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










Publication year range
1.
Emerg Microbes Infect ; 13(1): 2361019, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38804979

ABSTRACT

HLA-E expression plays a central role for modulation of NK cell function by interaction with inhibitory NKG2A and stimulatory NKG2C receptors on canonical and adaptive NK cells, respectively. Here, we demonstrate that infection of human primary lung tissue with SARS-CoV-2 leads to increased HLA-E expression and show that processing of the peptide YLQPRTFLL from the spike protein is primarily responsible for the strong, dose-dependent increase of HLA-E. Targeting the peptide site within the spike protein revealed that a single point mutation was sufficient to abrogate the increase in HLA-E expression. Spike-mediated induction of HLA-E differentially affected NK cell function: whereas degranulation, IFN-γ production, and target cell cytotoxicity were enhanced in NKG2C+ adaptive NK cells, effector functions were inhibited in NKG2A+ canonical NK cells. Analysis of a cohort of COVID-19 patients in the acute phase of infection revealed that adaptive NK cells were induced irrespective of the HCMV status, challenging the paradigm that adaptive NK cells are only generated during HCMV infection. During the first week of hospitalization, patients exhibited a selective increase of early NKG2C+CD57- adaptive NK cells whereas mature NKG2C+CD57+ cells remained unchanged. Further analysis of recovered patients suggested that the adaptive NK cell response is primarily driven by a wave of early adaptive NK cells during acute infection that wanes once the infection is cleared. Together, this study suggests that NK cell responses to SARS-CoV-2 infection are majorly influenced by the balance between canonical and adaptive NK cells via the HLA-E/NKG2A/C axis.


Subject(s)
COVID-19 , HLA-E Antigens , Histocompatibility Antigens Class I , Killer Cells, Natural , NK Cell Lectin-Like Receptor Subfamily C , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Humans , Killer Cells, Natural/immunology , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/genetics , COVID-19/immunology , COVID-19/virology , SARS-CoV-2/immunology , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/metabolism , NK Cell Lectin-Like Receptor Subfamily C/genetics , NK Cell Lectin-Like Receptor Subfamily C/metabolism , NK Cell Lectin-Like Receptor Subfamily C/immunology , Adaptive Immunity , Male , Female , Middle Aged , Lung/immunology , Lung/virology
2.
Cells ; 12(5)2023 02 21.
Article in English | MEDLINE | ID: mdl-36899812

ABSTRACT

Measure of drug-mediated immune reactions that are dependent on the patient's genotype determine individual medication protocols. Despite extensive clinical trials prior to the license of a specific drug, certain patient-specific immune reactions cannot be reliably predicted. The need for acknowledgement of the actual proteomic state for selected individuals under drug administration becomes obvious. The well-established association between certain HLA molecules and drugs or their metabolites has been analyzed in recent years, yet the polymorphic nature of HLA makes a broad prediction unfeasible. Dependent on the patient's genotype, carbamazepine (CBZ) hypersensitivities can cause diverse disease symptoms as maculopapular exanthema, drug reaction with eosinophilia and systemic symptoms or the more severe diseases Stevens-Johnson-Syndrome or toxic epidermal necrolysis. Not only the association between HLA-B*15:02 or HLA-A*31:01 but also between HLA-B*57:01 and CBZ administration could be demonstrated. This study aimed to illuminate the mechanism of HLA-B*57:01-mediated CBZ hypersensitivity by full proteome analysis. The main CBZ metabolite EPX introduced drastic proteomic alterations as the induction of inflammatory processes through the upstream kinase ERBB2 and the upregulation of NFκB and JAK/STAT pathway implying a pro-apoptotic, pro-necrotic shift in the cellular response. Anti-inflammatory pathways and associated effector proteins were downregulated. This disequilibrium of pro- and anti-inflammatory processes clearly explain fatal immune reactions following CBZ administration.


Subject(s)
Drug Hypersensitivity , Stevens-Johnson Syndrome , Humans , Janus Kinases , Anticonvulsants/therapeutic use , Up-Regulation , Proteomics , STAT Transcription Factors/genetics , Signal Transduction , Carbamazepine , HLA-B Antigens/genetics , Stevens-Johnson Syndrome/etiology , Stevens-Johnson Syndrome/genetics , NF-kappa B/genetics
3.
Biomedicines ; 10(3)2022 Mar 17.
Article in English | MEDLINE | ID: mdl-35327495

