Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 7.890
Filter
1.
Nat Commun ; 15(1): 8508, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39353943

ABSTRACT

Immune surveillance by cytotoxic T cells eliminates tumor cells and cells infected by intracellular pathogens. This process relies on the presentation of antigenic peptides by Major Histocompatibility Complex class I (MHC-I) at the cell surface. The loading of these peptides onto MHC-I depends on the peptide loading complex (PLC) at the endoplasmic reticulum (ER). Here, we uncovered that MHC-I antigen presentation is regulated by ER-associated degradation (ERAD), a protein quality control process essential to clear misfolded and unassembled proteins. An unbiased proteomics screen identified the PLC component Tapasin, essential for peptide loading onto MHC-I, as a substrate of the RNF185/Membralin ERAD complex. Loss of RNF185/Membralin resulted in elevated Tapasin steady state levels and increased MHC-I at the surface of professional antigen presenting cells. We further show that RNF185/Membralin ERAD complex recognizes unassembled Tapasin and limits its incorporation into PLC. These findings establish a novel mechanism controlling antigen presentation and suggest RNF185/Membralin as a potential therapeutic target to modulate immune surveillance.


Subject(s)
Antigen Presentation , Endoplasmic Reticulum-Associated Degradation , Endoplasmic Reticulum , Histocompatibility Antigens Class I , Membrane Transport Proteins , Ubiquitin-Protein Ligases , Humans , Membrane Transport Proteins/metabolism , Membrane Transport Proteins/genetics , Histocompatibility Antigens Class I/metabolism , Histocompatibility Antigens Class I/immunology , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Endoplasmic Reticulum/metabolism , HEK293 Cells , Membrane Proteins/metabolism , Membrane Proteins/genetics
2.
Nat Commun ; 15(1): 8722, 2024 Oct 08.
Article in English | MEDLINE | ID: mdl-39379371

ABSTRACT

Stevens-Johnson syndrome and toxic epidermal necrolysis (SJS/TEN) is a rare but life-threatening cutaneous drug reaction mediated by human leukocyte antigen (HLA) class I-restricted CD8+ T cells. For unbiased assessment of cellular immunopathogenesis, here we perform single-cell (sc) transcriptome, surface proteome, and T cell receptor (TCR) sequencing on unaffected skin, affected skin, and blister fluid from 15 SJS/TEN patients. From 109,888 cells, we identify 15 scRNA-defined subsets. Keratinocytes express markers indicating HLA class I-restricted antigen presentation and appear to trigger the proliferation of and killing by cytotoxic CD8+ tissue-resident T cells that express granulysin, granzyme B, perforin, LAG3, CD27, and LINC01871, and signal through the PKM, MIF, TGFß, and JAK-STAT pathways. In affected tissue, cytotoxic CD8+ T cells express private expanded and unexpanded TCRαß that are absent or unexpanded in unaffected skin, and mixed populations of macrophages and fibroblasts express pro-inflammatory markers or those favoring repair. This data identifies putative cytotoxic TCRs and therapeutic targets.


Subject(s)
CD8-Positive T-Lymphocytes , Keratinocytes , Receptors, Antigen, T-Cell , Single-Cell Analysis , Stevens-Johnson Syndrome , Humans , Stevens-Johnson Syndrome/immunology , Stevens-Johnson Syndrome/genetics , Single-Cell Analysis/methods , Keratinocytes/immunology , Keratinocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell/genetics , Skin/immunology , Skin/pathology , T-Lymphocytes, Cytotoxic/immunology , Granzymes/metabolism , Granzymes/genetics , Transcriptome , Male , Perforin/metabolism , Perforin/genetics , Female , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Antigens, Differentiation, T-Lymphocyte/genetics , Antigens, Differentiation, T-Lymphocyte/metabolism , Antigens, Differentiation, T-Lymphocyte/immunology , Macrophages/immunology , Macrophages/metabolism
3.
Sci Rep ; 14(1): 23401, 2024 10 08.
Article in English | MEDLINE | ID: mdl-39379424

