Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters










Publication year range
1.
Circ Res ; 124(2): 243-255, 2019 01 18.
Article in English | MEDLINE | ID: mdl-30582450

ABSTRACT

RATIONALE: Endothelial colony forming cells (ECFCs) or late blood outgrowth endothelial cells can be isolated from human cord or peripheral blood, display properties of endothelial progenitors, home into ischemic tissues and support neovascularization in ischemic disease models. OBJECTIVE: To assess the functions of CYTL1 (cytokine-like 1), a factor we found preferentially produced by ECFCs, in regard of vessel formation. METHODS AND RESULTS: We show by transcriptomic analysis that ECFCs are distinguished from endothelial cells of the vessel wall by production of high amounts of CYTL1. Modulation of expression demonstrates that the factor confers increased angiogenic sprouting capabilities to ECFCs and can also trigger sprouting of mature endothelial cells. The data further display that CYTL1 can be induced by hypoxia and that it functions largely independent of VEGF-A (vascular endothelial growth factor-A). By recombinant production of CYTL1 we confirm that the peptide is indeed a strong proangiogenic factor and induces sprouting in cellular assays and functional vessel formation in animal models comparable to VEGF-A. Mass spectroscopy corroborates that CYTL1 is specifically O-glycosylated on 2 neighboring threonines in the C-terminal part and this modification is important for its proangiogenic bioactivity. Further analyses show that the factor does not upregulate proinflammatory genes and strongly induces several metallothionein genes encoding anti-inflammatory and antiapoptotic proteins. CONCLUSIONS: We conclude that CYTL1 can mediate proangiogenic functions ascribed to endothelial progenitors such as ECFCs in vivo and may be a candidate to support vessel formation and tissue regeneration in ischemic pathologies.


Subject(s)
Angiogenic Proteins/metabolism , Autocrine Communication , Blood Proteins/metabolism , Corneal Neovascularization , Cytokines/metabolism , Endothelial Progenitor Cells/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Neovascularization, Physiologic , Paracrine Communication , Angiogenic Proteins/genetics , Animals , Blood Proteins/genetics , Cell Hypoxia , Cytokines/genetics , Disease Models, Animal , Female , Glycosylation , HEK293 Cells , Human Umbilical Vein Endothelial Cells/transplantation , Humans , Male , Mice, Inbred C57BL , Mice, SCID , Secretory Pathway , Signal Transduction , Spheroids, Cellular , Vascular Endothelial Growth Factor A/metabolism
2.
J Biol Chem ; 293(22): 8600-8613, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29669808

ABSTRACT

The plasminogen system is essential for dissolution of fibrin clots, and in addition, it is involved in a wide variety of other physiological processes, including proteolytic activation of growth factors, cell migration, and removal of protein aggregates. On the other hand, uncontrolled plasminogen activation contributes to many pathological processes (e.g. tumor cells' invasion in cancer progression). Moreover, some virulent bacterial species (e.g. Streptococci or Borrelia) bind human plasminogen and hijack the host's plasminogen system to penetrate tissue barriers. Thus, the conversion of plasminogen to the active serine protease plasmin must be tightly regulated. Here, we show that human lactoferrin, an iron-binding milk glycoprotein, blocks plasminogen activation on the cell surface by direct binding to human plasminogen. We mapped the mutual binding sites to the N-terminal region of lactoferrin, encompassed also in the bioactive peptide lactoferricin, and kringle 5 of plasminogen. Finally, lactoferrin blocked tumor cell invasion in vitro and also plasminogen activation driven by Borrelia Our results explain many diverse biological properties of lactoferrin and also suggest that lactoferrin may be useful as a potential tool for therapeutic interventions to prevent both invasive malignant cells and virulent bacteria from penetrating host tissues.


Subject(s)
Borrelia/metabolism , Fibrinolysin/metabolism , Fibrinolysis , Lactoferrin/metabolism , Plasminogen/antagonists & inhibitors , Streptococcus/metabolism , Cell Movement , Cells, Cultured , Crystallography, X-Ray , Humans , Lactoferrin/chemistry , Lactoferrin/genetics , Plasminogen/metabolism , Protein Conformation
3.
Clin Biochem ; 50(7-8): 444-451, 2017 May.
Article in English | MEDLINE | ID: mdl-28041932