ABSTRACT

Type B adverse drug reactions (ADRs) represent a significant threat as their occurrence arises unpredictable and despite proper application of the drug. The severe immune reaction Abacavir Hypersensitivity Syndrome (AHS) that arises in HIV+ patients treated with the antiretroviral drug Abacavir (ABC) strongly correlates to the presence of the human leukocyte antigen (HLA) genotype HLA-B*57:01 and discriminates HLA-B*57:01+ HIV+ patients from ABC treatment. However, not all HLA-B*57:01+ HIV+ patients are affected by AHS, implying the involvement of further patient-specific factors in the development of AHS. The establishment of a reliable assay to classify HLA-B*57:01 carriers as ABC sensitive or ABC tolerant allowed to investigate the T cell receptor (TCR) Vß chain repertoire of effector cells and revealed Vß6 and Vß24 as potential public TCRs in ABC sensitive HLA-B*57:01 carriers. Furthermore, distinct effects of ABC on the cellular proteome of ABC sensitive and tolerant volunteers were observed and suggest enhanced activation and maturation of dentritic cells (DC) in ABC sensitive volunteers. Analysis of ABC-naïve cellular proteomes identified the T cell immune regulator 1 (TCIRG1) as a potential prognostic biomarker for ABC susceptibility and the involvement of significantly upregulated proteins, particularly in peptide processing, antigen presentation, interferon (IFN), and cytokine regulation.

4.
J Pers Med ; 12(1)2022 Jan 04.
Article in English | MEDLINE | ID: mdl-35055355

ABSTRACT

Type B adverse drug reactions (ADRs) are unpredictable based on the drug's pharmacology and represent a key challenge in pharmacovigilance. For human leukocyte antigen (HLA)-mediated type B ADRs, it is assumed that the protein/small-molecule interaction alters the biophysical and mechanistic properties of the antigen presenting cells. Sophisticated methods enabled the molecular appreciation of HLA-mediated ADRs; in several instances, the drug molecule occupies part of the HLA peptide binding groove and modifies the recruited peptide repertoire thereby causing a strong T-cell-mediated immune response that is resolved upon withdrawal of medication. The severe ADR in HLA-B*57:01+ patients treated with the antiretroviral drug abacavir (ABC) in anti-HIV therapy is an example of HLA-drug-T cell cooperation. However, the long-term damages of the HLA-B*57:01-expressing immune cells following ABC treatment remain unexplained. Utilizing full proteome sequencing following ABC treatment of HLA-B*57:01+ cells, we demonstrate stringent proteomic alteration of the HLA/drug presenting cells. The proteomic content indisputably reflects the cellular condition; this knowledge directs towards individual pharmacovigilance for the development of personalized and safe medication.

5.
Front Immunol ; 11: 2046, 2020.
Article in English | MEDLINE | ID: mdl-32973812

ABSTRACT

Tumor immune escape is associated with both, the expression of immune checkpoint molecules on peripheral immune cells and soluble forms of the human leukocyte antigen-G (HLA-G) in the blood, which are consequently discussed as clinical biomarker for disease status and outcome of cancer patients. HLA-G preferentially interacts with the inhibitory receptor immunoglobulin-like transcript (ILT) receptor-2 in the blood and can be secreted as free soluble molecules (sHLA-G) or via extracellular vesicles (EV). To investigate the contribution of these two forms to the expression of checkpoint molecules in peripheral blood, we primed peripheral blood mononuclear cells with purified soluble sHLA-G1 protein, or EV preparations derived from SUM149 cells transfected with membrane-bound HLA-G1 or control vector prior to anti-CD3/CD28 T cell activation. Our study demonstrated that priming of PBMC with sHLA-G1 protein prior to 48 h activation resulted in enhanced frequencies of ILT-2 expressing CD8+ T cells, and in an upregulation of immune checkpoint molecules CTLA-4, PD-1, TIM-3, and CD95 exclusively on ILT-2 positive CD8+ T cells. In contrast, when PBMC were primed with EV (containing HLA-G1 or not) upregulation of CTLA-4, PD-1, TIM-3, and CD95 occurred exclusively on ILT-2 negative CD8+ T cells. Taken together, our data suggest that priming with sHLA-G forms induces a pronounced immunosuppressive/exhausted phenotype and that priming with sHLA-G1 protein or EV derived from HLA-G1 positive or negative SUM149 cells affects CD8+ T cells complementary by targeting either the ILT-2 positive or negative subpopulation, respectively, after T cell activation.