ABSTRACT

Natural killer group 2 member D ligands (NKG2DLs) are expressed as stress response proteins in cancer cells. NKG2DLs induce immune cell activation or tumor escape responses, depending on their expression. Human pancreatic cancer cells, PANC-1, express membrane MHC class I polypeptide-related sequence A/B (mMICA/B), whereas soluble MICB (sMICB) is detected in the culture supernatant. We hypothesized that sMICB saturates NKG2D in NKG2DLow T cells and inhibits the activation signal from mMICB to NKG2D. Knockdown of MICB by siRNA reduced sMICB level, downregulated mMICB expression, maintained NKG2DLow T cell activation, and inhibited NKG2DHigh T cell activation. To maintain mMICB expression and downregulate sMICB expression, we inhibited a disintegrin and metalloproteinase (ADAM), a metalloproteinase that sheds MICB. Subsequently, the shedding of MICB was prevented using ADAM17 inhibitors, and the activation of NKG2DLow T cells was maintained. In vivo xenograft model revealed that NKG2DHigh T cells have superior anti-tumor activity. These results elucidate the mechanism of immune escape via sMICB and show potential for the activation of NKG2DLow T cells within the tumor microenvironment.


Subject(s)
Histocompatibility Antigens Class I , Lymphocyte Activation , NK Cell Lectin-Like Receptor Subfamily K , Pancreatic Neoplasms , T-Lymphocytes , NK Cell Lectin-Like Receptor Subfamily K/metabolism , NK Cell Lectin-Like Receptor Subfamily K/genetics , Histocompatibility Antigens Class I/metabolism , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/genetics , Humans , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/genetics , Animals , Cell Line, Tumor , Mice , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Lymphocyte Activation/immunology , ADAM17 Protein/metabolism , ADAM17 Protein/genetics , Tumor Escape
4.
Nat Commun ; 15(1): 8569, 2024 Oct 03.
Article in English | MEDLINE | ID: mdl-39362877

ABSTRACT

Immunotherapy successfully complements traditional cancer treatment. However, primary and acquired resistance might limit efficacy. Reduced antigen presentation by MHC-I has been identified as potential resistance factor. Here we show that the epigenetic regulator ubiquitin-like with PHD and ring finger domains 1 (UHRF1), exhibits altered expression and aberrant cytosolic localization in cancerous tissues, where it promotes MHC-I ubiquitination and degradation. Cytoplasmic translocation of UHRF1 is induced by its phosphorylation on a specific serine in response to signals provided by factors present in the tumor microenvironment (TME), such as TGF-ß, enabling UHRF1 to bind MHC-I. Downregulation of MHC-I results in suppression of the antigen presentation pathway to establish an immune hostile TME. UHRF1 inactivation by genetic deletion synergizes with immune checkpoint blockade (ICB) treatment and induces an anti-tumour memory response by evoking low-affinity T cells. Our study adds to the understanding of UHRF1 in cancer immune evasion and provides a potential target to synergize with immunotherapy and overcome immunotherapeutic resistance.


Subject(s)
CCAAT-Enhancer-Binding Proteins , Cytoplasm , Tumor Microenvironment , Ubiquitin-Protein Ligases , Ubiquitination , Animals , Female , Humans , Mice , Antigen Presentation/immunology , CCAAT-Enhancer-Binding Proteins/metabolism , CCAAT-Enhancer-Binding Proteins/genetics , Cell Line, Tumor , Cytoplasm/metabolism , Gene Expression Regulation, Neoplastic , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Histocompatibility Antigens Class I/genetics , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Immunotherapy/methods , Mice, Inbred C57BL , Neoplasms/immunology , Neoplasms/therapy , Neoplasms/genetics , Phosphorylation , Tumor Microenvironment/immunology , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Male
5.
Front Immunol ; 15: 1371156, 2024.
Article in English | MEDLINE | ID: mdl-39351230