ABSTRACT

OBJECTIVES: Diamine oxidase (DAO) is essential for extracellular degradation of histamine. For decades activity assays with inherent limitations were used to quantify the relative amounts of DAO. No reference DAO standard is available. Absolute DAO amounts cannot be determined. Controversy exists, whether DAO is circulating or not in non-pregnant individuals. The role of DAO as biomarker in various diseases is ambiguous. It is not clear, whether precise quantification of human DAO antigen using commercially available enzyme-linked immunosorbent assays (ELISAs) is possible. The objective was to develop a precise and robust ELISA to quantify DAO in various biological fluids. DESIGN AND METHODS: A research prototype ELISA was established using a mouse monoclonal antibody for capturing and a polyclonal rabbit serum IgG fraction for the detection of human DAO. The limit of blank (LoB), limit of detection (LoD) and estimated limit of quantification (eLoQ) and normal DAO concentrations in serum and plasma were determined. RESULTS: The LoB, LoD and eLoQ derived from 42 standard curves are 0.27, 0.48 and 0.7ng/mL respectively. The detection range using the LoD as the lower and the highest DAO standard as the upper boundary is 0.5 to 450ng/mL. Serum and plasma mean/median concentrations are between 0.5 and 1.5ng/mL in healthy volunteers (n=58) and mastocytosis patients (n=19) and plateau at approximately 145ng/mL (n=16) during pregnancy. Accurate quantification was not influenced by heparin (DAO is a heparin-binding protein), lipaemic or hemolytic serum. The measured DAO antigen concentrations are in close agreement with published enzymatic activity data using radioactive putrescine as substrate. CONCLUSIONS: This research prototype ELISA is able to reliably and accurately quantify human DAO in different biological fluids. The potential of DAO as biomarker in various diseases can be evaluated.


Subject(s)
Amine Oxidase (Copper-Containing)/blood , Animals , Antibodies, Monoclonal, Murine-Derived/chemistry , Biomarkers/blood , Enzyme-Linked Immunosorbent Assay/methods , Female , Humans , Immunoglobulin G/chemistry , Male , Mice , Pregnancy , Rabbits
4.
J Gen Virol ; 98(3): 471-485, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27959783

ABSTRACT

Protein phosphorylation has important regulatory functions in cell homeostasis and is tightly regulated by kinases and phosphatases. The tegument of human cytomegalovirus (CMV) contains not only several proteins reported to be extensively phosphorylated but also cellular protein phosphatases (PP1 and PP2A). To investigate this apparent inconsistency, we evaluated the phosphorylation status of the tegument proteins pUL32 and pp65 by enzymatic dephosphorylation and MS. Enzymatic dephosphorylation with bacterial λ phosphatase, but not with PP1, shifted the pUL32-specific signal on reducing SDS-PAGE from ~150 to ~148 kDa, a mass still much larger than the ~118 kDa obtained from our diffusion studies and from the calculated protein mass of ~113 kDa. Remarkably, inhibition of phosphatases through treatment with the phosphatase inhibitors calyculin A and okadaic acid resulted in a shift to ~190 or ~180 kDa, respectively, indicating that a considerable number of potential phosphorylated residues on pUL32 are not phosphorylated under normal conditions. MS revealed a general state of hypophosphorylation of CMV phosphoproteins with only 17 phosphorylated residues detected on pUL32 and 19 on pp65, respectively. Moreover, bioinformatics analysis shows that the C-terminal two-thirds of pUL32 are intrinsically disordered and that most phosphorylations map to this region. In conclusion, we show that important CMV tegument proteins are indeed phosphorylated, though to a lesser extent than previously reported, and the difference in mobility on SDS-PAGE and calculated mass of pUL32 may not be attributed to phosphorylation but more likely due to the partially intrinsically disordered nature of pUL32.


Subject(s)
Cytomegalovirus/metabolism , Phosphoproteins/metabolism , Viral Matrix Proteins/metabolism , Electrophoresis, Polyacrylamide Gel , Enzyme Inhibitors/pharmacology , Humans , Marine Toxins , Okadaic Acid/pharmacology , Oxazoles/pharmacology , Phosphoprotein Phosphatases/antagonists & inhibitors , Phosphorylation , Protein Processing, Post-Translational
5.
Eur J Immunol ; 47(1): 193-205, 2017 01.
Article in English | MEDLINE | ID: mdl-27718235

ABSTRACT

The cytoplasmic tail of CD45 (ct-CD45) is proteolytically cleaved and released upon activation of human phagocytes. It acts on T cells as an inhibitory, cytokine-like factor in vitro. Here, we show that ct-CD45 is abundant in human peripheral blood plasma from healthy adults compared with plasma derived from umbilical cord blood and plasma from patients with rheumatoid arthritis or systemic lupus erythematosus. Plasma depleted of ct-CD45 enhanced T-cell proliferation, while addition of exogenous ct-CD45 protein inhibited proliferation and reduced cytokine production of human T lymphocytes in response to TCR signaling. Inhibition of T-cell proliferation by ct-CD45 was overcome by costimulation via CD28. T-cell activation in the presence of ct-CD45 was associated with an upregulation of the quiescence factors Schlafen family member 12 (SLFN12) and Krueppel-like factor 2 (KLF2) as well as of the cyclin-dependent kinase (CDK) inhibitor p27kip1. In contrast, positive regulators of the cell cycle such as cyclin D2 and D3 as well as CDK2 and CDK4 were found to be downregulated in response to ct-CD45. In summary, we demonstrate that ct-CD45 is present in human plasma and sets the threshold of T-cell activation.