Subject(s)
Antigens, CD/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Extracellular Vesicles/metabolism , HLA-G Antigens/immunology , HLA-G Antigens/metabolism , Leukocyte Immunoglobulin-like Receptor B1/metabolism , Antigens, CD/genetics , Biological Transport , Biomarkers , Cell Line , Culture Media, Conditioned/metabolism , Gene Expression , HLA-G Antigens/blood , Humans , Immune Checkpoint Proteins/metabolism , Immunomodulation , Leukocyte Immunoglobulin-like Receptor B1/genetics , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Phenotype
6.
Cancers (Basel) ; 11(8)2019 Aug 02.
Article in English | MEDLINE | ID: mdl-31382533

ABSTRACT

Extracellular vesicles (EV) and their tumor-supporting cargos provide a promising translational potential in liquid biopsies for risk assessment of epithelial ovarian cancer (EOC) patients frequently relapsing, despite initial complete therapy responses. As the immune checkpoint molecule HLA-G, which is operative in immune-escape, can be released by EV, we evaluate the abundance of EV and its vesicular-bound amount of HLA-G (HLA-GEV) as a biomarker in EOC. After enrichment of EV from plasma samples, we determined the EV particle number and amount of HLA-GEV by nanoparticle tracking analysis or ELISA. The association of results with the clinical status/outcome revealed that both, EV particle number and HLA-GEV were significantly elevated in EOC patients, compared to healthy females. However, elevated levels of HLA-GEV, but not EV numbers, were exclusively associated with a disadvantageous clinical status/outcome, including residual tumor, presence of circulating tumor cells, and disease progression. High HLA-GEV status was an independent predictor of progression, besides residual tumor burden and platinum-sensitivity. Especially among patients without residual tumor burden or with platinum-sensitivity, HLA-GEV identified patients with high risk of progression. Thus, this study highlights HLA-GEV as a potential novel biomarker for risk assessment of EOC patients with a rather beneficial prognosis defined by platinum-sensitivity or lack of residual tumor burden.

7.
Immunogenetics ; 71(7): 455-463, 2019 07.
Article in English | MEDLINE | ID: mdl-31250049

ABSTRACT

The main expression sites of HLA-G are human extravillous trophoblast cells. The interaction of HLA-G with uterine NK cells promotes their maturation and differentiation into decidual NK (dNK) cells. dNK cells secrete chemokines, cytokines, and proangiogenic factors in favor of a vascular remodeling and an immune suppressive microenvironment of the decidua. HLA-G is the most polymorphic member of the oligomorphic non-classical HLA molecule family; yet, the impact of polymorphic differences is not comprehensively understood. sHLA-G levels in embryo culture medium correlate with successful pregnancy; however, it remains questionable if HLA-G allelic diversity impacts on the outcome of dNK cell development. We utilized synthetic sHLA-G*01:01, 01:03, and 01:04 molecules and transduced K652/mHLA-G*01:01, 01:03, and 01:04 cells to study the biological interaction between HLA-G alleles and primary NK cells of human term placenta. Despite its low frequency, HLA-G*01:04 and not the most prevalent allele HLA-G*01:01 appear to be strong catalysts of dNK cell proliferation. Concluding, this study illustrates novel insights into the impact and binding efficiency of the three most common variants of HLA-G on primary placental NK cells.


Subject(s)
HLA-G Antigens/genetics , Killer Cells, Natural/metabolism , Placenta/cytology , CD56 Antigen/metabolism , Cell Proliferation , Decidua/cytology , Female , HLA-G Antigens/immunology , HLA-G Antigens/metabolism , Humans , K562 Cells , Killer Cells, Natural/immunology , Pregnancy
8.
Immunogenetics ; 71(5-6): 353-360, 2019 05.
Article in English | MEDLINE | ID: mdl-30941482

ABSTRACT

HLA-F belongs to the non-classical HLA-Ib molecules with a marginal polymorphic nature and tissue-restricted distribution. HLA-F is a ligand of the NK cell receptor KIR3DS1, whose activation initiates an antiviral downstream immune response and lead to delayed disease progression of HIV-1. During the time course of HIV infection, the expression of HLA-F is upregulated while its interaction with KIR3DS1 is diminished. Understanding HLA-F peptide selection and presentation is essential to a comprehensive understanding of this dynamic immune response and the molecules function. In this study, we were able to recover stable pHLA-F*01:01 complexes and analyze the characteristics of peptides naturally presented by HLA-F. These HLA-F-restricted peptides exhibit a non-canonical length without a defined N-terminal anchor. The peptide characteristics lead to a unique presentation profile and influence the stability of the protein. Furthermore, we demonstrate that almost all source proteins of HLA-F-restricted peptides are described to interact with HIV proteins. Understanding the balance switch between HLA-Ia and HLA-F expression and peptide selection will support to understand the role of HLA-F in viral pathogenesis.

