genetic Predisposition to Disease , Point Mutation , Vitronectin/genetics , Atypical Hemolytic Uremic Syndrome/microbiology , Female , Humans , Infant
8.
Immunol Lett ; 189: 73-81, 2017 09.
Article in English | MEDLINE | ID: mdl-28577901

ABSTRACT

The expression and role of CR3 (CD11b/CD18) and CR4 (CD11c/CD18) in B cells are not yet explored in contrast to myeloid cells, where these ß2-integrin type receptors are known to participate in various cellular functions, including phagocytosis, adherence and migration. Here we aimed to reveal the expression and role of CR3 and CR4 in human B cells. In B cells of healthy donors CR3 and CR4 are scarcely expressed. However, two patients with chronic lymphocytic leukemia (CLL) characterized by a peculiar immune-phenotype containing both CD5-positive and CD5-negative B cell populations made possible to study these molecules in distinct B cell subsets. We found that CD11b and CD11c were expressed on both CD5-positive and CD5-negative B cells, albeit to different extents. Our data suggest that these receptors are involved in spreading, since this activity of CpG-activated B cells on fibrinogen could be partially blocked by monoclonal antibodies specific for CD11b or CD11c. CpG-stimulation lead to proliferation of both CD5-positive and CD5-negative B cells of the patients with a less pronounced effect on the CD5-positive cells. In contrast to normal B cells, CLL B cells of both patients reacted to CpG-stimulation with robust IL-10 production. The concomitant, suboptimal stimulus via the BCR and TLR9 exerted either a synergistic enhancing effect or resulted in inhibition of proliferation and IL-10 production of patients' B cells. Our data obtained studying B cells of leukemic patients point to the role of CR3 and probably CR4 in the interaction of tumor cells with the microenvironment and suggest the involvement of IL-10 producing B cells in the pathologic process.


Subject(s)
B-Lymphocytes/physiology , Integrin alphaXbeta2/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Macrophage-1 Antigen/metabolism , Receptors, Antigen, B-Cell/metabolism , Aged , CD18 Antigens/chemistry , CD18 Antigens/metabolism , CD5 Antigens/metabolism , Cells, Cultured , Female , Gene Expression Regulation , Humans , Integrin alphaXbeta2/chemistry , Interleukin-10/metabolism , Macrophage-1 Antigen/chemistry , Toll-Like Receptor 9/metabolism , Tumor Microenvironment
9.
JAMA Ophthalmol ; 135(1): 39-46, 2017 Jan 01.
Article in English | MEDLINE | ID: mdl-27918759

ABSTRACT

IMPORTANCE: In age-related macular degeneration (AMD), rare variants in the complement system have been described, but their functional consequences remain largely unexplored. OBJECTIVES: To identify new rare variants in complement genes and determine the functional effect of identified variants on complement levels and complement regulation in serum samples from carriers and noncarriers. DESIGN, SETTING, AND PARTICIPANTS: This study evaluated affected (n = 114) and unaffected (n = 60) members of 22 families with AMD and a case-control cohort consisting of 1831 unrelated patients with AMD and 1367 control individuals from the European Genetic Database from March 29, 2006, to April 26, 2013, in Nijmegen, the Netherlands, and Cologne, Germany. Exome sequencing data of families were filtered for rare variants in the complement factor H (CFH), complement factor I (CFI), complement C9 (C9), and complement C3 (C3) genes. The case-control cohort was genotyped with allele-specific assays. Serum samples were obtained from carriers of identified variants (n = 177) and age-matched noncarriers (n = 157). Serum concentrations of factor H (FH), factor I (FI), C9, and C3 were measured, and C3b degradation ability was determined. MAIN OUTCOMES AND MEASURES: Association of rare variants in the CFH, CFI, C9, and C3 genes with AMD, serum levels of corresponding proteins, and C3b degradation ability of CFH and CFI variant carriers. RESULTS: The 1831 unrelated patients with AMD had a mean (SD) age of 75.0 (9.4) years, and 60.5% were female. The 1367 unrelated control participants had a mean (SD) age of 70.4 (7.0), and 58.7% were female. All individuals were of European descent. Rare variants in CFH, CFI, C9, and C3 contributed to an increased risk of developing AMD (odds ratio, 2.04; 95% CI, 1.47-2.82; P < .001). CFI carriers had decreased median FI serum levels (18.2 µg/mL in Gly119Arg carriers and 16.2 µg/mL in Leu131Arg carriers vs 27.2 and 30.4 µg/mL in noncarrier cases and controls, respectively; both P < .001). Elevated C9 levels were observed in Pro167Ser carriers (10.7 µg/mL vs 6.6 and 6.1 µg/mL in noncarrier cases and controls, respectively; P < .001). The median FH serum levels were 299.4 µg/mL for CFH Arg175Gln and 266.3 µg/mL for CFH Ser193Leu carriers vs 302.4 and 283.0 µg/mL for noncarrier cases and controls, respectively. The median C3 serum levels were 943.2 µg/mL for C3 Arg161Trp and 946.7 µg/mL for C3 Lys155Gln carriers vs 874.0 and 946.7 µg/mL for noncarrier cases and controls, respectively. The FH and FI levels correlated with C3b degradation in noncarriers (R2 = 0.35 and R2 = 0.31, respectively; both P < .001). CONCLUSIONS AND RELEVANCE: Reduced serum levels were associated with C3b degradation in carriers of CFI but not CFH variants, suggesting that CFH variants affect functional activity of FH rather than serum levels. Carriers of CFH (Arg175Gln and Ser193Leu) and CFI (Gly119Arg and Leu131Arg) variants have an impaired ability to regulate complement activation and may benefit more from complement-inhibiting therapy than patients with AMD in general.