Subject(s)
Cell Cycle , Leukocyte Common Antigens/blood , Protein Domains , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Adult , Aged , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/metabolism , Biomarkers , Cell Cycle/genetics , Cell Cycle/immunology , Female , Gene Expression Profiling , Gene Expression Regulation , Humans , Immunomodulation , Immunophenotyping , Leukocyte Common Antigens/chemistry , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/metabolism , Lymphocyte Activation/immunology , Male , Middle Aged , Phenotype , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
6.
Biomark Med ; 10(8): 797-810, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27416002

ABSTRACT

AIM: Oxidized phospholipids (OxPL) are the major pathogenic component of oxidized low-density lipoproteins (OxLDL). Endogenous anti-OxPL activity, defined as the ability to neutralize adverse effects of oxidized lipids, may have biomarker potential. METHODS & RESULTS: Using two anti-OxPL monoclonal antibodies (commercial mAB-E06 and custom mAB-509) we developed a novel ELISA that measures the global capacity of plasma to inactivate OxPL. Preincubation of OxLDL with plasma inhibits its binding of anti-OxPL mABs. This phenomenon ('masking') reflects anti-OxPL plasma activity. A pilot clinical application of the assay revealed reduced anti-OxPL activity in hypertension, coronary artery disease, acute coronary syndrome and diabetes. CONCLUSION: Inadequate anti-OxPL protection may contribute to cardiovascular disease and have biomarker potential in conditions associated with abnormal lipid peroxidation.


Subject(s)
Blood Chemical Analysis/methods , Enzyme-Linked Immunosorbent Assay , Phospholipids/blood , Acute Coronary Syndrome/blood , Acute Coronary Syndrome/pathology , Adult , Aged , Antibodies, Monoclonal/immunology , Coronary Artery Disease/blood , Coronary Artery Disease/pathology , Diabetes Mellitus/blood , Diabetes Mellitus/pathology , Female , Humans , Hypertension/blood , Hypertension/pathology , Lipoproteins, LDL/blood , Male , Middle Aged , Oxidation-Reduction , Phospholipids/immunology
7.
Immunology ; 149(3): 280-296, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27392084

ABSTRACT

Co-receptors, being either co-stimulatory or co-inhibitory, play a pivotal role in T-cell immunity. Several studies have indicated that CD43, one of the abundant T-cell surface glycoproteins, acts not only as a potent co-receptor but also as a negative regulator for T-cell activation. Here we demonstrate that co-stimulation of human peripheral blood (PB) T cells through two distinct CD43 epitopes recognized by monoclonal antibodies (mAb) CD43-6E5 (T6E5-act ) and CD43-10G7 (T10G7-act ) potently induced T-cell proliferation. However, T-cell co-stimulation through two CD43 epitopes differentially regulated activation of nuclear factor of activated T cells (NFAT) and nuclear factor-κB (NF-κB) transcription factors, T-cell cytokine production and effector function. T6E5-act produced high levels of interleukin-22 (IL-22) and interferon-γ (IFN-γ) similar to T cells activated via CD28 (TCD28-act ), whereas T10G7-act produced low levels of inflammatory cytokines but higher levels of regulatory cytokines transforming growth factor-ß (TGF-ß) and interleukin-35 (IL-35). Compared with T6E5-act or to TCD28-act , T10G7-act performed poorly in response to re-stimulation and further acquired a T-cell suppressive function. T10G7-act did not directly inhibit proliferation of responder T cells, but formed stable heterotypic clusters with dendritic cells (DC) via CD2 to constrain activation of responder T cells. Together, our data demonstrate that CD43 is a unique and polarizing regulator of T-cell function.


Subject(s)
Dendritic Cells/immunology , Epitopes, T-Lymphocyte/metabolism , Leukosialin/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , CD28 Antigens/metabolism , Cell Differentiation , Cells, Cultured , Costimulatory and Inhibitory T-Cell Receptors/metabolism , Cytokines/metabolism , Humans , Immune Tolerance , Leukosialin/immunology , NF-kappa B/metabolism , NFATC Transcription Factors/metabolism , Receptor Cross-Talk , Receptors, Antigen, T-Cell/metabolism , Signal Transduction
8.
J Immunol Methods ; 430: 10-20, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26780292

ABSTRACT

Engagement of the T cell receptor complex reprograms T cells for proliferation, cytokine production and differentiation towards effector cells. This process depends on activating costimulatory signals and is counteracted by coinhibitory molecules. Three transcription factors, namely NF-κB, NFAT and AP-1, have a major role in inducing the transcriptional program that is required for T cell activation and differentiation. Here we describe the generation of a triple parameter reporter based on the human Jurkat T cell line, where response elements for NF-κB, NFAT and AP-1 drive the expression of the fluorescent proteins CFP, eGFP and mCherry, respectively. The emission spectra of these proteins allow simultaneous assessment of NF-κB, NFAT and AP-1 activity in response to stimulation. Ligation of the TCR complex induced moderate reporter activity, which was strongly enhanced upon coengagement of the costimulatory receptors CD2 or CD28. Moreover, we have generated and tested triple parameter reporter cells that harbor costimulatory and inhibitory receptors not endogenously expressed in the Jurkat cells. In these experiments we could show that engagement of the costimulatory molecule 4-1BB enhances NF-κB and AP-1 activity, whereas coinhibition via PD-1 or BTLA strongly reduced the activation of NF-κB and NFAT. Engagement of BTLA significantly inhibited AP-1, whereas PD-1 had little effect on the activation of this transcription factor. Our triple parameter reporter T cell line is an excellent tool to assess the effect of costimulatory and coinhibitory receptors on NF-κB, NFAT and AP-1 activity and has a wide range of applications beyond the evaluation of costimulatory pathways.