9.
Immunogenetics ; 71(5-6): 361, 2019 May.
Article in English | MEDLINE | ID: mdl-31020357

ABSTRACT

The original version of this article contained errors. The Article Title, Figures 1 and 3, and Electronic Supplementary Materials were incorrectly shown in the wrong version. The original article has been corrected.

10.
J Immunol Res ; 2018: 5086503, 2018.
Article in English | MEDLINE | ID: mdl-30302345

ABSTRACT

Among patients treated with the anticonvulsive and psychotropic drug carbamazepine (CBZ), approximately 10% develop severe and life-threatening adverse drug reactions. These immunological conditions are resolved upon withdrawal of the medicament, suggesting that the drug does not manifest in the body in long term. The HLA allele B∗15:02 has been described to be a genomic biomarker for CBZ-mediated immune reactions. It is not well understood if the immune reactions are triggered by the original drug or by its metabolite carbamazepine-10,11-epoxide (EPX) and how the interaction between the drug and the distinct HLA molecule occurs. Genetically engineered human B-lymphoblastoid cells expressing soluble HLA-B∗15:02 molecules were treated with the drug or its metabolite. Functional pHLA complexes were purified; peptides were eluted and sequenced. Applying mass spectrometric analysis, CBZ and EPX were monitored by analyzing the heavy chain and peptide fractions separately for the presence of the drug. This method enabled the detection of the drug in a biological situation post-pHLA assembly. Both drugs were bound to the HLA-B∗15:02 heavy chain; however, solely EPX altered the peptide-binding motif of B∗15:02-restricted peptides. This observation could be explained through structural insight; EPX binds to the peptide-binding region and alters the biochemical features of the F pocket and thus the peptide motif. Understanding the nature of immunogenic interactions between CBZ and EPX with the HLA immune complex will guide towards effective and safe medications.


Subject(s)
Allergens/adverse effects , Anticonvulsants/adverse effects , B-Lymphocytes/drug effects , Carbamazepine/analogs & derivatives , Carbamazepine/adverse effects , Drug Hypersensitivity/immunology , Drug-Related Side Effects and Adverse Reactions/immunology , Allergens/chemistry , Allergens/therapeutic use , Anticonvulsants/chemistry , Anticonvulsants/therapeutic use , Antigen Presentation , B-Lymphocytes/physiology , Binding Sites , Carbamazepine/chemistry , Carbamazepine/pharmacology , Carbamazepine/therapeutic use , Cell Line , HLA-B15 Antigen/genetics , HLA-B15 Antigen/metabolism , Humans , Immunomodulation , Mass Spectrometry , Peptide Fragments/metabolism , Protein Binding
11.
Immunogenetics ; 70(10): 639-646, 2018 11.
Article in English | MEDLINE | ID: mdl-30128813

ABSTRACT

Peptide selection in infected cells is not fully understood yet, but several indications point to the fact that there are differences to uninfected cells, especially in productive HCMV infection, since HCMV evolved various strategies to disable the hosts immune system, including presentation of peptide-HLA complexes to immune effector cells. Therefore, peptide predictions for specific HLA alleles are limited in these cases and the naturally presented peptide repertoire of HCMV-infected cells is of major interest to optimize adoptive T cell therapies. The allotypes HLA-B*35:01 and B*35:08 differ at a single amino acid at position 156 and have been described to differ in their peptide features and in their association with the peptide loading complex. Virus specific T cells recognizing the allelic pHLA-B*35 complexes could be detected, indicating a significant role of this HLA subtypes in viral immunity. However, naturally selected and presented viral peptides have not been described so far. In this study, we analyzed the peptide binding repertoire for HLA-B*35:01 and HLA-B*35:08 in HCMV-infected cells. The isolated peptides from both allelic subtypes were of extraordinary length, however differed in their features, origin, and sequence. For these HCMV-originated peptides, no overlap in the peptide repertoire could be observed between the two allelic subtypes. These findings reveal the discrepancies between predicted and naturally presented immunogenic epitopes and support the need of comprehensive peptide recruitment data for personalized and effective cellular therapies.