Subject(s)
Complement C3b/genetics , DNA/genetics , Macular Degeneration/genetics , Aged , Aged, 80 and over , Alleles , Complement C3b/metabolism , Exome , Female , Genotype , Germany/epidemiology , Humans , Macular Degeneration/epidemiology , Macular Degeneration/metabolism , Male , Middle Aged , Netherlands/epidemiology , Polymorphism, Single Nucleotide , Prevalence , Retrospective Studies
10.
J Immunol Res ; 2016: 5758192, 2016.
Article in English | MEDLINE | ID: mdl-27981054

ABSTRACT

Complement receptors (CRs) play an integral role in innate immunity and also function to initiate and shape the adaptive immune response. Our earlier results showed that complement receptor type 1 (CR1, CD35) is a potent inhibitor of the B cell receptor- (BCR-) induced functions of human B lymphocytes. Here we show that this inhibition occurs already at the initial steps of B cell activation since ligation of CR1 reduces the BCR-induced phosphorylation of key signaling molecules such as Syk and mitogen activated protein kinases (MAPKs). Furthermore, our data give evidence that although B lymphocytes of active systemic lupus erythematosus (SLE) patients express lower level of CR1, the inhibitory capacity of this complement receptor is still maintained and its ligand-induced clustering results in significant inhibition of the main B cell functions, similar to that found in the case of healthy individuals. Since we have found that reduced CR1 expression of SLE patients does not affect the inhibitory capacity of the receptor, our results further support the therapeutical potential of CD35 targeting the decrease of B cell activation and autoantibody production in autoimmune patients.


Subject(s)
B-Lymphocytes/immunology , Lupus Erythematosus, Systemic/immunology , Receptors, Complement 3b/immunology , Receptors, Complement 3b/metabolism , Adult , Autoantibodies/immunology , B-Lymphocytes/physiology , Cell Proliferation , Cells, Cultured , Female , Flow Cytometry , Humans , Lymphocyte Activation , Male , Middle Aged , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Receptors, Antigen, B-Cell/immunology , Syk Kinase/metabolism
11.
Immunol Rev ; 274(1): 127-140, 2016 11.
Article in English | MEDLINE | ID: mdl-27782338

ABSTRACT

The complement system is a major component of immune defense. Activation of the complement cascade by foreign substances and altered self-structures may lead to the elimination of the activating agent, and during the enzymatic cascade, several biologically active fragments are generated. Most immune regulatory effects of complement are mediated by the activation products of C3, the central component. The indispensable role of C3 in opsonic phagocytosis as well as in the regulation of humoral immune response is known for long, while the involvement of complement in T-cell biology have been revealed in the past few years. In this review, we discuss the immune modulatory functions of C3-derived fragments focusing on their role in processes which have not been summarized so far. The importance of locally synthesized complement will receive special emphasis, as several immunological processes take place in tissues, where hepatocyte-derived complement components might not be available at high concentrations. We also aim to call the attention to important differences between human and mouse systems regarding C3-mediated processes.