Subject(s)
Lymphocyte Activation , NF-kappa B/analysis , NFATC Transcription Factors/analysis , Transcription Factor AP-1/analysis , CD2 Antigens/physiology , CD28 Antigens , CD3 Complex/metabolism , Genes, Reporter , Humans , Jurkat Cells , Luminescent Proteins , NF-kappa B/genetics , NF-kappa B/metabolism , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , Signal Transduction , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism , Red Fluorescent Protein
9.
In Vitro Cell Dev Biol Anim ; 50(1): 56-65, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23982913

ABSTRACT

The Grey horse phenotype, caused by a 4.6 kb duplication in Syntaxin 17, is strongly associated with high incidence of melanoma. In contrast to most human melanomas with an early onset of metastasis, the Grey horse melanomas have an extended period of benign growth, after which 50% or more eventually undergo progression and may metastasize. In efforts to define changes occurring during Grey horse melanoma progression, we established an in vitro model comprised of two cell lines, HoMel-L1 and HoMel-A1, representing a primary and a metastatic stage of the melanoma, respectively. The cell lines were examined for their growth and morphological characteristics, in vitro and in vivo oncogenic potential, chromosome numbers, and expression of melanocytic antigens and tumor suppressors. Both cell lines exhibited malignant characteristics; however, the metastatic HoMel-A1 showed a more aggressive phenotype characterized by higher proliferation rates, invasiveness, and a stronger tumorigenic potential both in vitro and in vivo. HoMel-A1 displayed a near-haploid karyotype, whereas HoMel-L1 was near-diploid. The cell lines expressed melanocytic lineage markers such as TYR, TRP1, MITF, PMEL, ASIP, MC1R, POMC, and KIT. The tumor suppressor p53 was strongly expressed in both cell lines, while the tumor suppressors p16 and PTEN were absent in HoMel-A1, potentially implicating significance of these pathways in the melanoma progression. This in vitro model system will not only aid in understanding of the Grey horse melanoma pathogenesis, but also in unraveling the steps during melanoma progression in general as well as being an invaluable tool for development of new therapeutic strategies.


Subject(s)
Cell Line, Tumor , Horses , Melanoma/veterinary , Animals , Cell Proliferation , Chromosomes, Mammalian , Karyotype , Melanoma/genetics , Melanoma/pathology , Neoplasm Metastasis/genetics , Neoplasm Metastasis/pathology
10.
PLoS One ; 7(10): e46776, 2012.
Article in English | MEDLINE | ID: mdl-23056446

ABSTRACT

Langerhans cells (LCs) are suspected to initiate inflammatory immune responses to contact allergens and pathogenic bacteria. In chronic infectious diseases, programmed death ligand (PD-L) 1 exhibits both inhibitory and costimulatory functions on T cell-mediated activation and tolerance. Here, we investigated the effects of contact allergens and bacterial stimuli on PD-L1 expression in LCs and the effects of altered PD-L1 expression on cytokine release of subsequently cocultured T cells. Monocyte-derived LCs (MoLCs), LCs, and skin sections of patients suffering from allergic contact dermatitis were challenged with nickel and then analyzed for PD-L1 expression by confocal laser scanning microscopy and flow cytometry. In blocking experiments, we found that the release of Th cell specific cytokines was dependent on both stimulation of LCs and inhibition of PD-L1-PD-1 interactions. Stimulation with peptidoglycan (PGN) or lipopolysaccharide (LPS) and blockage of PD-L1 with a specific antibody triggered the release of high levels of IL-17, IL-22, TNF-α, and IFN-γ in CD4(+)T cells. If nickel was used as a stimulus, blockage of PD-L1 led to high amounts of TNF-α and IL-22. A closer look revealed PD-L1-dependent upregulation of IL-17 secretion in FACS-sorted CCR6(+)/CCR4(+) T memory cells. In the presence of anti-PD-L1, PGN induced secretion of IFN-γ and IL-17 in total CCR6(+) cells, while nickel triggered secretion of IFN-γ and IL-17 exclusively in CCR6(+)/CCR4(+) cells. Our findings suggest that PD-L1 on LCs plays a crucial role in type IV allergic reactions and in response to bacterial stimuli by controlling the nature of inflammatory Th cell responses.


