Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Proteomics ; 18(12): e1700465, 2018 06.
Article in English | MEDLINE | ID: mdl-29786170

ABSTRACT

The recent development in immune checkpoint inhibitors and chimeric antigen receptor (CAR) T-cells in the treatment of cancer has not only demonstrated the potency of utilizing T-cell reactivity for cancer therapy, but has also highlighted the need for developing new approaches to discover targets suitable for such novel therapeutics. Here we analyzed the immunopeptidomes of six HLA-A2-positive triple negative breast cancer (TNBC) samples by nano-ultra performance liquid chromatography tandem mass spectrometry (nUPLC-MS2 ). Immunopeptidomic profiling identified a total of 19 675 peptides from tumor and adjacent normal tissue and 130 of the peptides were found to have higher abundance in tumor than in normal tissues. To determine potential therapeutic target proteins, we calculated the average tumor-associated cohort coverage (aTaCC) that represents the percentage coverage of each protein in this cohort by peptides that had higher tumoral abundance. Cofilin-1 (CFL-1), interleukin-32 (IL-32), proliferating cell nuclear antigen (PCNA), syntenin-1 (SDCBP), and ribophorin-2 (RPN-2) were found to have the highest aTaCC scores. We propose that these antigens could be evaluated further for their potential as targets in breast cancer immunotherapy and the small cohort immunopeptidomics analysis technique could be used in a wide spectrum of target discovery. Data are available via ProteomeXchange with identifier PXD009738.


Subject(s)
Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism , HLA-A2 Antigen/metabolism , Immunotherapy , Peptide Fragments/metabolism , Triple Negative Breast Neoplasms/metabolism , Adult , Aged , Aged, 80 and over , Antigens, Neoplasm/analysis , Female , HLA-A2 Antigen/immunology , Humans , Middle Aged , Peptide Fragments/immunology , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/pathology
2.
PLoS One ; 10(6): e0129063, 2015.
Article in English | MEDLINE | ID: mdl-26098415

ABSTRACT

OBJECTIVE: The leukocyte immunoglobulin-like receptor (LILR) family includes inhibitory and stimulatory members which bind to classical and non-classical HLA-class I. The ligands for many LILR including LILRB5 have not yet been identified. METHODS: We generated C-terminal eGFP and N-terminal FLAG-tagged fusion constructs for monitoring LILR expression. We screened for LILR binding to HLA-class I by tetramer staining of 293T cells transfected with LILRA1, A4, A5 A6 and LILRB2 and LILRB5. We also studied HLA class I tetramer binding to LILRB5 on peripheral monocyte cells. LILRB5 binding to HLA-class I heavy chains was confirmed by co-immunoprecipitation. RESULTS: HLA-B27 (B27) free heavy chain (FHC) dimer but not other HLA-class I stained LILRB5-transfected 293T cells. B27 dimer binding to LILRB5 was blocked with the class I heavy chain antibody HC10 and anti-LILRB5 antisera. B27 dimers also bound to LILRB5 on peripheral monocytes. HLA-B7 and B27 heavy chains co-immunoprecipitated with LILRB5 in transduced B and rat basophil RBL cell lines. CONCLUSIONS: Our findings show that class I free heavy chains are ligands for LILRB5. The unique binding specificity of LILRB5 for HLA-class I heavy chains probably results from differences in the D1 and D2 immunoglobulin-like binding domains which are distinct from other LILR which bind to ß2m-associated HLA-class I.


Subject(s)
Antigens, CD/metabolism , HLA-B27 Antigen/metabolism , HLA-B7 Antigen/metabolism , Receptors, Immunologic/metabolism , Animals , Cell Line , Humans , Protein Binding , Rats
3.
J Leukoc Biol ; 96(1): 105-11, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24585933

ABSTRACT

The collagenous C-type lectin, SP-D, is a multitrimeric glycoprotein present at mucosal surfaces and is involved in host defense against infections in mammals. SP-D has immunomodulatory properties, but the underlying mechanisms are incompletely understood. SP-D contains collagen domains. LAIR-1 is an inhibitory immune receptor at the cell surface of various immune-competent cells that binds collagen. We hypothesized that the immunomodulatory functions of SP-D can be mediated via interactions between its collagen domain and LAIR-1. Binding assays show that SP-D interacts via its collagenous domain with LAIR-1 and the related LAIR-2. This does not affect the mannan-binding capacities of SP-D, which induces cross-linking of LAIR-1 in a cellular reporter assay. Functional assays show that SP-D inhibits the production of FcαR-mediated reactive oxygen via LAIR-1. Our studies indicate that SP-D is a functional ligand of the immune inhibitory receptor LAIR-1. Thus, we have identified a novel pathway for the immunomodulatory functions of SP-D mediated via binding of its collagenous domains to LAIR-1. This may provide a mechanism for the unexplained immunomodulatory function of the collagenous domains of SP-D.