Subject(s)
Complement C3/immunology , Liver/immunology , Peptides/immunology , Phagocytosis/immunology , T-Lymphocytes/immunology , Animals , Humans , Immunity, Cellular , Immunity, Humoral , Immunomodulation , Mice , Models, Animal
12.
Immunol Lett ; 178: 37-44, 2016 10.
Article in English | MEDLINE | ID: mdl-27485187

ABSTRACT

B cell functions triggered by the clonally-rearranged antigen-specific B cell receptor (BCR) are regulated by several germ-line encoded receptors - including Toll-like receptors (TLRs) and complement receptors (CRs). Simultaneous or sequential engagement of these structures expressed either on the cell membrane or intracellularly, may fundamentally alter and fine tune activation, antibody and cytokine production of B cells. Here we review the expression and function of TLRs and various C3 fragment binding CRs on B cells, emphasizing their role in different human B cell subsets under physiological and pathological conditions. Studies underlining the importance of the crosstalk between TLRs and CRs in regulating B cell functions are also highlighted.


Subject(s)
B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Complement System Proteins/immunology , Complement System Proteins/metabolism , Immunomodulation , Toll-Like Receptors/metabolism , Animals , Autoimmunity , Disease Susceptibility , Gene Expression Regulation , Humans , Immunity, Innate , Protein Binding , Receptors, Complement/metabolism , Signal Transduction
13.
Cell Mol Life Sci ; 73(23): 4531-4545, 2016 12.
Article in English | MEDLINE | ID: mdl-27125884

ABSTRACT

Tunneling nanotubes (TNTs) are long intercellular connecting structures providing a special transport route between two neighboring cells. To date TNTs have been reported in different cell types including immune cells such as T-, NK, dendritic cells, or macrophages. Here we report that mature, but not immature, B cells spontaneously form extensive TNT networks under conditions resembling the physiological environment. Live-cell fluorescence, structured illumination, and atomic force microscopic imaging provide new insights into the structure and dynamics of B cell TNTs. Importantly, the selective interaction of cell surface integrins with fibronectin or laminin extracellular matrix proteins proved to be essential for initiating TNT growth in B cells. These TNTs display diversity in length and thickness and contain not only F-actin, but their majority also contain microtubules, which were found, however, not essential for TNT formation. Furthermore, we demonstrate that Ca2+-dependent cortical actin dynamics exert a fundamental control over TNT growth-retraction equilibrium, suggesting that actin filaments form the TNT skeleton. Non-muscle myosin 2 motor activity was shown to provide a negative control limiting the uncontrolled outgrowth of membranous protrusions. Moreover, we also show that spontaneous growth of TNTs is either reduced or increased by B cell receptor- or LPS-mediated activation signals, respectively, thus supporting the critical role of cytoplasmic Ca2+ in regulation of TNT formation. Finally, we observed transport of various GM1/GM3+ vesicles, lysosomes, and mitochondria inside TNTs, as well as intercellular exchange of MHC-II and B7-2 (CD86) molecules which may represent novel pathways of intercellular communication and immunoregulation.


Subject(s)
B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Nanotubes/chemistry , Actin Cytoskeleton/metabolism , Animals , Biological Transport , Calcium/metabolism , Cell Line , Cell Membrane/metabolism , Cell Proliferation , Cellular Microenvironment , Flow Cytometry , Humans , Mice , Myosins/metabolism
14.
Immunol Lett ; 168(1): 13-21, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26306739

ABSTRACT

The opportunistic pathogenic yeast Candida albicans employs several mechanisms to interfere with the human complement system. This includes the acquisition of host complement regulators, the release of molecules that scavenge complement proteins or block cellular receptors, and the secretion of proteases that inactivate complement components. Secreted aspartic protease 2 (Sap2) was previously shown to cleave C3b, C4b and C5. C. albicans also recruits the complement inhibitor factor H (FH), but yeast-bound FH can enhance the antifungal activity of human neutrophils via binding to complement receptor type 3 (CR3). In this study, we characterized FH binding to human monocyte-derived macrophages. Inhibition studies with antibodies and siRNA targeting CR3 (CD11b/CD18) and CR4 (CD11c/CD18), as well as analysis of colocalization of FH with these integrins indicated that both function as FH receptors on macrophages. Preincubation of C. albicans yeast cells with FH induced increased production of IL-1ß and IL-6 in macrophages. Furthermore, FH enhanced zymosan-induced production of these cytokines. C. albicans Sap2 cleaved FH, diminishing its complement regulatory activity, and Sap2-treatment resulted in less detectable CR3 and CR4 on macrophages. These data show that FH enhances the activation of human macrophages when bound on C. albicans. However, the fungus can inactivate both FH and its receptors on macrophages by secreting Sap2, which may represent an additional means for C. albicans to evade the host innate immune system.