ABSTRACT

Effective treatment and immunoprophylaxis of viral respiratory infections with neutralizing monoclonal antibodies (mAbs) require maintaining inhibitory concentrations of mAbs at the airway surface. While engineered mAbs with increased affinity to the neonatal Fc receptor (FcRn) are increasingly employed, little is known how increased affinity of Fc to FcRn influences basal-to-apical transepithelial transport (transcytosis) of mAbs across the airway epithelium. To investigate this, we utilized a model of well-differentiated human airway epithelium (WD-HAE) that exhibited robust FcRn expression, and measured the transepithelial transport of a mAb against SARS-CoV-2 Spike protein (CR3022) with either wildtype IgG1-Fc or Fc modified with YTE or LS mutations known to increase affinity for FcRn. Despite the marked differences in the affinity of these CR3022 variants for FcRn, we did not find substantial differences in basal-to-apical transport reflective of systemic dosing, or apical-to-basal transport reflective of inhaled dosing, compared to the transport of wildtype IgG1-Fc. These results suggest increasing FcRn affinity may only have limited influence over transcytosis rates of systemically dosed mAbs across the human airway epithelium over short time scales. Over longer time scales, the elevated circulating levels of mAbs with greater FcRn affinity, due to more effective FcRn-mediated recycling, may better resupply mAb into the respiratory tract, leading to more effective extended immunoprophylaxis.


Subject(s)
Antibodies, Monoclonal , Histocompatibility Antigens Class I , Immunoglobulin G , Receptors, Fc , Respiratory Mucosa , Transcytosis , Humans , Receptors, Fc/metabolism , Receptors, Fc/immunology , Histocompatibility Antigens Class I/metabolism , Histocompatibility Antigens Class I/immunology , Immunoglobulin G/immunology , Antibodies, Monoclonal/immunology , Respiratory Mucosa/metabolism , Respiratory Mucosa/immunology , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/metabolism , SARS-CoV-2/immunology , SARS-CoV-2/physiology , Antibodies, Neutralizing/immunology , COVID-19/immunology , COVID-19/prevention & control
6.
BMC Vet Res ; 20(1): 454, 2024 Oct 09.
Article in English | MEDLINE | ID: mdl-39379944

ABSTRACT

Pseudorabies virus (PRV) can establish lifelong latent infection in peripheral nervous ganglion, and persistent infections in peripheral blood lymphocytes. Establishing an infection in the lymphocytes does not only enable the PRV to escape host immune surveillance but pass through the placental barrier, leading to fetal death and abortion. Due to the pathogenicity of the PRV, it poses a huge challenge in its prevention and control. The PRV escapes host immunity through downregulation of swine leukocyte antigen class I (SLA I) molecules on infected cells. However, data on the molecular mechanisms of the SLA I suppression remains scant. Here, in order to verify the effect of candidate proteins PRV pUL44 and pUS6 on PRV immune escape related molecules SLA I and peptide loading complex (PLC), we detected the expression of SLA I and PLC components after expressing PRV pUL44 and pUS6. The effects of pUS6 and pUL44 on SLA I and PLC were analyzed by qRT-PCR and Western blot at mRNA and protein level, respectively. Cells expressing pUS6 or pUL44 genes showed a significantly suppressed expression of surface and total SLA I molecules. In addition, unlike UL44, the US6 gene was shown to downregulate the transporter associated with antigen processing 1 (TAP1), TAP2 and Tapasin molecules. The results show that PRV pUS6 may participate in virus immune escape by directly regulating the SLA I, TAP dimer and Tapasin molecules, thus blocking the transportation of TAP-bound peptides to the ER to bind SLA I molecules. We provide a theoretical basis on the mechanism of TAP mediated immune escape by the PRV.