Subject(s)
Cytomegalovirus Infections/immunology , Cytomegalovirus/immunology , Epitopes/immunology , HLA-B Antigens/genetics , Alleles , Amino Acid Sequence/genetics , CD8-Positive T-Lymphocytes/immunology , Cytomegalovirus/pathogenicity , Cytomegalovirus Infections/genetics , Cytomegalovirus Infections/virology , Epitopes/genetics , HLA-B Antigens/immunology , Humans , Peptides/genetics , Peptides/immunology
12.
Immunogenetics ; 70(8): 485-494, 2018 08.
Article in English | MEDLINE | ID: mdl-29602958

ABSTRACT

HLA-G is known for its strictly restricted tissue distribution. HLA-G expression could be detected in immune privileged organs and many tumor entities such as leukemia, multiple myeloma, and non-Hodgkin and Hodgkin's lymphoma. This functional variability from mediation of immune tolerance to facilitation of tumor immune evasion strategies might translate to a differential NK cell inhibition between immune-privileged organs and tumor cells. The biophysical invariability of the HLA-G heavy chain and its contrary diversity in immunity implicates a strong influence of the bound peptides on the pHLA-G structure. The aim was to determine if HLA-G displays a tissue-specific peptide repertoire. Therefore, using soluble sHLA-G technology, we analyzed the K562 and HDLM-2 peptide repertoires. Although both cell lines possess a comparable proteome and recruit HLA-G-restricted peptides through the same peptide-loading pathway, the peptide features appear to be cell specific. HDLM-2 derived HLA-G peptides are anchored by an Arg at p1 and K562-derived peptides are anchored by a Lys. At p2, no anchor motif could be determined while peptides were anchored at pΩ with a Leu and showed an auxiliary anchor motif Pro at p3. To appreciate if the peptide anchor alterations are due to a cell-specific differential peptidome, we performed analysis of peptide availability within the different cell types. Yet, the comparison of the cell-specific proteome and HLA-G-restricted ligandome clearly demonstrates a tissue-specific peptide selection by HLA-G molecules. This exclusive and unexpected observation suggests an exquisite immune function of HLA-G.


Subject(s)
HLA-G Antigens/metabolism , HLA-G Antigens/physiology , Alleles , Amino Acid Sequence/genetics , Cell Line, Tumor , HLA-G Antigens/genetics , Humans , Killer Cells, Natural/immunology , Organ Specificity/physiology , Peptides/chemistry , Polymorphism, Genetic/genetics
13.
Hum Immunol ; 79(6): 453-462, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29605689

ABSTRACT

The trade-off from HLA class I expression to HLA-G expression support the immune evasion of malignant cells. The essential role of the virtually invariant HLA-G in immune tolerance, tumor immunology and its expression frequency in immune privileged tissues is known; however the specific importance of allelic subtypes in immune responses is still not well understood. HLA-G∗01:01, ∗01:03 and ∗01:04 are the most prevalent allelic variants differing at residues 31 and 110, respectively. In cytotoxicity assays applying K562 cells transduced with the HLA-G variants as targets and NK cells as effectors the differential protective potential of HLA-G variants was analyzed. Their peptide profiles were determined utilizing soluble HLA technology. An increased protective potential of HLA-G∗01:04 could be observed. All variants exhibit a unique peptide repertoire with marginal overlap, while G∗01:04 differs in its peptide anchor profile substantially. The functional differences between HLA-G subtypes could be explained by the constraint of the bound peptides, modifying the pHLA-G accessible surface. For the first time a contribution of amino acid alterations within the HLA-G heavy chain for peptide selection and NK cell recognition could be observed. These results will be a step towards understanding immune tolerance and will guide towards personalized immune therapeutic strategies.


Subject(s)
HLA-G Antigens/metabolism , Killer Cells, Natural/immunology , Peptides/metabolism , Alleles , Amino Acids/genetics , Cytotoxicity, Immunologic , Genotype , HLA-G Antigens/genetics , Humans , Immune Tolerance , Immunomodulation , K562 Cells , Lymphocyte Activation , Mutation/genetics , Peptides/genetics , Polymorphism, Genetic , Protein Domains/genetics
14.
Immunogenetics ; 68(1): 29-41, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26552660

ABSTRACT

Human leukocyte antigen (HLA)-E molecules are potent inhibitors of NK cell-mediated killing. Low in polymorphisms, two alleles are widely expressed among diverse populations: HLA-E*01:01 and HLA-E*01:03. Both alleles are distinguished by one SNP resulting in the substitution Arg107Gly. Both alleles present a limited set of peptides derived from class I leader sequences physiologically; however, HLA-E*01:01 presents non-canonical peptides in the absence of HLA class I molecules. To further assess the functional differences between both alleles, we analyzed the peptide repertoire of HLA-E*01:03 by applying soluble HLA technology followed by mass-spectrometric peptide sequencing. HLA-E*01:03 restricted peptides showed a length of 9-17 amino acids and differed in their biophysical properties, no overlap in the peptide repertoire of both allelic variants could be observed; however, both alleles shared marginal peptides from the same proteomic content. Artificial APCs expressing empty HLA-E*01:01 or E*01:03 molecules were generated and stabilized using cognate HLA class I-derived peptide ligands to analyze the impact of residue 107 within the HLA-E heavy chain on the NKG2/CD94 receptor engagement. Differences in peptide stabilization could be translated to the density and half-life time of peptide-HLA-E molecules on the cell surface that subsequently impacted NK cell inhibition as verified by cytotoxicity assays. Taken together, these data illustrate functional differences of HLA-E allelic variants induced by a single amino acid. Furthermore, the function of HLA-E in pathophysiologic situations when the HLA processing machinery is interrupted seems to be more emphasized than previously described, implying a crucial role for HLA-E in tumor or viral immune episodes.


