Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 622
Filter
Add more filters

Publication year range
1.
Nat Immunol ; 24(7): 1087-1097, 2023 07.
Article in English | MEDLINE | ID: mdl-37264229

ABSTRACT

Human leukocyte antigen (HLA)-E binds epitopes derived from HLA-A, HLA-B, HLA-C and HLA-G signal peptides (SPs) and serves as a ligand for CD94/NKG2A and CD94/NKG2C receptors expressed on natural killer and T cell subsets. We show that among 16 common classical HLA class I SP variants, only 6 can be efficiently processed to generate epitopes that enable CD94/NKG2 engagement, which we term 'functional SPs'. The single functional HLA-B SP, known as HLA-B/-21M, induced high HLA-E expression, but conferred the lowest receptor recognition. Consequently, HLA-B/-21M SP competes with other SPs for providing epitope to HLA-E and reduces overall recognition of target cells by CD94/NKG2A, calling for reassessment of previous disease models involving HLA-B/-21M. Genetic population data indicate a positive correlation between frequencies of functional SPs in humans and corresponding cytomegalovirus mimics, suggesting a means for viral escape from host responses. The systematic, quantitative approach described herein will facilitate development of prediction algorithms for accurately measuring the impact of CD94/NKG2-HLA-E interactions in disease resistance/susceptibility.


Subject(s)
Killer Cells, Natural , Protein Sorting Signals , Humans , Histocompatibility Antigens Class I , HLA Antigens/metabolism , Histocompatibility Antigens Class II/metabolism , NK Cell Lectin-Like Receptor Subfamily D/genetics , NK Cell Lectin-Like Receptor Subfamily D/metabolism , Lectins, C-Type/metabolism , Receptors, Natural Killer Cell/metabolism , HLA-E Antigens
2.
Annu Rev Immunol ; 31: 529-61, 2013.
Article in English | MEDLINE | ID: mdl-23298204

ABSTRACT

The MHC fold is found in proteins that have a range of functions in the maintenance of an organism's health, from immune regulation to fat metabolism. Well adapted for antigen presentation, as seen for peptides in the classical MHC molecules and for lipids in CD1 molecules, the MHC fold has also been modified to perform Fc-receptor activity (e.g., FcRn) and for roles in host homeostasis (e.g., with HFE and ZAG). The more divergent MHC-like molecules, such as some of those that interact with the NKG2D receptor, represent the minimal MHC fold, doing away with the α3 domain and ß2m while maintaining the α1/α2 platform domain for receptor engagement. Viruses have also co-opted the MHC fold for immune-evasive functions. The variations on the theme of a ß-sheet topped by two semiparallel α-helices are discussed in this review, highlighting the fantastic adaptability of this fold for good and for bad.


Subject(s)
Antigen Presentation/immunology , Histocompatibility Antigens Class I/chemistry , Histocompatibility Antigens Class I/physiology , Immunity, Innate , Animals , HLA-G Antigens/metabolism , Histocompatibility Antigens Class I/metabolism , Humans , Mice , Protein Folding , Structure-Activity Relationship , HLA-E Antigens
3.
Cell ; 175(7): 1744-1755.e15, 2018 12 13.
Article in English | MEDLINE | ID: mdl-30503208

ABSTRACT

Tumor-infiltrating CD8 T cells were found to frequently express the inhibitory receptor NKG2A, particularly in immune-reactive environments and after therapeutic cancer vaccination. High-dimensional cluster analysis demonstrated that NKG2A marks a unique immune effector subset preferentially co-expressing the tissue-resident CD103 molecule, but not immune checkpoint inhibitors. To examine whether NKG2A represented an adaptive resistance mechanism to cancer vaccination, we blocked the receptor with an antibody and knocked out its ligand Qa-1b, the conserved ortholog of HLA-E, in four mouse tumor models. The impact of therapeutic vaccines was greatly potentiated by disruption of the NKG2A/Qa-1b axis even in a PD-1 refractory mouse model. NKG2A blockade therapy operated through CD8 T cells, but not NK cells. These findings indicate that NKG2A-blocking antibodies might improve clinical responses to therapeutic cancer vaccines.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines , Immunity, Cellular , NK Cell Lectin-Like Receptor Subfamily C , Neoplasm Proteins , Neoplasms, Experimental , Vaccination , Animals , Antibodies, Neoplasm/immunology , Antigens, CD/immunology , CD8-Positive T-Lymphocytes/pathology , Cancer Vaccines/immunology , Cancer Vaccines/pharmacology , Cell Line, Tumor , Histocompatibility Antigens Class I/immunology , Humans , Integrin alpha Chains/immunology , Mice , NK Cell Lectin-Like Receptor Subfamily C/antagonists & inhibitors , NK Cell Lectin-Like Receptor Subfamily C/immunology , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/immunology , Neoplasms, Experimental/immunology , Neoplasms, Experimental/pathology , Neoplasms, Experimental/therapy , HLA-E Antigens
4.
Proc Natl Acad Sci U S A ; 121(19): e2318003121, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38691588