Subject(s)
Aspartic Acid Endopeptidases/immunology , Candida albicans/immunology , Complement Factor H/immunology , Fungal Proteins/immunology , Integrin alphaXbeta2/immunology , Macrophage-1 Antigen/immunology , Aspartic Acid Endopeptidases/metabolism , Blotting, Western , CD11b Antigen/genetics , CD11b Antigen/immunology , CD11b Antigen/metabolism , CD11c Antigen/genetics , CD11c Antigen/immunology , CD11c Antigen/metabolism , CD18 Antigens/genetics , CD18 Antigens/immunology , CD18 Antigens/metabolism , Candida albicans/enzymology , Candida albicans/physiology , Cell Line, Tumor , Cells, Cultured , Complement Factor H/metabolism , Cytokines/immunology , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Fungal Proteins/metabolism , Host-Pathogen Interactions/immunology , Humans , Integrin alphaXbeta2/genetics , Integrin alphaXbeta2/metabolism , Macrophage-1 Antigen/genetics , Macrophage-1 Antigen/metabolism , RNA Interference
15.
Cell Mol Life Sci ; 72(11): 2223-36, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25543269

ABSTRACT

B cells are efficiently activated by CpG oligodeoxynucleotides (ODNs) to produce pro-inflammatory cytokines and antibody (Ab). Here, we describe a so far unidentified, spleen tyrosine kinase (Syk)-dependent pathway, which is indispensable for CpG-induced human B cell activation. We show that triggering of B cells by CpG results in Syk and src kinase phosphorylation, proliferation, as well as cytokine and Ab production independent of the BCR. Notably, all these functions are abrogated when Syk is inhibited. We demonstrate that CpG-induced Syk activation originates from the cell surface in a TLR9-dependent manner. While inhibition of Syk does not influence the uptake of CpG ODNs, activation of the kinase is a prerequisite for the delivery of CpG into TLR9-containing endolysosomes and for the CpG-induced up-regulation of TLR9 expression. Our results reveal an alternative, Syk-dependent pathway of CpG-induced B cell stimulation, which is initiated at the plasma membrane and seems to be an upstream requirement for endosomal TLR9-driven B cell proliferation and differentiation.


Subject(s)
B-Lymphocytes/immunology , CpG Islands/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Oligodeoxyribonucleotides/metabolism , Protein-Tyrosine Kinases/metabolism , Toll-Like Receptor 9/metabolism , Cell Differentiation/immunology , Cell Proliferation , Humans , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Lymphocyte Activation/genetics , Mitogen-Activated Protein Kinases/metabolism , Myeloid Differentiation Factor 88/antagonists & inhibitors , Myeloid Differentiation Factor 88/metabolism , NF-kappa B/metabolism , Oligodeoxyribonucleotides/immunology , Phosphorylation , Protein Binding , Protein-Tyrosine Kinases/antagonists & inhibitors , RNA Interference , RNA, Small Interfering , Syk Kinase , Toll-Like Receptor 9/biosynthesis , Toll-Like Receptor 9/genetics , Up-Regulation , src-Family Kinases/metabolism
16.
Int Immunol ; 25(1): 25-33, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22962438