Subject(s)
Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Amino Acid Sequence , Arginine/genetics , Cell Line , Cytotoxicity Tests, Immunologic , Genes, MHC Class I , Glycine/genetics , Histocompatibility Antigens Class I/metabolism , Humans , Killer Cells, Natural/immunology , Lymphocytes/immunology , Membrane Transport Proteins/metabolism , Molecular Sequence Data , Peptides/metabolism , Polymorphism, Single Nucleotide , Protein Isoforms , Protein Sorting Signals/physiology , HLA-E Antigens
15.
Stem Cells Int ; 2015: 346714, 2015.
Article in English | MEDLINE | ID: mdl-26366178

ABSTRACT

The HLA-E locus encodes a nonclassical class Ib molecule that serves many immune functions from inhibiting NK cells to activating CTLs. Structural analysis of HLA-E/NKG2A complexes visualized fine-tuning of protective immune responses through AA interactions between HLA-E, the bound peptide, and NKG2A/CD94. A loss of cellular protection through abrogation of the HLA-E/NKG2A engagement is dependent on the HLA-E bound peptide. The role of HLA-E in posttransplant outcomes is not well understood but might be attributed to its peptide repertoire. To investigate the self-peptide repertoire of HLA-E (∗) 01:01 in the absence of protective HLA class I signal peptides, we utilized soluble HLA technology in class I negative LCL cells in order to characterize HLA-E (∗) 01:01-bound ligands by mass-spectrometry. To understand the immunological impact of these analyzed ligands on NK cell reactivity, we performed cellular assays. Synthesized peptides were loaded onto recombinant T2 cells expressing HLA-E (∗) 01:01 molecules and applied in cytotoxicity assays using the leukemia derived NK cell line (NKL) as effector. HLA-E in complex with the self-peptides demonstrated a shift towards cytotoxicity and a loss of cell protection. Our data highlights the fact that the HLA-E-peptidome is not as restricted as previously thought and support the suggestion of a posttransplant role for HLA-E.

16.
Biochim Biophys Acta ; 1840(3): 1004-13, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24211252

ABSTRACT

BACKGROUND: CD7 expression is found on ~30% of acute myeloblastic leukemias (AML). The leukemic progenitor cell line KG1a (CD7+) constitutively expresses GM-CSF while the parental KG1 (CD7-) cell line does not. This study focuses on the molecular basis of CD7 mediated GM-CSF regulation. METHODS: KG1a cells were treated with recombinant SECTM1-Fc protein, the PI3K kinase inhibitors wortmannin, LY292004, or PI4K activator spermine. Stable KG1-CD7+, KG1a-shCD7, KG1a-shETS1 as well as KG1a-GFP, KG1a-PKCßII-GFP cell lines were generated and the levels of CD7, GM-CSF and ETS-1 mRNA and protein were compared by real-time-PCR, western blotting, flow cytometry and ELISA. RESULTS: SECTM1 is expressed in Human Bone Marrow Endothelial Cells (HBMEC) and its expression can be upregulated by both IFN-γ. KG1a cells demonstrated high expression levels of CD7 and ETS-1 allowing a constitutative signaling through the PI3K/Atk pathway to promote GM-CSF expression, while KG1 cells with low expression of CD7 and ETS-1 showed low GM-CSF expression. On KG1a cells GM-CSF expression could be negatively regulated by PI3K inhibitors or by recombinant SECTM1-Fc. Overexpression of CD7 in KG1 cells was insufficient to promote GM-CSF expression, while silencing of CD7 or ETS-1 resulted in reduced GM-CSF expression levels. Differentiation capable KG1a cells overexpressing PKCßII illustrated complete loss of CD7, but maintained normal levels of both ETS-1 and GM-CSF expression. CONCLUSION: These findings add an additional layer to the previously described autocrine/paracrine signaling between leukemic progenitor cells and the bone marrow microenvironment and highlight a role for SECTM1 in both normal and malignant hematopoiesis. GENERAL SIGNIFICANCE: This work shows that SECTM1 secreted from bone marrow stromal cells may interact with CD7 to influence GM-CSF expression in leukemic cells.