Subject(s)
Bacterial Infections/metabolism , Langerhans Cells/metabolism , Nickel/immunology , Programmed Cell Death 1 Receptor/metabolism , T-Lymphocytes, Helper-Inducer/metabolism , Bacterial Infections/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cells, Cultured , Cytokines/metabolism , Dermatitis, Allergic Contact/immunology , Dermatitis, Allergic Contact/metabolism , Flow Cytometry , Humans , Immunohistochemistry , In Vitro Techniques , Interferon-gamma/metabolism , Interleukin-17/metabolism , Interleukins/metabolism , Microscopy, Confocal , Microscopy, Fluorescence , T-Lymphocytes, Helper-Inducer/immunology , Tumor Necrosis Factor-alpha/metabolism , Interleukin-22
11.
Transplantation ; 93(1): 32-40, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-22186936

ABSTRACT

BACKGROUND: Antithymocyte globulins (ATGs) are used to prevent and treat allograft rejection and graft versus host disease. They are purified IgG fractions derived from rabbits immunized with the Jurkat T-cell line (ATG-Fresenius) or thymus cells (Thymoglobulin). Differences not only in the amounts of leukocyte reactive antibodies but also in the antigens targeted by ATGs could potentially affect the clinical efficacy of different batches of these polyclonal antibody preparations. METHODS: Four batches of ATG-Fresenius and Thymoglobulin were compared regarding their capacity to interact with human leukocytes from healthy donors and kidney transplant recipients. Using flow cytometric assays, we analyzed the reactivity of these ATG preparations with Jurkat cells and with primary leukocytes. In addition, ATGs derived from different batches were probed with a panel of cell lines expressing high levels of ATG antigens. Their ability to mediate complement-mediated lysis of human monocytes and lymphocytes was also compared. RESULTS: Binding studies to leukocyte antigens and functional analysis pointed to a high conformity among different batches in both ATG preparations. CONCLUSIONS: From our in vitro data, it can be expected that ATGs derived from different batches will not differ in their clinical efficacy. Furthermore, the methods described in this study allow for a reliable analysis of ATG batches.


Subject(s)
Antilymphocyte Serum/immunology , Cytotoxicity Tests, Immunologic , Immunomodulation/immunology , Leukocytes, Mononuclear/immunology , Adult , Aged , Animals , Antilymphocyte Serum/pharmacology , Cell Death/drug effects , Cell Line , Cells, Cultured , Humans , In Vitro Techniques , Jurkat Cells/cytology , Jurkat Cells/immunology , Kidney Transplantation/immunology , Leukocytes, Mononuclear/cytology , Lymphocytes/cytology , Lymphocytes/drug effects , Mice , Middle Aged , Monocytes/cytology , Monocytes/drug effects , Rabbits , Thymus Gland/cytology , Thymus Gland/immunology , Treatment Outcome
12.
Immunol Lett ; 140(1-2): 74-80, 2011 Oct 30.
Article in English | MEDLINE | ID: mdl-21756939

ABSTRACT

Immunosuppression is an important treatment modality in transplantation and human diseases that are associated with aberrant T cell activation. There are considerable differences regarding the cellular processes targeted by the immunosuppressive drugs that are in clinical use. Drugs like azathioprine (Aza) mainly act by halting proliferation of fast dividing cells, whereas others like cyclosporine A (CsA) specifically target signaling pathways in T cells. Since the outcome of T cell responses critically depends on the quality and strength of costimulatory signals, this study has addressed the interplay between costimulation and the immunosuppressive agents CsA and Aza during the in vitro activation of human T cells. We used an experimental system that allows analyzing T cells activated in the presence of selected costimulatory ligands to study T cells stimulated via CD28, CD2, LFA-1, ICOS or 4-1BB. The mean inhibitory concentrations (IC(50)) for Aza and CsA were determined for the proliferation of T cells receiving different costimulatory signals as well as for T cells activated in the absence of costimulation. CD28 signals but not costimulation via CD2, 4-1BB, ICOS or LFA-1 greatly increased the IC(50) for CsA. By contrast, the inhibitory effects of Aza were not influenced by T cell costimulatory signals. Our results might have implications for combining standard immunosuppressive drugs with CTLA-4Ig fusion proteins, which act by blocking CD28 costimulation.


Subject(s)
Azathioprine/pharmacology , CD28 Antigens/metabolism , Cyclosporine/pharmacology , Immunosuppression Therapy , T-Lymphocytes/drug effects , 4-1BB Ligand/immunology , 4-1BB Ligand/metabolism , CD2 Antigens/immunology , CD2 Antigens/metabolism , CD28 Antigens/immunology , Cell Proliferation/drug effects , Cells, Cultured , Humans , Inducible T-Cell Co-Stimulator Protein/immunology , Inducible T-Cell Co-Stimulator Protein/metabolism , Lymphocyte Activation/drug effects , Lymphocyte Function-Associated Antigen-1/immunology , Lymphocyte Function-Associated Antigen-1/metabolism , Receptor Cross-Talk/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology
13.
J Immunol ; 186(9): 5333-44, 2011 May 01.
Article in English | MEDLINE | ID: mdl-21451110