Subject(s)
Antigen Presentation , Herpesvirus 1, Suid , Herpesvirus 1, Suid/immunology , Animals , Swine , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Viral Proteins/metabolism , Viral Proteins/genetics , Pseudorabies/immunology , Pseudorabies/virology , Cell Line
7.
J Immunother Cancer ; 12(9)2024 Sep 18.
Article in English | MEDLINE | ID: mdl-39299754

ABSTRACT

BACKGROUND: Major histocompatibility complex class I (MHC-I) loss is frequent in non-small cell lung cancer (NSCLC) rendering tumor cells resistant to T cell lysis. NK cells kill MHC-I-deficient tumor cells, and although previous work indicated their presence at NSCLC margins, they were functionally impaired. Within, we evaluated whether NK cell and CD8 T cell infiltration and activation vary with MHC-I expression. METHODS: We used single-stain immunohistochemistry (IHC) and Kaplan-Meier analysis to test the effect of NK cell and CD8 T cell infiltration on overall and disease-free survival. To delineate immune covariates of MHC-I-disparate lung cancers, we used multiplexed immunofluorescence (mIF) imaging followed by multivariate statistical modeling. To identify differences in infiltration and intercellular communication between IFNγ-activated and non-activated lymphocytes, we developed a computational pipeline to enumerate single-cell neighborhoods from mIF images followed by multivariate discriminant analysis. RESULTS: Spatial quantitation of tumor cell MHC-I expression revealed intratumoral and intertumoral heterogeneity, which was associated with the local lymphocyte landscape. IHC analysis revealed that high CD56+ cell numbers in patient tumors were positively associated with disease-free survival (HR=0.58, p=0.064) and overall survival (OS) (HR=0.496, p=0.041). The OS association strengthened with high counts of both CD56+ and CD8+ cells (HR=0.199, p<1×10-3). mIF imaging and multivariate discriminant analysis revealed enrichment of both CD3+CD8+ T cells and CD3-CD56+ NK cells in MHC-I-bearing tumors (p<0.05). To infer associations of functional cell states and local cell-cell communication, we analyzed spatial single-cell neighborhood profiles to delineate the cellular environments of IFNγ+/- NK cells and T cells. We discovered that both IFNγ+ NK and CD8 T cells were more frequently associated with other IFNγ+ lymphocytes in comparison to IFNγ- NK cells and CD8 T cells (p<1×10-30). Moreover, IFNγ+ lymphocytes were most often found clustered near MHC-I+ tumor cells. CONCLUSIONS: Tumor-infiltrating NK cells and CD8 T cells jointly affected control of NSCLC tumor progression. Coassociation of NK and CD8 T cells was most evident in MHC-I-bearing tumors, especially in the presence of IFNγ. Frequent colocalization of IFNγ+ NK cells with other IFNγ+ lymphocytes in near-neighbor analysis suggests NSCLC lymphocyte activation is coordinately regulated.


Subject(s)
CD8-Positive T-Lymphocytes , Carcinoma, Non-Small-Cell Lung , Histocompatibility Antigens Class I , Killer Cells, Natural , Lung Neoplasms , Humans , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Lung Neoplasms/immunology , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Histocompatibility Antigens Class I/metabolism , Histocompatibility Antigens Class I/immunology , Male , Female , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism
8.
Sci Adv ; 10(38): eado6491, 2024 Sep 20.
Article in English | MEDLINE | ID: mdl-39292790

ABSTRACT

Neoantigens arising from somatic mutations are tumor specific and induce antitumor host T cell responses. However, their sequences are individual specific and need to be identified for each patient for therapeutic applications. Here, we present a proteogenomic approach for neoantigen identification, named Neoantigen Selection using a Surrogate Immunopeptidome (NESSIE). This approach uses an autologous wild-type immunopeptidome as a surrogate for the tumor immunopeptidome and allows human leukocyte antigen (HLA)-agnostic identification of both HLA class I (HLA-I) and HLA class II (HLA-II) neoantigens. We demonstrate the direct identification of highly immunogenic HLA-I and HLA-II neoantigens using NESSIE in patients with colorectal cancer and endometrial cancer. Fresh or frozen tumor samples are not required for analysis, making it applicable to many patients in clinical settings. We also demonstrate tumor prevention by vaccination with selected neoantigens in a preclinical mouse model. This approach may benefit personalized T cell-mediated immunotherapies.