Subject(s)
Pulmonary Surfactant-Associated Protein D/metabolism , Receptors, Immunologic/metabolism , Antigens, CD/genetics , Antigens, CD/immunology , Antigens, CD/metabolism , HL-60 Cells , Humans , Protein Binding , Protein Structure, Tertiary , Pulmonary Surfactant-Associated Protein D/genetics , Pulmonary Surfactant-Associated Protein D/immunology , Receptors, Fc/genetics , Receptors, Fc/immunology , Receptors, Fc/metabolism , Receptors, Immunologic/agonists , Receptors, Immunologic/genetics , Receptors, Immunologic/immunology
4.
Clin Immunol ; 150(1): 31-42, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24333531

ABSTRACT

Inhibitory receptors are thought to be important in balancing immune responses. The general assumption is that lack of inhibition predisposes for autoimmune diseases. As reviewed here, various experimental and clinical data support this assumption. However, in humans genetic evidence implicates only a limited number of inhibitory receptors. GWAS have established common variation in a few inhibitory receptor genes, such as FCγRIIB, PD-1 and CTLA-4 as risk factors. The question arises whether inhibitory receptor function is a major determinant of autoimmune disease. In this respect, the finding that genetic variation in CSK and PTPN22 is strongly associated with multiple autoimmune diseases is of interest. We propose a model in which the molecules encoded by these genes are downstream of inhibitory receptors. We conclude that common genetic variation of inhibitory receptors, with few exceptions, is not a determining factor for autoimmunity in humans. However, common downstream signaling pathways are.


Subject(s)
Autoimmune Diseases/immunology , Receptors, Immunologic/immunology , Animals , Autoimmunity/genetics , Autoimmunity/immunology , Humans
5.
J Innate Immun ; 6(3): 284-92, 2014.
Article in English | MEDLINE | ID: mdl-24192271

ABSTRACT

Activation of complement may cause severe tissue damage in antibody-mediated allograft rejection and other antibody-mediated clinical conditions; therefore, novel potent complement inhibitors are needed. Previously, we described binding of the inhibitory receptor LAIR-1 and its soluble family member LAIR-2 to collagen. Here, we investigated binding of LAIR-1 and LAIR-2 to the complement proteins C1q and MBL, which both have collagen-like domains, and evaluated the effect of this binding on complement function. We demonstrate specific binding of recombinant LAIR proteins to both C1q and MBL. Surface plasmon resonance experiments showed that LAIR-2-Fc protein bound C1q and MBL with the highest affinity compared to LAIR-2-HIS. We, therefore, hypothesized that LAIR-2-Fc is a potent complement inhibitor. Indeed, LAIR-2-Fc inhibited C4 fixation to IgG or mannan, reduced activation of C4 by aggregated IgG in plasma and inhibited iC3b deposition on cells. Finally, LAIR-2-Fc inhibited complement-mediated lysis of cells sensitized with anti-HLA antibodies in an ex vivo model for antibody-mediated transplant rejection. Thus, LAIR-2-Fc is an effective novel complement inhibitor for the treatment and prevention of antibody-mediated allograft rejection and antibody-mediated clinical conditions.


Subject(s)
Graft Rejection/prevention & control , Immune Complex Diseases/therapy , Immunotherapy , Receptors, Immunologic/metabolism , Recombinant Fusion Proteins/metabolism , Antibody-Dependent Cell Cytotoxicity/drug effects , Complement C1q/metabolism , Complement Pathway, Mannose-Binding Lectin/drug effects , Graft Rejection/immunology , HLA Antigens/immunology , Humans , Immune Complex Diseases/immunology , Immunoglobulin Constant Regions/genetics , Isoantibodies/metabolism , K562 Cells , Protein Binding/drug effects , Receptors, Immunologic/administration & dosage , Receptors, Immunologic/genetics , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics
6.
PLoS One ; 8(12): e83920, 2013.
Article in English | MEDLINE | ID: mdl-24386309