ABSTRACT

Peptides presented by HLA-E, a molecule with very limited polymorphism, represent attractive targets for T cell receptor (TCR)-based immunotherapies to circumvent the limitations imposed by the high polymorphism of classical HLA genes in the human population. Here, we describe a TCR-based bispecific molecule that potently and selectively binds HLA-E in complex with a peptide encoded by the inhA gene of Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis in humans. We reveal the biophysical and structural bases underpinning the potency and specificity of this molecule and demonstrate its ability to redirect polyclonal T cells to target HLA-E-expressing cells transduced with mycobacterial inhA as well as primary cells infected with virulent Mtb. Additionally, we demonstrate elimination of Mtb-infected cells and reduction of intracellular Mtb growth. Our study suggests an approach to enhance host T cell immunity against Mtb and provides proof of principle for an innovative TCR-based therapeutic strategy overcoming HLA polymorphism and therefore applicable to a broader patient population.


Subject(s)
Histocompatibility Antigens Class I , Mycobacterium tuberculosis , Receptors, Antigen, T-Cell , T-Lymphocytes , Mycobacterium tuberculosis/immunology , Humans , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , T-Lymphocytes/immunology , HLA-E Antigens , Bacterial Proteins/immunology , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Tuberculosis/immunology
5.
J Immunol ; 212(4): 715-722, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38149913

ABSTRACT

Immune checkpoint molecules are promising targets for suppressing the immune response but have received little attention in immune tolerance induction in organ transplantation. In this study, we found that IFN-ß could induce the expression of HLA-E as well as PD-L1 on human renal tubular epithelial cell line HK-2 and renal tissue of the C57BL/6 mouse. The JAK/STAT2 pathway was necessary for this process. Upregulation of both HLA-E and PD-L1 was fully abrogated by the JAK1/2 inhibitor ruxolitinib. Signaling pathway molecules, including STAT1, STAT2, mTOR, Tyk2, and p38 MAPK, were involved in HLA-E and PD-L1 upregulation. IRF7 is the key transcription factor responsible for the activation of HLA-E and PD-L1 promoters. Through screening an epigenetic regulation library, we found a natural compound, bisdemethoxycurcumin, enhanced IFN-ß-induced HLA-E and PD-L1 expression in vitro and in vivo. In PBMC-derived CD56+ NK cells, we found that NKG2A but not PD1 was constitutively expressed, indicating HLA-E/NKG2A as a more potent target to induce tolerance to innate immune cells. Pretreating HK-2 cells by IFN-ß significantly attenuated the degranulation of their coincubated NK cells and protected cells from NK-mediated lysis. In conclusion, IFN-ß pretreatment could activate HLA-E and PD-L1 transcription through the JAK/STAT/IRF7 pathway and then could protect renal tubular epithelial cells from allogeneic immune attack mediated by NK cells.


Subject(s)
HLA-E Antigens , Hematopoietic Stem Cell Transplantation , Mice , Animals , Humans , B7-H1 Antigen/metabolism , Leukocytes, Mononuclear , Epigenesis, Genetic , Mice, Inbred C57BL , Histocompatibility Antigens Class I , Killer Cells, Natural , Epithelial Cells
6.
Blood ; 141(13): 1560-1573, 2023 03 30.
Article in English | MEDLINE | ID: mdl-36477802