Subject(s)
Antigens, Neoplasm , Histocompatibility Antigens Class II , Histocompatibility Antigens Class I , Humans , Antigens, Neoplasm/immunology , Animals , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/genetics , Mice , Histocompatibility Antigens Class II/immunology , Female , Cancer Vaccines/immunology , Colorectal Neoplasms/immunology , Peptides/immunology , Endometrial Neoplasms/immunology , Immunotherapy/methods
9.
Front Immunol ; 15: 1442783, 2024.
Article in English | MEDLINE | ID: mdl-39301027

ABSTRACT

Advances in immunotherapy rely on targeting novel cell surface antigens, including therapeutically relevant peptide fragments presented by HLA molecules, collectively known as the actionable immunopeptidome. Although the immunopeptidome of classical HLA molecules is extensively studied, exploration of the peptide repertoire presented by non-classical HLA-E remains limited. Growing evidence suggests that HLA-E molecules present pathogen-derived and tumor-associated peptides to CD8+ T cells, positioning them as promising targets for universal immunotherapies due to their minimal polymorphism. This mini-review highlights recent developments in mass spectrometry (MS) technologies for profiling the HLA-E immunopeptidome in various diseases. We discuss the unique features of HLA-E, its expression patterns, stability, and the potential for identifying new therapeutic targets. Understanding the broad repertoire of actionable peptides presented by HLA-E can lead to innovative treatments for viral and pathogen infections and cancer, leveraging its monomorphic nature for broad therapeutic efficacy.


Subject(s)
HLA-E Antigens , Histocompatibility Antigens Class I , Immunotherapy , Mass Spectrometry , Humans , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Mass Spectrometry/methods , Immunotherapy/methods , mRNA Vaccines , Neoplasms/therapy , Neoplasms/immunology , Peptides/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Antigen Presentation/immunology
10.
Front Immunol ; 15: 1447980, 2024.
Article in English | MEDLINE | ID: mdl-39295866

ABSTRACT

The ubiquitous Torque teno virus (TTV) establishes a chronically persistent infection in the human host. TTV has not been associated with any apparent disease, but, as part of the human virome, it may confer a regulatory imprint on the human immune system with as yet unclear consequences. However, so far, only few studies have characterized the TTV-specific immune responses or the overall immunological imprints by TTV. Here, we reveal that TTV infection leads to a highly exhausted TTV-specific CD8+ T-cell response, hallmarked by decreased IFN-γ production and the expression of the inhibitory NKG2A-receptor. On a functional level, we identified a panel of highly polymorphic TTV-encoded peptides that lead to an expansion of regulatory NKG2A+ natural killer, NKG2A+CD4+, and NKG2A+CD8+ T cells via the stabilization of the non-classical HLA-E molecule. Our results thus demonstrate that TTV leads to a distinct imprint on the human immune system that may further regulate overall human immune responses in infectious, autoimmune, and malignant diseases.


Subject(s)
CD8-Positive T-Lymphocytes , DNA Virus Infections , HLA-E Antigens , Histocompatibility Antigens Class I , NK Cell Lectin-Like Receptor Subfamily C , Torque teno virus , Humans , Torque teno virus/immunology , NK Cell Lectin-Like Receptor Subfamily C/metabolism , NK Cell Lectin-Like Receptor Subfamily C/immunology , CD8-Positive T-Lymphocytes/immunology , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , DNA Virus Infections/immunology , Interferon-gamma/metabolism , Interferon-gamma/immunology , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Male
11.
Front Immunol ; 15: 1445338, 2024.
Article in English | MEDLINE | ID: mdl-39247192