ABSTRACT

Allergic inflammation is based on the cross-linking of mast cell and basophil-bound IgE Abs and requires at least two binding sites for IgE on allergens, which are difficult to characterize because they are often conformational in nature. We studied the IgE recognition of birch pollen allergen Bet v 1, a major allergen for >100 million allergic patients. Monoclonal and polyclonal Abs raised against Bet v 1-derived peptides were used to compete with allergic patients' IgE binding to Bet v 1 to search for sequences involved in IgE recognition. Strong inhibitions of patients' IgE binding to Bet v 1 (52-75%) were obtained with mAbs specific for two peptides comprising aa 29-58 (P2) and aa 73-103 (P6) of Bet v 1. As determined by surface plasmon resonance, mAb2 specific for P2 and mAb12 specific for P6 showed high affinity, but only polyclonal rabbit anti-P2 and anti-P6 Abs or a combination of mAbs inhibited allergen-induced basophil degranulation. Thus, P2 and P6 define a surface patch on the Bet v 1 allergen, which allows simultaneous binding of several different IgE Abs required for efficient basophil and mast cell activation. This finding explains the high allergenic activity of the Bet v 1 allergen. The approach of using peptide-specific Abs for the mapping of conformational IgE epitopes on allergens may be generally applicable. It may allow discriminating highly allergenic from less allergenic allergen molecules and facilitate the rational design of active and passive allergen-specific immunotherapy strategies.


Subject(s)
Antibodies, Monoclonal/immunology , Antibody Specificity/immunology , Antigens, Plant/immunology , Epitope Mapping/methods , Epitopes, B-Lymphocyte/immunology , Immunoglobulin E/immunology , Amino Acid Sequence , Animals , Antigens, Plant/chemistry , Basophils/immunology , Betula/immunology , Binding, Competitive , Cross Reactions , Enzyme-Linked Immunosorbent Assay , Epitopes, B-Lymphocyte/chemistry , Humans , Hypersensitivity/immunology , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Peptides/chemistry , Peptides/immunology , Pollen/immunology , Rabbits , Sequence Homology, Amino Acid
14.
Clin Immunol ; 139(2): 199-207, 2011 May.
Article in English | MEDLINE | ID: mdl-21414849

ABSTRACT

The therapeutic CD11a antibody Efalizumab interferes with psoriasis pathogenesis by blocking T cell activation and migration. We have performed a detailed analysis on its effects during the activation of human T cells and found that the capability of Efalizumab to inhibit proliferation and cytokine production of T cells critically depends on the quality and quantity of costimulatory signals. Efalizumab potently inhibited the proliferation and cytokine production of human T cells costimulated via ICOS, OX40, CD27 or 4-1BB, but did not significantly inhibit T cells that received stimuli via CD2 or CD28. The capacity of CD2 and CD28 signals to interfere with the T cell inhibitory effects of Efalizumab was also observed upon stimulation of T cells with allogeneic DC. Furthermore, studies with T cells from psoriasis patients indicated that Efalizumab therapy induces inhibition of T cell responses that can be reverted by CD2 or CD28 signals.


Subject(s)
Antibodies, Monoclonal/pharmacology , CD11a Antigen/immunology , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , 4-1BB Ligand/pharmacology , Abatacept , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Antigen Presentation/immunology , Antigens, CD/pharmacology , B7-1 Antigen/immunology , CD28 Antigens/immunology , CD3 Complex/immunology , CD58 Antigens/immunology , Cell Proliferation/drug effects , Dendritic Cells/immunology , Humans , Immunoconjugates/pharmacology , Immunosuppressive Agents/pharmacology , Immunosuppressive Agents/therapeutic use , Inducible T-Cell Co-Stimulator Ligand , Intercellular Adhesion Molecule-1/metabolism , Interferon-gamma/metabolism , Interleukin-10/metabolism , Interleukin-2/pharmacology , Interleukin-4/metabolism , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Lymphocyte Function-Associated Antigen-1/metabolism , Protein Binding/drug effects , Protein Binding/immunology , Psoriasis/drug therapy , Psoriasis/immunology , T-Lymphocytes/metabolism
15.
J Immunol Methods ; 362(1-2): 131-41, 2010 Oct 31.
Article in English | MEDLINE | ID: mdl-20858499

ABSTRACT

It is well established that full activation of T cells requires the interaction of the TCR complex with the peptide-MHC complex (Signal 1) and additional signals (Signal 2). These second signals are generated by the interaction of costimulatory ligands expressed on antigen presenting cells with activating receptors on T cells. In addition, T cell responses are negatively regulated by inhibitory costimulatory pathways. Since professional antigen presenting cells (APC) harbour a plethora of stimulating and inhibitory surface molecules, the contribution of individual costimulatory molecules is difficult to assess on these cells. We have developed a system of stimulator cells that can give signal 1 to human T cells via a membrane bound anti-CD3 antibody fragment. By expressing human costimulatory ligands on these cells, their role in T cell activation processes can readily be analyzed. We demonstrate that T cell stimulator cells are excellent tools to study various aspects of human T cell costimulation, including the effects of immunomodulatory drugs or how costimulatory signals contribute to the in vitro expansion of T cells. T cell stimulator cells are especially suited for the functional evaluation of ligands that are implicated in costimulatory processes. In this study we have evaluated the role of the CD2 family member CD150 (SLAM) and the TNF family member TL1A (TNFSF15) in the activation of human T cells. Whereas our results do not point to a significant role of CD150 in T cell activation we found TL1A to potently costimulate human T cells. Taken together our results demonstrate that T cell stimulator cells are excellent tools to study various aspects of costimulatory processes.