ABSTRACT

Primary Epstein-Barr virus (EBV) infections may cause infectious mononucleosis (IM), whereas EBV reactivations in solid organ and hematopoietic stem cell transplant recipients are associated with posttransplantation lymphoproliferative disorders (PTLDs). It is still unclear why only a minority of primary EBV-infected individuals develop IM, and why only some patients progress to EBV+PTLD after transplantation. We now investigated whether nonclassic human leukocyte antigen E (HLA-E)-restricted immune responses have a significant impact on the development of EBV diseases in the individual host. On the basis of a large study cohort of 1404 patients and controls as well as on functional natural killer (NK) and CD8+ T-cell analyses, we could demonstrate that the highly expressed HLA-E∗0103/0103 genotype is protective against IM, due to the induction of potent EBV BZLF1-specific HLA-E-restricted CD8+ T-cell responses, which efficiently prevent the in vitro viral dissemination. Furthermore, we provide evidence that the risk of symptomatic EBV reactivations in immunocompetent individuals as well as in immunocompromised transplant recipients depends on variations in the inhibitory NKG2A/LMP-1/HLA-E axis. We show that EBV strains encoding for the specific LMP-1 peptide variants GGDPHLPTL or GGDPPLPTL, presented by HLA-E, elicit strong inhibitory NKG2A+ NK and CD8+ T-cell responses. The presence of EBV strains encoding for both peptides was highly associated with symptomatic EBV reactivations. The further progression to EBV+PTLD was highly associated with the presence of both peptide-encoding EBV strains and the expression of HLA-E∗0103/0103 in the host. Thus, HLA-E-restricted immune responses and the NKG2A/LMP-1/HLA-E axis are novel predictive markers for EBV+PTLD in transplant recipients and should be considered for future EBV vaccine design.


Subject(s)
Epstein-Barr Virus Infections , Infectious Mononucleosis , Lymphoproliferative Disorders , Humans , Epstein-Barr Virus Infections/complications , Herpesvirus 4, Human/genetics , Lymphoproliferative Disorders/etiology , Lymphoproliferative Disorders/prevention & control , HLA Antigens , Infectious Mononucleosis/prevention & control , Immunity , HLA-E Antigens
7.
PLoS Comput Biol ; 20(9): e1011718, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39283896

ABSTRACT

In addition to the classical HLA genes, the major histocompatibility complex (MHC) harbors a high number of other polymorphic genes with less established roles in disease associations and transplantation matching. To facilitate studies of the non-classical and non-HLA genes in large patient and biobank cohorts, we trained imputation models for MICA, MICB, HLA-E, HLA-F and HLA-G alleles on genome SNP array data. We show, using both population-specific and multi-population 1000 Genomes references, that the alleles of these genes can be accurately imputed for screening and research purposes. The best imputation model for MICA, MICB, HLA-E, -F and -G achieved a mean accuracy of 99.3% (min, max: 98.6, 99.9). Furthermore, validation of the 1000 Genomes exome short-read sequencing-based allele calling against a clinical-grade reference data showed an average accuracy of 99.8%, testifying for the quality of the 1000 Genomes data as an imputation reference. We also fitted the models for Infinium Global Screening Array (GSA, Illumina, Inc.) and Axiom Precision Medicine Research Array (PMRA, Thermo Fisher Scientific Inc.) SNP content, with mean accuracies of 99.1% (97.2, 100) and 98.9% (97.4, 100), respectively.


Subject(s)
Alleles , Histocompatibility Antigens Class I , Polymorphism, Single Nucleotide , Humans , Polymorphism, Single Nucleotide/genetics , Histocompatibility Antigens Class I/genetics , Genome, Human/genetics , HLA-E Antigens , Computational Biology/methods
8.
Mol Ther ; 32(3): 678-688, 2024 Mar 06.
Article in English | MEDLINE | ID: mdl-38219014

ABSTRACT

Naturally occurring T cells that recognize microbial peptides via HLA-E, a nonpolymorphic HLA class Ib molecule, could provide the foundation for new universal immunotherapeutics. However, confidence in the biological relevance of putative ligands is crucial, given that the mechanisms by which pathogen-derived peptides can access the HLA-E presentation pathway are poorly understood. We systematically interrogated the HIV proteome using immunopeptidomic and bioinformatic approaches, coupled with biochemical and cellular assays. No HIV HLA-E peptides were identified by tandem mass spectrometry analysis of HIV-infected cells. In addition, all bioinformatically predicted HIV peptide ligands (>80) were characterized by poor complex stability. Furthermore, infected cell elimination assays using an affinity-enhanced T cell receptor bispecific targeted to a previously reported HIV Gag HLA-E epitope demonstrated inconsistent presentation of the peptide, despite normal HLA-E expression on HIV-infected cells. This work highlights the instability of the HIV HLA-E peptidome as a major challenge for drug development.