Subject(s)
Antigens, CD7/physiology , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Leukemia, Myeloid, Acute/metabolism , Membrane Proteins/physiology , Neoplastic Stem Cells/metabolism , Proto-Oncogene Protein c-ets-1/physiology , Cell Line, Tumor , Humans , Phosphatidylinositol 3-Kinases/physiology , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Transcription, Genetic
17.
Biochim Biophys Acta ; 1810(12): 1294-301, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21749909

ABSTRACT

BACKGROUND: SECTM1 is a T/NK cell "co-stimulatory" molecule that is expressed in the peripheral blood by neutrophils and monocytes. METHODS: We used qRT-PCR to investigate the mRNA expression of SECTM1 in human monocytic cells after stimulation with interferons and LPS and confirmed the protein expression by flow cytometry. RESULTS: The kinetics of interferon induced SECTM1 mRNA expression in MM6 cells are time dependent occurring rapidly within 3h of stimulation and reaching a maximal level at ~6h for IFN-α and ~12h for IFN-ß and IFN-γ. Co-treatment of MM6 cells with IFN-γ and cycloheximide caused a superinduction of SECTM1 mRNA expression while cycloheximide alone had no effect, illustrating that de novo protein synthesis is not required for IFN-γ enhanced expression of SECTM1 mRNA, a characteristic of IFN early response genes. The kinetics of IFN induced SECTM1 mRNA expression in primary monocytes is comparable although it occurs much quicker with rapid induction by IFN-α, IFN-ß and IFN-γ and maximal levels reached in <6h. Human monocytic cells also displayed a pronounced negative regulation of SECTM1 mRNA expression by LPS, while at the protein level SECTM1 expression was also shown to be regulated by IFN and LPS. Bioinformatic analysis of the SECTM1 promoter region identified STAT1α/GAS, STAT3, ISRE, NFκB and putative p63 binding sites suggesting a complex transcriptional control. This tight regulation of SECTM1 gene expression and rapid upregulation highlights its relevance in the innate immune response. CONCLUSION: Human monocytes produce SECTM1 in response to interferon stimuli that is negatively regulated by LPS. GENERAL SIGNIFICANCE: The level of SECTM1 expression is likely to be a key factor in innate immune responses and in the immune tolerance of cancerous cells.


Subject(s)
Gene Expression Regulation/drug effects , Killer Cells, Natural/immunology , Lipopolysaccharides/pharmacology , Membrane Proteins/physiology , Monocytes/drug effects , T-Lymphocytes/immunology , Amino Acid Sequence , Base Sequence , Cell Line , Flow Cytometry , Humans , Membrane Proteins/genetics , Molecular Sequence Data , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction
18.
Haematologica ; 96(1): 110-8, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20934997

ABSTRACT

BACKGROUND: Polymorphic differences between human leukocyte antigen (HLA) molecules affect the specificity and conformation of their bound peptides and lead to differential selection of the T-cell repertoire. Mismatching during allogeneic transplantation can, therefore, lead to immunological reactions. DESIGN AND METHODS: We investigated the structure-function relationships of six members of the HLA-B*41 allelic group that differ by six polymorphic amino acids, including positions 80, 95, 97 and 114 within the antigen-binding cleft. Peptide-binding motifs for B*41:01, *41:02, *41:03, *41:04, *41:05 and *41:06 were determined by sequencing self-peptides from recombinant B*41 molecules by electrospray ionization tandem mass spectrometry. The crystal structures of HLA-B*41:03 bound to a natural 16-mer self-ligand (AEMYGSVTEHPSPSPL) and HLA-B*41:04 bound to a natural 11-mer self-ligand (HEEAVSVDRVL) were solved. RESULTS: Peptide analysis revealed that all B*41 alleles have an identical anchor motif at peptide position 2 (glutamic acid), but differ in their choice of C-terminal pΩ anchor (proline, valine, leucine). Additionally, B*41:04 displayed a greater preference for long peptides (>10 residues) when compared to the other B*41 allomorphs, while the longest peptide to be eluted from the allelic group (a 16mer) was obtained from B*41:03. The crystal structures of HLA-B*41:03 and HLA-B*41:04 revealed that both alleles interact in a highly conserved manner with the terminal regions of their respective ligands, while micropolymorphism-induced changes in the steric and electrostatic properties of the antigen-binding cleft account for differences in peptide repertoire and auxiliary anchoring. CONCLUSIONS: Differences in peptide repertoire, and peptide length specificity reflect the significant functional evolution of these closely related allotypes and signal their importance in allogeneic transplantation, especially B*41:03 and B*41:04, which accommodate longer peptides, creating structurally distinct peptide-HLA complexes.