ABSTRACT

Background: Defective ribosomal products (DRiPs) are non-functional proteins rapidly degraded during or after translation being an essential source for MHC class I ligands. DRiPs are characterized to derive from a substantial subset of nascent gene products that degrade more rapidly than their corresponding native retiree pool. So far, mass spectrometry analysis revealed that a large number of HLA class I peptides derive from DRiPs. However, a specific viral DRiP on protein level was not described. In this study, we aimed to characterize and identify DRiPs derived from a viral protein. Methods: Using the nucleoprotein (NP) of the lymphocytic choriomeningitis virus (LCMV) which is conjugated N-terminally to ubiquitin, or the ubiquitin-like modifiers FAT10 or ISG15 the occurrence of DRiPs was studied. The formation and degradation of DRiPs was monitored by western blot with the help of a FLAG tag. Flow cytometry and cytotoxic T cells were used to study antigen presentation. Results: We identified several short lived DRiPs derived from LCMV-NP. Of note, these DRiPs could only be observed when the LCMV-NP was modified with ubiquitin or ubiquitin-like modifiers, but not in the wild type form. Using proteasome inhibitors, we could show that degradation of LCMV-NP derived DRiPs were proteasome dependent. Interestingly, the synthesis of DRiPs could be enhanced when cells were stressed with the help of FCS starvation. An enhanced NP118-126 presentation was observed when the LCMV-NP was modified with ubiquitin or ubiquitin-like modifiers, or under FCS starvation. Conclusion: Taken together, we visualize for the first time DRiPs derived from a viral protein. Furthermore, DRiPs formation, and therefore MHC-I presentation, is enhanced under cellular stress conditions. Our investigations on DRiPs in MHC class I antigen presentation open up new approaches for the development of vaccination strategies.


Subject(s)
Antigen Presentation , Histocompatibility Antigens Class I , Lymphocytic choriomeningitis virus , Antigen Presentation/immunology , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Lymphocytic choriomeningitis virus/immunology , Animals , Humans , Stress, Physiological/immunology , T-Lymphocytes, Cytotoxic/immunology , Mice , Ubiquitins/metabolism , Ubiquitins/genetics , Ribosomal Proteins/metabolism , Ribosomal Proteins/immunology , Proteolysis , Nucleoproteins/immunology , Nucleoproteins/metabolism
12.
PLoS Comput Biol ; 20(9): e1012380, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39226310

ABSTRACT

Molecules of the Major Histocompatibility Complex (MHC) present short protein fragments on the cell surface, an important step in T cell immune recognition. MHC-I molecules process peptides from intracellular proteins; MHC-II molecules act in antigen-presenting cells and present peptides derived from extracellular proteins. Here we show that the sequence-dependent energy landscapes of MHC-peptide binding encode class-specific nonlinearities (epistasis). MHC-I has a smooth landscape with global epistasis; the binding energy is a simple deformation of an underlying linear trait. This form of epistasis enhances the discrimination between strong-binding peptides. In contrast, MHC-II has a rugged landscape with idiosyncratic epistasis: binding depends on detailed amino acid combinations at multiple positions of the peptide sequence. The form of epistasis affects the learning of energy landscapes from training data. For MHC-I, a low-complexity problem, we derive a simple matrix model of binding energies that outperforms current models trained by machine learning. For MHC-II, higher complexity prevents learning by simple regression methods. Epistasis also affects the energy and fitness effects of mutations in antigen-derived peptides (epitopes). In MHC-I, large-effect mutations occur predominantly in anchor positions of strong-binding epitopes. In MHC-II, large effects depend on the background epitope sequence but are broadly distributed over the epitope, generating a bigger target for escape mutations due to loss of presentation. Together, our analysis shows how an energy landscape of protein-protein binding constrains the target of escape mutations from T cell immunity, linking the complexity of the molecular interactions to the dynamics of adaptive immune response.