Subject(s)
HIV Infections , HLA-E Antigens , Humans , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/metabolism , Epitopes , HIV Infections/therapy , Peptides/metabolism
9.
Mol Ther ; 32(8): 2711-2727, 2024 Aug 07.
Article in English | MEDLINE | ID: mdl-38943249

ABSTRACT

Natural killer (NK) cells eliminate infected or cancer cells via their cytotoxic capacity. NKG2A is an inhibitory receptor on NK cells and cancer cells often overexpress its ligand HLA-E to evade NK cell surveillance. Given the successes of immune checkpoint blockade in cancer therapy, NKG2A is an interesting novel target. However, anti-NKG2A antibodies have shown limited clinical response. In the pursuit of enhancing NK cell-mediated anti-tumor responses, we devised a Cas9-based strategy to delete KLRC1, encoding NKG2A, in human primary NK cells. Our approach involved electroporation of KLRC1-targeting Cas9 ribonucleoprotein resulting in effective ablation of NKG2A expression. Compared with anti-NKG2A antibody blockade, NKG2AKO NK cells exhibited enhanced activation, reduced suppressive signaling, and elevated expression of key transcription factors. NKG2AKO NK cells overcame inhibition from HLA-E, significantly boosting NK cell activity against solid and hematologic cancer cells. We validated this efficacy across multiple cell lines, a xenograft mouse model, and primary human leukemic cells. Combining NKG2A knockout with antibody coating of tumor cells further enhanced cytotoxicity through ADCC. Thus, we provide a comprehensive comparison of inhibition of the NKG2A pathway using genetic ablation and antibodies and provide novel insight in the observed differences in molecular mechanisms, which can be translated to enhance adoptive NK cell immunotherapy.


Subject(s)
Killer Cells, Natural , NK Cell Lectin-Like Receptor Subfamily C , Xenograft Model Antitumor Assays , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , NK Cell Lectin-Like Receptor Subfamily C/genetics , NK Cell Lectin-Like Receptor Subfamily C/metabolism , Animals , Mice , Cell Line, Tumor , HLA-E Antigens , Neoplasms/immunology , Neoplasms/therapy , Neoplasms/genetics , Antibodies, Monoclonal/pharmacology , CRISPR-Cas Systems , Gene Deletion , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/metabolism , Cytotoxicity, Immunologic
10.
J Immunol ; 209(8): 1555-1565, 2022 10 15.
Article in English | MEDLINE | ID: mdl-36096642

ABSTRACT

Tuberculosis (TB) remains one of the deadliest infectious diseases worldwide, posing great social and economic burden to affected countries. Novel vaccine approaches are needed to increase protective immunity against the causative agent Mycobacterium tuberculosis (Mtb) and to reduce the development of active TB disease in latently infected individuals. Donor-unrestricted T cell responses represent such novel potential vaccine targets. HLA-E-restricted T cell responses have been shown to play an important role in protection against TB and other infections, and recent studies have demonstrated that these cells can be primed in vitro. However, the identification of novel pathogen-derived HLA-E binding peptides presented by infected target cells has been limited by the lack of accurate prediction algorithms for HLA-E binding. In this study, we developed an improved HLA-E binding peptide prediction algorithm and implemented it to identify (to our knowledge) novel Mtb-derived peptides with capacity to induce CD8+ T cell activation and that were recognized by specific HLA-E-restricted T cells in Mycobacterium-exposed humans. Altogether, we present a novel algorithm for the identification of pathogen- or self-derived HLA-E-presented peptides.


Subject(s)
Mycobacterium tuberculosis , Tuberculosis , Antigens, Bacterial , CD8-Positive T-Lymphocytes , Epitopes, T-Lymphocyte , Histocompatibility Antigens Class I , Humans , Peptides , HLA-E Antigens
11.
Immunology ; 168(3): 526-537, 2023 03.
Article in English | MEDLINE | ID: mdl-36217755

ABSTRACT

There is growing interest in HLA-E-restricted T-cell responses as a possible novel, highly conserved, vaccination targets in the context of infectious and malignant diseases. The developing field of HLA multimers for the detection and study of peptide-specific T cells has allowed the in-depth study of TCR repertoires and molecular requirements for efficient antigen presentation and T-cell activation. In this study, we developed a method for efficient peptide thermal exchange on HLA-E monomers and multimers allowing the high-throughput production of HLA-E multimers. We optimized the thermal-mediated peptide exchange, and flow cytometry staining conditions for the detection of TCR and NKG2A/CD94 receptors, showing that this novel approach can be used for high-throughput identification and analysis of HLA-E-binding peptides which could be involved in T-cell and NK cell-mediated immune responses. Importantly, our analysis of NKG2A/CD94 interaction in the presence of modified peptides led to new molecular insights governing the interaction of HLA-E with this receptor. In particular, our results reveal that interactions of HLA-E with NKG2A/CD94 and the TCR involve different residues. Altogether, we present a novel HLA-E multimer technology based on thermal-mediated peptide exchange allowing us to investigate the molecular requirements for HLA-E/peptide interaction with its receptors.