Subject(s)
Antigens, CD/immunology , Biological Assay/methods , Lymphocyte Activation/immunology , Receptors, Cell Surface/immunology , T-Lymphocytes/immunology , Tumor Necrosis Factor Ligand Superfamily Member 15/immunology , Animals , Antibodies/pharmacology , Cell Line, Tumor , Humans , Immunologic Factors/pharmacology , Lymphocyte Activation/drug effects , Major Histocompatibility Complex/immunology , Mice , Peptides/immunology , Receptor-CD3 Complex, Antigen, T-Cell/immunology , Signaling Lymphocytic Activation Molecule Family Member 1
16.
Cell Immunol ; 263(2): 188-95, 2010.
Article in English | MEDLINE | ID: mdl-20392439

ABSTRACT

Chemokine-like factor 1 (CKLF1) was the first member of the CKLF-like MARVEL transmembrane domain containing member (CMTM) family to be discovered. Its expression level is increased clearly in peripheral blood lymphocytes upon phytohemagglutinin stimulation, but little is known about the expression and function of CKLF1 in dendritic cells (DCs), which are the most potent antigen-presenting cells. In the present study, we showed that CKLF1 was highly expressed in monocytes. During differentiation from monocytes to immature DCs, CKLF1 was increased dramatically on day 2 and then decreased from day 3 to 5. Upon maturation with different stimuli, CKLF1 was down-regulated. Two peptides of CKLF1, C19 and C27, stimulated the effect of immature DCs (imDCs) on T-cell proliferation and IFN-gamma production. Further study on DC maturation showed that C19 and C27 up-regulated HLA-DR expression and IL-12 secretion, with no obvious effects on CD80, CD83 or CD86. Thus, CKLF1-C19 and -C27 can stimulate antigen-presenting capability of imDCs.


Subject(s)
Cell Differentiation , Chemokines/metabolism , Dendritic Cells/cytology , Dendritic Cells/immunology , Gene Expression Regulation , Cell Differentiation/drug effects , Chemokines/pharmacology , Dendritic Cells/drug effects , Down-Regulation , Humans , Interleukin-12/metabolism , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/immunology , MARVEL Domain-Containing Proteins
17.
Eur J Immunol ; 40(2): 321-9, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19950173

ABSTRACT

IL-35 is a heterodimer of EBV-induced gene 3 and of the p35 subunit of IL-12, and recently identified as an inhibitory cytokine produced by natural Treg in mice, but not in humans. Here we demonstrate that DC activated by human rhinoviruses (R-DC) induce IL-35 production and release, as well as a suppressor function in CD4(+) and CD8(+) T cells derived from human peripheral blood but not in naïve T cells from cord blood. The induction of IL-35-producing T cells by R-DC was FOXP3-independent, but blocking of B7-H1 (CD274) and sialoadhesin (CD169) on R-DC with mAb against both receptors prevented the induction of IL-35. Thus, the combinatorial signal delivered by R-DC to T cells via B7-H1 and sialoadhesin is crucial for the induction of human IL-35(+) Treg. These results demonstrate a novel pathway and its components for the induction of immune-inhibitory T cells.


Subject(s)
Antigens, CD/metabolism , Dendritic Cells/metabolism , Interleukins/metabolism , Membrane Glycoproteins/metabolism , Receptors, Immunologic/metabolism , Rhinovirus/physiology , T-Lymphocytes, Regulatory/metabolism , B7-H1 Antigen , Blotting, Western , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/metabolism , Cell Line , Cell Proliferation/drug effects , Cells, Cultured , Coculture Techniques , Culture Media, Conditioned/chemistry , Culture Media, Conditioned/pharmacology , Dendritic Cells/cytology , Flow Cytometry , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Host-Pathogen Interactions , Humans , Interferon-alpha/metabolism , Interleukin-10/metabolism , Interleukins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sialic Acid Binding Ig-like Lectin 1 , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/virology , Transforming Growth Factor beta/metabolism
18.
Blood ; 114(27): 5481-9, 2009 Dec 24.
Article in English | MEDLINE | ID: mdl-19864645

ABSTRACT

Dendritic cells (DCs) are the key cell type in the regulation of an adaptive immune response. Under inflammatory conditions monocytes can give rise to immunostimulatory DCs, depending on microenvironmental stimuli. Here we show that oxidized phospholipids (Ox-Pls), which are generated during inflammatory reactions, dysregulate the differentiation of DCs. DCs generated in the presence of Ox-Pls up-regulated the typical DC marker DC-SIGN but did not express CD1a, CD1b, and CD1c. These DCs generated in the presence of Ox-Pls had a substantially diminished T cell-stimulating capacity after stimulation with Toll-like receptor ligands. Toll-like receptor ligand-induced production of interleukin-12 also was strongly diminished, whereas induction of CD83 was not altered. In addition, we found that Ox-Pls strongly inhibit inflammatory stimuli-induced phosphorylation of histone H3, a key step of interleukin-12 production, yet leaving activation of nuclear factor-kappaB unaltered. Taken together, Ox-Pls present during differentiation yielded DCs with a reduced capacity to become immunostimulatory mature DCs. Furthermore, the presence of Ox-Pls blocked histone modifications required for full activation of DCs. Therefore, inflammation-derived Ox-Pls control DC functions in part by epigenetic mechanisms.