Subject(s)
Peptides , Protein Binding , Peptides/chemistry , Peptides/metabolism , Peptides/immunology , Humans , Computational Biology , Histocompatibility Antigens Class II/chemistry , Histocompatibility Antigens Class II/metabolism , Histocompatibility Antigens Class II/immunology , Epistasis, Genetic , Histocompatibility Antigens Class I/metabolism , Histocompatibility Antigens Class I/chemistry , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/genetics , Thermodynamics , Major Histocompatibility Complex/immunology
13.
Sci Rep ; 14(1): 20924, 2024 09 09.
Article in English | MEDLINE | ID: mdl-39251790

ABSTRACT

Human herpes viruses (HHV) are ubiquitous and have been implicated in numerous long-term health conditions. Since the association between viral exposure and long-term health impacts is partially influenced by variation in human leukocyte antigen (HLA) genes, we evaluated in silico the binding affinities of 9 HHV envelope glycoproteins with 127 common HLA Class I and Class II molecules. The findings show substantial variability in HHV binding affinity across viruses, HLA Class, HLA genes, and HLA alleles. Specific findings were as follows: (1) the predicted binding affinities of HHVs were characterized by four distinct groupings-[HHV1, HHV2], [HHV3, HHV4, HHV5], [HHV6A], [HHV6B, HHV7, HHV8]-with relatively lower binding affinities for HHV1, HHV2, and HHV6a compared to other HHVs; (2) significantly higher binding affinity was found for HLA Class I relative to Class II; (3) analyses within each class demonstrated that alleles of the C gene (for Class I) and DRB1 gene (for Class II) had the highest binding affinities; and (4) for each virus, predicted binding affinity to specific alleles varied, with HHV6a having the lowest affinity for HHV-HLA complexes, and HHV3, HHV4, and HHV5 having the highest. Since HLA-antigen binding is the first step in initiating an immune response to foreign antigens, these relative differences in HHV binding affinities are likely to influence long-term health impacts such that the cells infected with viruses associated with higher binding affinities across common HLA alleles may be more reduced in numbers, thereby lowering the potential for long-term sequelae of their infections.


Subject(s)
Alleles , Viral Envelope Proteins , Humans , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology , Viral Envelope Proteins/metabolism , Herpesviridae/immunology , Herpesviridae/genetics , HLA Antigens/genetics , HLA Antigens/immunology , Protein Binding , Immunogenetics , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology
14.
Vaccine ; 42(24): 126266, 2024 Oct 24.
Article in English | MEDLINE | ID: mdl-39232399

ABSTRACT

Tuberculosis (TB) is one of the leading causes of death from infectious diseases, killing approximately 1.3 million people worldwide in 2022 alone. The current vaccine for TB contains a live attenuated bacterium, Mycobacterium bovis BCG (Bacille Calmette-Guérin). The BCG vaccine is highly effective in preventing severe forms of childhood TB but does not protect against latent infection or disease in older age groups. A new or improved BCG vaccine for prevention of pulmonary TB is urgently needed. In this study, we infected murine bone marrow derived dendritic cells from C57BL/6 mice with M. bovis BCG followed by elution and identification of BCG-derived MHC class I and class II-bound peptides using tandem mass spectrometry. We identified 1436 MHC-bound peptides of which 94 were derived from BCG. Fifty-five peptides were derived from MHC class I molecules and 39 from class II molecules. We tested the 94 peptides for their immunogenicity using IFN- γ ELISPOT assay with splenocytes purified from BCG immunized mice and 10 showed positive responses. Seven peptides were derived from MHC II and three from MHC class I. In particular, MHC class II binding peptides derived from the mycobacterial surface lipoprotein Mpt83 were highly antigenic. Further evaluations of these immunogenic BCG peptides may identify proteins useful as new TB vaccine candidates.