Subject(s)
Histocompatibility Antigens Class I , Killer Cells, Natural , Protein Binding , Histocompatibility Antigens Class I/metabolism , Peptides , Receptors, Antigen, T-Cell , NK Cell Lectin-Like Receptor Subfamily D/chemistry , NK Cell Lectin-Like Receptor Subfamily D/metabolism , NK Cell Lectin-Like Receptor Subfamily C , HLA-E Antigens
12.
Eur J Immunol ; 52(4): 618-632, 2022 04.
Article in English | MEDLINE | ID: mdl-35108401

ABSTRACT

The nonpolymorphic class Ib molecule, HLA-E, primarily presents peptides from HLA class Ia leader peptides, providing an inhibitory signal to NK cells via CD94/NKG2 interactions. Although peptides of pathogenic origin can also be presented by HLA-E to T cells, the molecular basis underpinning their role in antigen surveillance is largely unknown. Here, we solved a co-complex crystal structure of a TCR with an HLA-E presented peptide (pHLA-E) from bacterial (Mycobacterium tuberculosis) origin, and the first TCR-pHLA-E complex with a noncanonically presented peptide from viral (HIV) origin. The structures provided a molecular foundation to develop a novel method to introduce cysteine traps using non-natural amino acid chemistry that stabilized pHLA-E complexes while maintaining native interface contacts between the TCRs and different pHLA-E complexes. These pHLA-E monomers could be used to isolate pHLA-E-specific T cells, with obvious utility for studying pHLA-E restricted T cells, and for the identification of putative therapeutic TCRs.


Subject(s)
Amino Acids , HLA Antigens , Histocompatibility Antigens Class I , Peptides , Receptors, Antigen, T-Cell , HLA-E Antigens
13.
Ann Surg Oncol ; 30(2): 1240-1252, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36149610

ABSTRACT

BACKGROUND: Some reports showed the immune tolerance of soluble human leukocyte antigen E (HLA-E), but the role that soluble HLA-E plays in gastric cancer (GC) is unknown. We aimed to clarify the molecular mechanism and clinical significance of soluble HLA-E in GC. METHODS: We examined the expression of HLA-E on GC cells and soluble HLA-E under co-culture with natural killer (NK) cells in a time-dependent manner. Changes in NK cell activity were investigated using anti-NK group 2 member A (NKG2A) antibodies in the presence of soluble HLA-E. Expression of soluble HLA-E in the serum of GC patients was determined. RESULTS: Whereas HLA-E expression on GC cells peaked with interferon (IFN)-γ secretion by NK cells in a time-dependent manner, soluble HLA-E was upregulated in conditioned medium. Pre-incubation with anti-NKG2A antibodies increased the activation of NKG2A+ NK cells in the presence of soluble HLA-E. Expression of soluble HLA-E in the serum of GC patients correlated with disease progression. CONCLUSIONS: HLA-E expression dynamically changes on GC cells and in conditioned medium. Furthermore, soluble HLA-E can contribute to immune escape in GC cell lines, which may have significance in clinical practice. Moreover, soluble HLA-E may be a potential prognostic biomarker.


Subject(s)
Stomach Neoplasms , Humans , Stomach Neoplasms/metabolism , Culture Media, Conditioned/metabolism , Histocompatibility Antigens Class I , Killer Cells, Natural , HLA Antigens/metabolism , HLA-E Antigens
14.
J Chem Inf Model ; 63(11): 3486-3499, 2023 06 12.
Article in English | MEDLINE | ID: mdl-37207294