Subject(s)
Cell Differentiation/drug effects , Dendritic Cells/drug effects , Monocytes/drug effects , Phosphatidylcholines/pharmacology , Antigens, CD/genetics , Blotting, Western , Cell Line , Cells, Cultured , Dendritic Cells/cytology , Dendritic Cells/metabolism , Dose-Response Relationship, Drug , Epigenesis, Genetic , Flow Cytometry , Gene Expression Regulation/drug effects , Histones/metabolism , Humans , Immunoglobulins/genetics , Interleukin-12/metabolism , Lipopolysaccharides/pharmacology , Membrane Glycoproteins/genetics , Monocytes/cytology , Monocytes/metabolism , NF-kappa B/metabolism , Phosphorylation/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , CD83 Antigen
19.
J Immunol ; 183(7): 4440-8, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19734213

ABSTRACT

Dendritic cells (DCs) use pattern recognition receptors to sense invading viruses and triggering of these receptors induces a maturation program. Human rhinoviruses (HRVs) belong to the family of Picornaviridae, which have a single-stranded, coding RNA genome. Because HRV does not replicate in DCs, we used genomic RNA from HRV in this study to analyze the impact of natural occurring viral ssRNA on DC function. We found that transfection of human monocyte-derived DCs with viral ssRNA induced type I IFN production but failed to activate the NF-kappaB pathway in DCs. In line with this observation, the up-regulation of typical maturation markers such as CD83 or the production of the proinflammatory cytokines IL-12p40, IL-6, and TNF-alpha was not detectable. Most importantly, the T cell stimulatory capacity of viral ssRNA-treated DCs was not enhanced and remained at the level of immature DCs. Taken together, our results demonstrate that viral ssRNA efficiently activates the innate defense arm of DCs, whereas it is insufficient to activate the stimulatory capacity of DCs for the adaptive defense responses.


Subject(s)
Cell Differentiation/immunology , Dendritic Cells/immunology , Down-Regulation/immunology , Genome, Viral/immunology , Interferon Type I/biosynthesis , Monocytes/immunology , RNA, Viral/immunology , Rhinovirus/immunology , Biomarkers/metabolism , Cell Differentiation/genetics , Cell Line , Dendritic Cells/pathology , Dendritic Cells/virology , Down-Regulation/genetics , Growth Inhibitors/genetics , Growth Inhibitors/physiology , HeLa Cells , Humans , Immunity, Innate/genetics , Immunophenotyping , Inflammation Mediators/metabolism , Interferon Type I/genetics , Lymphocyte Activation/genetics , Monocytes/pathology , Monocytes/virology , RNA, Viral/antagonists & inhibitors , RNA, Viral/metabolism , Rhinovirus/genetics , Virus Replication/genetics , Virus Replication/immunology
20.
Blood ; 114(11): 2323-32, 2009 Sep 10.
Article in English | MEDLINE | ID: mdl-19617579

ABSTRACT

In hematopoietic stem cell transplant (HSCT) recipients, the recognition of polymorphic antigens by the donor-derived immune system is an important mechanism underlying both graft-versus-host disease and graft-versus-leukemia (GVL) effect. Here we show that a subset of HSCT recipients (13.9%, n = 108) have antibodies directed to surface molecules of dendritic cells. We have used one such serum in conjunction with retroviral expression cloning to identify the highly polymorphic surface molecule immunoglobulin-like transcript 5 (ILT5) as one of the targets of dendritic cell-reactive antibodies. ILT5 reactive antibodies were found in 5.4% of HSCT patients but not in solid organ transplantation recipients, patients with collagen diseases, multiparous women, or polytransfused or healthy persons. We show that ILT5-specific antibodies can mediate killing of ILT5-bearing cells and furthermore demonstrate ILT5 expression in some leukemic cells, indicating that it might be a target for GVL effects. Thus, our results represent the first description of potent allogeneic antibody responses to a non-major histocompatibility complex cell surface molecule in hematopoietic stem cell transplanted patients and warrant further studies to elucidate the role of antibodies to polymorphic cell surface molecules in GVL and graft-versus-host responses.


Subject(s)
Antibody Formation/immunology , Antigens, CD/immunology , Dendritic Cells/immunology , Hematopoietic Stem Cell Transplantation , Isoantibodies/immunology , Receptors, Immunologic/immunology , Animals , Collagen Diseases/immunology , Female , Follow-Up Studies , Graft vs Host Disease/immunology , Graft vs Leukemia Effect/immunology , Male , Mice , Parity/immunology , Pregnancy , Retrospective Studies , Transplantation, Homologous
SELECTION OF CITATIONS
SEARCH DETAIL
...