Subject(s)
Epitopes, T-Lymphocyte/genetics , HLA-B Antigens/genetics , Peptide Fragments/immunology , Polymorphism, Single Nucleotide/genetics , Recombinant Proteins/genetics , HLA-B Antigens/chemistry , HLA-B Antigens/metabolism , Humans , Peptide Fragments/metabolism , Protein Binding , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Spectrometry, Mass, Electrospray Ionization , T-Lymphocytes, Cytotoxic/immunology
19.
Haematologica ; 95(8): 1373-80, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20220067

ABSTRACT

BACKGROUND: The functional integrity of human leukocyte antigen low expression variants is a prerequisite for considering them as essential in the matching process of hematopoietic stem cell donors and recipients to diminish the risk of serious complications such as graft-versus-host disease or graft rejection. The HLA-A*3014L variant has a disulfide bridge missing in the alpha2 domain which could affect peptide binding and presentation to T cells. DESIGN AND METHODS: HLA-A*3014L and HLA-A*3001 were expressed as truncated variants and peptides were eluted and subjected to pool sequencing by Edman degradation as well as to single-peptide sequencing by mass spectrometry. Quantitative analysis of binding peptides presented in vivo was performed by a flow cytometric peptide-binding assay using HLA-A*3001 and HLA-A*3014L-expressing B-LCLs. RESULTS: The truncated HLA-A*3014L protein was secreted in the supernatant and it was possible to elute and sequence peptides. Sequence analysis of these eluted peptides revealed no relevant differences to the peptide motif of HLA-A*3001, indicating that the Cys164Ser substitution does not substantially alter the spectrum of presented peptides. Strong binding of one of the shared in vivo identified HLA-A*3001/3014L ligands was confirmed in the peptide-binding assay. CONCLUSIONS: This study is the first to demonstrate that HLA low expression variants are able to present peptides and, thus, can be considered as functionally active. When comparing peptide motifs, it is likely that HLA-A*3014L and HLA-A*3001 represent a permissive mismatch with low allogenicity in hematopoietic stem cell transplantation. These results indicate that surface expression, as well as peptide-binding data of HLA variants with similar disulfide bridge variations (e.g. HLA-A*3211Q) need to be considered as functionally active in an allogeneic hematopoietic stem cell transplantation setting as long as the opposite has not been shown. Otherwise a relevant but not considered HLA mismatch could result in a severe allogeneic T-cell response and graft-versus-host disease.


Subject(s)
Alleles , Amino Acid Motifs/genetics , HLA-A Antigens/genetics , Peptides/genetics , Amino Acid Sequence , Amino Acid Substitution , Binding Sites/genetics , Gene Expression , HEK293 Cells , HLA-A Antigens/metabolism , Humans , Mass Spectrometry , Models, Molecular , Mutagenesis , Peptides/chemistry , Peptides/metabolism , Protein Binding , Protein Structure, Tertiary , Sequence Analysis, Protein
20.
Anticancer Res ; 27(4A): 2075-7, 2007.
Article in English | MEDLINE | ID: mdl-17649824

ABSTRACT

BACKGROUND: Human leukocyte antigen (HLA) class I plays a central role in the immune defense against microbes and tumors by presenting endogenously processed peptides to T-cells. Few studies have addressed the question as to what extent variants of the same allelic HLA group differ in their peptide motif. MATERIALS AND METHODS: To answer this question for the HLA-A*66 family, which is a sub-group of the widespread serological HLA-A10 group, peptides eluted from recombinant A*6602 and A*6603 molecules were sequenced and have with data previously reported for A*6601. RESULTS: The amino acid exchange between A*6602 and A*6603 (Gln70His) has no impact on the peptide motif. By contrast, the amino acid exchange Arg163Glu that distinguishes A*6601 from A*6602/6603 results in a change of the peptides preferentially bound. CONCLUSION: Minor amino acid differences between HLA molecules may have a large impact on peptide presentation. For immunotherapy based on tumor-related peptides, it is crucial that the tumor specific peptide is presented by the patients' HLA molecules. For this reason, patients need to be typed not only at the HLA group level, but also at the allelic level.


Subject(s)
Alleles , Antigen Presentation/genetics , HLA-A Antigens/genetics , Immunotherapy/methods , Neoplasms/therapy , Amino Acid Sequence , HLA-A Antigens/chemistry , Humans , Molecular Sequence Data , Phylogeny , Protein Structure, Quaternary
SELECTION OF CITATIONS
SEARCH DETAIL
...