ABSTRACT

Natural killer (NK) cells play an important role in the innate immune response against tumors and various pathogens such as viruses and bacteria. Their function is controlled by a wide array of activating and inhibitory receptors, which are expressed on their cell surface. Among them is a dimeric NKG2A/CD94 inhibitory transmembrane (TM) receptor which specifically binds to the non-classical MHC I molecule HLA-E, which is often overexpressed on the surface of senescent and tumor cells. Using the Alphafold 2 artificial intelligence system, we constructed the missing segments of the NKG2A/CD94 receptor and generated its complete 3D structure comprising extracellular (EC), TM, and intracellular regions, which served as a starting point for the multi-microsecond all-atom molecular dynamics simulations of the receptor with and without the bound HLA-E ligand and its nonameric peptide. The simulated models revealed that an intricate interplay of events is taking place between the EC and TM regions ultimately affecting the intracellular immunoreceptor tyrosine-based inhibition motif (ITIM) regions that host the point at which the signal is transmitted further down the inhibitory signaling cascade. Signal transduction through the lipid bilayer was also coupled with the changes in the relative orientation of the NKG2A/CD94 TM helices in response to linker reorganization, mediated by fine-tuned interactions in the EC region of the receptor, taking place after HLA-E binding. This research provides atomistic details of the cells' protection mechanism against NK cells and broadens the knowledge regarding the TM signaling of ITIM-bearing receptors.


Subject(s)
NK Cell Lectin-Like Receptor Subfamily C , Receptors, Immunologic , NK Cell Lectin-Like Receptor Subfamily C/metabolism , Receptors, Natural Killer Cell/metabolism , Receptors, Immunologic/chemistry , Receptors, Immunologic/metabolism , Ligands , Artificial Intelligence , Histocompatibility Antigens Class I/metabolism , Signal Transduction , Carrier Proteins/metabolism , HLA-E Antigens
15.
BMC Genomics ; 23(1): 455, 2022 Jun 20.
Article in English | MEDLINE | ID: mdl-35725386

ABSTRACT

BACKGROUND: Congenital cytomegalovirus immunopathogenesis is largely unknown and multifactorial due to the complex interactions between viral, maternal, placental, and child factors. Polymorphisms in the HLA-E binding UL4015-23 peptide mimics HLA-E complexed peptides from certain HLA-A, -B, -C and -G alleles, which regulate the cellular immune response driven by natural killer-cells (NK) and CD8 + T cells. The aim of this study was to compare UL4015-23 peptides distribution in congenital CMV and the counterpart HLA Class I peptides in a healthy cohort to investigate risk factors and markers for cCMV disease. In this 10-year retrospective study, the UL40 gene was directly sequenced from 242 clinical samples from 199 cases of congenital CMV (166 children and 33 pregnant or breast feeding women). Distribution of HLA-E binding UL4015-23 peptides was analyzed and compared to those of HLA Class I observed in a cohort of 444 healthy individuals. RESULTS: Nineteen different HLA-E binding UL4015-23 peptides were found. Three of them (VMAPRTLIL, VMAPRTLLL, VMAPRTLVL) were found in 88.3% of UL40 and 100% of HLA Class I of healthy individuals. In contrast, 15 of them (10.7%) were not found in HLA Class I. The VMAPRTLFL peptide was found in 1% of UL40 and all HLA-G alleles. Significant differences in peptide (VMAPRTLIL, VMAPRTLLL, VMAPRTLVL, other UL4015-23 peptides, other HLA Class I peptides) distribution between UL4015-23 from congenital CMV and HLA-A, -B, -C and -G from healthy individuals were found. CONCLUSIONS: Our findings suggest that a mismatch between UL4015-23 peptides and HLA Class I peptides between children and mothers might play a role in congenital CMV disease, and it may account for differences in outcome, morbidity and sequelae.


Subject(s)
Cytomegalovirus Infections , Cytomegalovirus , Histocompatibility Antigens Class I , Viral Proteins , CD8-Positive T-Lymphocytes , Child , Cytomegalovirus/genetics , Cytomegalovirus Infections/genetics , Cytomegalovirus Infections/metabolism , Female , HLA-A Antigens/metabolism , Humans , Peptides/genetics , Peptides/metabolism , Placenta/metabolism , Pregnancy , Retrospective Studies , Risk Factors , Viral Proteins/genetics , HLA-E Antigens
16.
Immunology ; 166(4): 507-521, 2022 08.
Article in English | MEDLINE | ID: mdl-35596615