ABSTRACT

The involvement of B cells, complement activation and subsequent immune complex deposition has all been implicated in the pathogenesis of rheumatoid arthritis (RA). Although the reduced expression of complement receptor 1 (CR1, CD35) and 2 (CR2, CD21) on the B cells of RA patients has been known for a long time, their exact role in B-cell tolerance and autoimmunity is not yet fully understood. To get a deeper insight into the possible mechanisms, we studied the expression and function of CR1 and CR2 on various subsets of B cells of healthy donors and RA patients at various stages of the disease by FACS analysis, (3)H-thymidine incorporation and ELISA. We found that CD19(+)CD27(-) naive B cells up-regulate the expression of the inhibitory CR1 during differentiation to CD19(+)CD27(+) memory B cells both in healthy donors and in RA patients, whereas the expression of the activatory CR2 is down-regulated. This clearly demonstrates that the expression of these two antagonistic complement receptors is regulated differentially during the development of human B cells, a phenomenon which may influence the maintenance of peripheral B-cell tolerance. Our functional studies show that after clustering CR1 both by its natural ligand and To5 mAb, the inhibitory function of CD35 is maintained in RA patients, despite its significantly reduced expression compared with healthy individuals. Besides blocking B-cell receptor-induced proliferation, CR1 inhibits the differentiation of B cells to plasmablasts and their immunoglobulin production. Since the reduced expression of CR1 in RA patients does not affect its inhibitory function, this receptor might serve as a new target for therapeutical interventions.


Subject(s)
Arthritis, Rheumatoid/immunology , B-Lymphocytes/immunology , Immunity, Humoral , Receptors, Complement 3b/immunology , Receptors, Complement 3d/immunology , Adult , Aged , Antigens, CD19/genetics , Antigens, CD19/immunology , Arthritis, Rheumatoid/pathology , Autoimmunity , B-Lymphocytes/pathology , Case-Control Studies , Cell Differentiation , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Gene Expression Regulation/immunology , Humans , Immunologic Memory , Male , Middle Aged , Peripheral Tolerance , Receptors, Antigen, B-Cell/genetics , Receptors, Antigen, B-Cell/immunology , Receptors, Complement 3b/genetics , Receptors, Complement 3d/genetics , Signal Transduction , Tumor Necrosis Factor Receptor Superfamily, Member 7/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 7/immunology
17.
Immunol Lett ; 143(1): 131-6, 2012 Mar 30.
Article in English | MEDLINE | ID: mdl-22553783

ABSTRACT

Although the complement system is thought to be mainly involved in innate immunity and in the humoral arm of adaptive responses, evidence implicating that complement impacts T cell responses are accumulating recently. The role of the various activation products of the major complement component C3 were mainly studied so far in animal systems, and investigations regarding the effect of different C3-fragments on human T cells are sparse. Here we show that anti-CD3 activated human T lymphocytes derived from the blood and tonsil of healthy individuals produce C3, and the major cleavage fragment that appears on the T cell surface is iC3b. Based on studies carried out in allogenic system we demonstrate that the T cell membrane bound iC3b binds to the CR3 and probably to CR4 receptors expressed on monocyte-derived dendritic cells, and this interaction leads to significantly enhanced T-cell proliferation. Since neither C3aR and nor C3a binding could be detected on the membrane of anti-CD3 activated T cells, our findings indicate that in humans ­ in contrast to mice ­ the C3a peptide is most probably not involved directly in the T cell activation process.


Subject(s)
Complement C3b/immunology , Lymphocyte Activation , T-Lymphocytes/immunology , Animals , Cell Line , Cell Proliferation , Dendritic Cells/cytology , Dendritic Cells/immunology , Humans , Mice
18.
Mol Immunol ; 46(14): 2767-73, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19559484

ABSTRACT

The involvement of complement in the development and regulation of antibody responses under both healthy and pathological conditions is known for long. Unravelling the molecular mechanisms underlying the events however is still in progress. This review focuses on the role of complement receptors CR1 (CD35) and CR2 (CD21) expressed on T and B cells. Alteration in the expression and function of these receptors may contribute to the initiation and maintenance of immune complex mediated autoimmune diseases such as systemic lupus erythematosus and rheumatoid arthritis. Recent data regarding complement receptor expression on T lymphocytes and on memory B cells are also discussed.


Subject(s)
Autoimmune Diseases/immunology , B-Lymphocytes/immunology , Receptors, Complement 3b/immunology , Receptors, Complement 3d/immunology , T-Lymphocytes/immunology , Antigen-Antibody Complex/immunology , Antigen-Antibody Complex/metabolism , Apoptosis/immunology , Autoimmunity , B-Lymphocytes/metabolism , Humans , Receptors, Complement 3b/metabolism , Receptors, Complement 3d/metabolism , T-Lymphocytes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...