ABSTRACT

The soluble form of the inhibitory immune receptor leukocyte-Associated Ig-like Receptor-1 (sLAIR-1) is present in plasma, urine and synovial fluid and correlates to inflammation. We and others previously showed inflammatory protein expression in normal amniotic fluid at term. We hypothesized that sLAIR-1 is present in amniotic fluid during term parturition and is related to fetal lung function development. sLAIR-1 was detectable in all amniotic fluid samples (n=355) collected during term spontaneous deliveries. First, potential intra-uterine origins of amniotic fluid sLAIR-1 were explored. Although LAIR-1 was expressed on the surface of amniotic fluid neutrophils, LAIR-1 was not secreted upon ex vivo neutrophil stimulation with LPS, or PMA/ionomycin. Cord blood concentrations of sLAIR-1 were fourfold lower than and not related to amniotic fluid concentrations and placentas showed no or only sporadic LAIR-1 positive cells. Similarly, in post-mortem lung tissue of term neonates that died of non-pulmonary disorders LAIR-1 positive cells were absent or only sporadically present. In fetal urine samples, however, sLAIR-1 levels were even higher than in amniotic fluid and correlated with amniotic fluid sLAIR-1 concentrations. Second, the potential relevance of amniotic fluid sLAIR-1 was studied. sLAIR-1 concentrations had low correlation to amniotic fluid cytokines. We measured neonatal lung function in a convenient subset of 152 infants, using the single occlusion technique, at a median age of 34 days (IQR 30-39). The amniotic fluid concentration of sLAIR-1 was independently correlated to airway compliance (ρ=0.29, P=.001). Taken together, we show the consistent presence of sLAIR-1 in amniotic fluid, which originates from fetal urine. Concentrations of sLAIR-1 in amniotic fluid during term deliveries are independent from levels of other soluble immune mediators. The positive association between concentrations of amniotic fluid sLAIR-1 and neonatal lung compliance suggests that amniotic fluid sLAIR-1 may be useful as a novel independent marker of neonatal lung maturation.


Subject(s)
Amniotic Fluid/metabolism , Fetus/physiology , Receptors, Immunologic/chemistry , Receptors, Immunologic/metabolism , Amniotic Fluid/immunology , Biomarkers/chemistry , Biomarkers/metabolism , Female , Fetus/embryology , Fetus/metabolism , Gene Expression Regulation , Humans , Infant , Infant, Newborn , Lung/embryology , Lung Compliance , Neutrophils/metabolism , Pregnancy , Solubility , Term Birth
7.
Arthritis Rheum ; 63(12): 3749-57, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22127695

ABSTRACT

OBJECTIVE: Human leukocyte-associated immunoglobulin-like receptor 1 (hLAIR-1) is an immune inhibitory receptor for collagen that is expressed on most immune cells. We previously showed that the LAIR-1-collagen interaction could be antagonized by the secreted homolog hLAIR-2, which can be detected in the synovial fluid of rheumatoid arthritis (RA) patients. In addition, the extracellular part of hLAIR-1 is a putative antagonist upon shedding from the cell membrane. The purpose of this study was to determine the relative roles of hLAIR-2 and soluble hLAIR-1 (shLAIR-1) in the regulation of the LAIR-1-collagen interaction. METHODS: The ability of recombinant LAIR proteins to abrogate LAIR-1-collagen binding was tested by flow cytometry and adhesion assays. Collagen binding capacity was analyzed by surface plasmon resonance. Plasma, urine, and synovial fluid were screened for the presence of sLAIR-1 and LAIR-2 by enzyme-linked immunosorbent assay. RESULTS: Recombinant LAIR-2 proteins abrogated the binding of collagen to LAIR-1 more efficiently than did recombinant sLAIR-1. Consistent with these findings, surface plasmon resonance analysis showed that LAIR-2 had a higher affinity for collagen than did LAIR-1. Activated CD4+ T cells were the main producers of LAIR-2, whereas the source of sLAIR-1 remains elusive. Both soluble LAIR-1 and LAIR-2 could be detected in the plasma and urine of healthy control subjects and patients with RA. Urinary levels of both proteins were significantly increased in RA patients, and LAIR-2 levels in urine were significantly correlated with markers of inflammation. CONCLUSION: Our data suggest that LAIR-2 is a more potent antagonist of LAIR-1 function in vivo, while both sLAIR-1 and LAIR-2 are potential biomarkers that may be used to monitor urine samples for evidence of systemic inflammation.


Subject(s)
Arthritis, Rheumatoid/metabolism , Collagen/metabolism , Receptors, Immunologic/metabolism , Biomarkers/metabolism , Case-Control Studies , Cell Line , Cohort Studies , Collagen/antagonists & inhibitors , Female , Humans , Male , Osteoarthritis/metabolism , Protein Binding/physiology , Receptors, Immunologic/antagonists & inhibitors , Synovial Fluid/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...