ABSTRACT

NKG2A has emerged as a new immunotherapy target and its blockade with the novel immune checkpoint inhibitor (ICI) monalizumab can boost both NK cell and CD8+ T cell responses. NKG2A forms heterodimers with CD94 and binds to the human non-classical MHC class I molecule HLA-E. HLA-E forms complexes with a limited set of peptides mainly derived from the leader sequences of the classical MHC class I molecules (HLA-A, HLA-B and HLA-C) and the non-classical class I paralogue HLA-G, and it is well established that the interaction between CD94/NKG2x receptors and its ligand HLA-E is peptide-sensitive. Here, we have evaluated peptide dependence of NKG2A-mediated inhibition and the efficiency of interference by monalizumab in a transcriptional T cell reporter system. NKG2A inhibition was mediated by cell-expressed HLA-E molecules stably presenting disulfate-trapped peptide ligands. We show that different HLA-class I leader peptides mediate varying levels of inhibition. We have used NKG2A/NKG2C chimeric receptors to map the binding site of NKG2A and NKG2C blocking antibodies. Furthermore, we determined the functional EC50 values of blocking NKG2A antibodies and show that they greatly depend on the HLA-leader peptide presented by HLA-E. Monalizumab was less effective in augmenting NK cell-mediated killing of target cells displaying HLA-G peptide on HLA-E, than cells expressing HLA-E complexed with HLA-A, HLA-B and HLA-C peptides. Our results indicate that peptides displayed by HLA-E molecules on tumour cells might influence the effectivity of NKG2A-ICI therapy and potentially suggest novel approaches for patient stratification, for example, based on tumoral HLA-G levels.


Subject(s)
HLA-C Antigens , HLA-G Antigens , NK Cell Lectin-Like Receptor Subfamily C/metabolism , HLA-A Antigens , Histocompatibility Antigens Class I/metabolism , Humans , Ligands , NK Cell Lectin-Like Receptor Subfamily D , Peptides , HLA-E Antigens
17.
Eur J Immunol ; 51(10): 2513-2521, 2021 10.
Article in English | MEDLINE | ID: mdl-34323289

ABSTRACT

Recent studies have indicated the antitumor activity and reduced allogeneic response of universal chimeric antigen receptor-modified T (UCAR T) cells lacking endogenous T cell receptors and beta-2 microglobulin (B2M) generated using gene-editing technologies. However, these cells are vulnerable to lysis by allogeneic natural killer (NK) cells due to their lack of human leukocyte antigen (HLA) class I molecule expression. Here, constitutive expression of mutant B2M-HLA-E (mBE) and B2M-HLA-G (mBG) fusion proteins in anti-CD19 UCAR T (UCAR T-19) cells was conducted to protect against allogeneic NK cell-mediated lysis. The ability of cells expressing mBE or mBG to resist NK cell-mediated lysis was observed in gene-edited Jurkat CAR19 cells. UCAR T-19 cells constitutively expressing the mBE and mBG fusion proteins were manufactured and showed effective and specific anti-tumor activity. Constitutive expression of the mBE and mBG fusion proteins in UCAR T-19 cells prevented allogeneic NK cell-mediated lysis. In addition, these cells were not recognizable by allogeneic T cells. Additional experiments, including those in animal models and clinical trials, are required to evaluate the safety and efficacy of UCAR T-19 cells that constitutively express mBE and mBG.


Subject(s)
Cytotoxicity, Immunologic/genetics , HLA-G Antigens/genetics , Histocompatibility Antigens Class I/genetics , Mutation , Receptors, Chimeric Antigen/genetics , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , beta 2-Microglobulin/genetics , Antigens, CD19/immunology , Gene Knockout Techniques , HLA-G Antigens/immunology , Histocompatibility Antigens Class I/immunology , Humans , Immunophenotyping , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Lymphocyte Activation/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Receptors, Chimeric Antigen/immunology , beta 2-Microglobulin/immunology , HLA-E Antigens
18.
Ann Surg Oncol ; 29(8): 4951-4960, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35412205

ABSTRACT

BACKGROUND: The NKG2A/HLA-E pathway functions as an immune checkpoint with potential for inhibition using therapeutic antibodies. Through this pathway, immune cells lose activity, which allows cancers to progress. We aimed to determine whether HLA-E expression combined with NK cell status serves as a prognostic biomarker for gastric cancer (GC). METHODS: We enrolled patients (n = 232) with advanced GC who underwent curative gastrectomy. Immunohistochemical analyses of global HLA-E expression, and the expression of CD56 and CD3 to identify NK cells were performed. Survival analysis was performed to evaluate the significance of HLA-E expression and NK status. RESULTS: Patients with HLA-E-positive was 104 (41.3%) and had significantly worse prognosis of relapse-free survival (RFS) compared with those with HLA-E-negative. Moreover, patients with NK Low status had worse prognoses for RFS compared with those with NK High status. Statistical analysis of RFS demonstrated that HLA-E expression was a significant independent factor for poor prognosis (HR 1.57, 95% CI 1.04-2.36, P = 0.031). Furthermore, HLA-E-positive patients with low NK low status experienced the shortest RFS, particularly those in the upper GC group. CONCLUSIONS: HLA-E served as a prognostic factor after curative resection of GC, and HLA-E expression combined with NK status served as a sensitive prognostic biomarker for advanced GC.


Subject(s)
Stomach Neoplasms , Biomarkers/metabolism , Histocompatibility Antigens Class I , Humans , Killer Cells, Natural , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/surgery , Prognosis , Stomach Neoplasms/pathology , HLA-E Antigens
19.
Am J Obstet Gynecol ; 227(4): 641.e1-641.e13, 2022 10.
Article in English | MEDLINE | ID: mdl-35863458

ABSTRACT

BACKGROUND: The extravillous trophoblast expresses each of the nonclassical major histocompatibility complex class I antigens-human leukocyte antigens E, F, and G-and a single classical class I antigen, human leukocyte antigen C. We recently demonstrated dynamic expression patterns of human leukocyte antigens C, G, and F during early extravillous trophoblast invasion and placentation. OBJECTIVE: This study aimed to investigate the hypothesis that the immune inflammatory mediated complications of pregnancy such as early preeclampsia and preterm labor may show altered expression profiles of nonclassical human leukocyte antigens. STUDY DESIGN: Real-time quantitative polymerase chain reaction, western blot, and immunohistochemistry were performed on placental villous tissues and basal plate sections from term nonlaboring deliveries, preterm deliveries, and severe early-onset preeclampsia, both with and without small-for-gestational-age neonates. RESULTS: Human leukocyte antigen G is strongly and exclusively expressed by the extravillous trophoblast within the placental basal plate, and its levels increase in pregnancies complicated by severe early-onset preeclampsia with small-for-gestational-age neonates relative to those of healthy term controls. Human leukocyte antigen C shows a similar profile in the extravillous trophoblast of preeclamptic pregnancies, but significantly decreases in the villous placenta. Human leukocyte antigen F protein levels are decreased in both extravillous trophoblast and villous placenta of severe early-onset preeclamptic pregnancies, both with and without small-for-gestational-age neonates, compared with those found in term and preterm birth deliveries. Human leukocyte antigen E decreases in blood vessels in placentas from preeclamptic pregnancies relative to its levels in term and preterm birth deliveries. Placental levels of human leukocyte antigens F and C are increased in cases of preterm birth with chorioamnionitis relative to those of cases of idiopathic preterm birth. CONCLUSION: Dysregulation of placental human leukocyte antigen expression at the maternal-fetal interface may contribute to compromised maternal tolerance in preterm birth with chorioamnionitis and excessive maternal systemic inflammation associated with severe early-onset preeclampsia.


Subject(s)
Chorioamnionitis , Pre-Eclampsia , Premature Birth , Chorioamnionitis/metabolism , Female , Fetal Growth Retardation/metabolism , HLA-C Antigens/metabolism , HLA-G Antigens/metabolism , Histocompatibility Antigens Class I , Humans , Infant, Newborn , Placenta/metabolism , Placentation , Pre-Eclampsia/metabolism , Pregnancy , Premature Birth/metabolism , Trophoblasts/metabolism , HLA-E Antigens
20.
Crit Rev Immunol ; 41(4): 39-53, 2021.
Article in English | MEDLINE | ID: mdl-35381142

ABSTRACT

The TCR-mediated recognition of self and microbial protein antigens by CD8+ T cells presented by the relatively nonvariable, class Ib MHC molecule, Qa-1 in mice and HLA-E in humans, is emerging as an important arm of the immune response. In this brief review, we will cover key examples of Qa1/HLA-E-restricted CD8+ T cells and their role in immunity against microbes and in cancer, but also as an important immunoregulatory pathway complementary to the FoxP3+CD4+ Treg. Although much remains to be learned, increased understanding of HLA-E-targeted immune responses can be potentially exploited in the development of broader and complementary immunotherapeutics against bacteria/viruses, tumors, and autoimmune diseases.


Subject(s)
Histocompatibility Antigens Class I , Neoplasms , Animals , CD8-Positive T-Lymphocytes , Histocompatibility Antigens Class I/metabolism , Humans , Mice , Neoplasms/metabolism , Neoplasms/therapy , T-Lymphocytes, Regulatory , HLA-E Antigens
SELECTION OF CITATIONS
SEARCH DETAIL