Subject(s)
Antigens, Bacterial , BCG Vaccine , Bacterial Proteins , Dendritic Cells , Mice, Inbred C57BL , Mycobacterium bovis , Animals , Antigens, Bacterial/immunology , Mycobacterium bovis/immunology , Mice , BCG Vaccine/immunology , Bacterial Proteins/immunology , Dendritic Cells/immunology , Vaccine Development , Female , Proteomics/methods , T-Lymphocytes/immunology , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class II/immunology , Lipoproteins/immunology , Tuberculosis/prevention & control , Tuberculosis/immunology , Peptides/immunology , Membrane Proteins
15.
J Immunother Cancer ; 12(9)2024 Sep 06.
Article in English | MEDLINE | ID: mdl-39244214

ABSTRACT

BACKGROUND: Immunodeficient mice engrafted with peripheral blood mononuclear cells (PBMCs) are models to study new cancer immunotherapy agents. However, this approach is associated with xenograft-versus-host disease (xGVHD), which starts early after PBMC transfer and limits the duration and interpretation of experiments. Here, we explore different approaches to overcome xGVHD and better support the development of cancer immunotherapies. METHODS: Immunodeficient NOD-scid IL2Rgnull (NSG) mice were intravenously transferred with human PBMCs and subcutaneously co-engrafted with HT29 human colon carcinoma cells. Diverse strategies to reduce xGVHD while preserving the antitumor activity of human immune cells were evaluated: (1) ex vivo immune graft modification by depleting CD4+ T cells pre-transfer using magnetic beads, (2) post-transplantation cyclophosphamide administration to eliminate proliferating xenoreactive T-cell clones and (3) using major histocompatibility complex (MHC) class I and II-deficient NSG mice: (Kb Db)null (IA)null (MHC-dKO NSG). Body weight and plasma murine alanine aminotransferase levels were measured as indicators of xGVHD and tumor size was measured every 2-3 days to monitor antitumor activity. The antitumor effects and pharmacodynamics of nivolumab plus ipilimumab and an anti-epithelial cell adhesion molecule (EpCAM)/CD3 T-cell engager (αEpCAM/CD3 bispecific antibody (BsAb)) were evaluated in the model. RESULTS: CD4+ T-cell depletion attenuates xGVHD but also abrogates the antitumor activity. Cyclophosphamide limits the antitumor response and does not substantially prevent xGVHD. In contrast, xGVHD was significantly attenuated in MHC-dKO NSG recipients, while the antitumor effect of human PBMCs was preserved. Furthermore, the administration of nivolumab plus ipilimumab caused exacerbated xGVHD in conventional NSG mice, thereby precluding the observation of their antitumor effects. Severe xGVHD did not occur in MHC-dKO NSG mice thus enabling the study of complete and durable tumor rejections. Similarly, NSG mice treated with an αEpCAM/CD3 BsAb showed complete tumor regressions, but died due to xGVHD. In contrast, MHC-dKO NSG mice on treatment with the αEpCAM/CD3 BsAb achieved complete tumor responses without severe xGVHD. A significant proportion of mice rendered tumor-free showed tumor rejection on rechallenge with HT29 cells without further treatment. Finally, tumor-infiltrating CD8+ T-cell number increase, activation and CD137 upregulation were observed on αEpCAM/CD3 BsAb treatment. CONCLUSION: Humanized MHC-dKO immunodeficient mice allow and refine the preclinical testing of immunotherapy agents for which experimentation is precluded in conventional immunodeficient mice due to severe xGVHD.


Subject(s)
Immune Checkpoint Inhibitors , Animals , Humans , Mice , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Mice, SCID , Mice, Inbred NOD , Histocompatibility Antigens Class I/immunology , Xenograft Model Antitumor Assays , Histocompatibility Antigens Class II/immunology , Graft vs Host Disease/drug therapy , Graft vs Host Disease/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
16.
MAbs ; 16(1): 2406788, 2024.
Article in English | MEDLINE | ID: mdl-39324549

ABSTRACT

Prenatal administration of monoclonal antibodies (mAbs) is a strategy that could be exploited to prevent viral infections during pregnancy and early life. To reach protective levels in fetuses, mAbs must be transported across the placenta, a selective barrier that actively and specifically promotes the transfer of antibodies (Abs) into the fetus through the neonatal Fc receptor (FcRn). Because FcRn also regulates Ab half-life, Fc mutations like the M428L/N434S, commonly known as LS mutations, and others have been developed to enhance binding affinity to FcRn and improve drug pharmacokinetics. We hypothesized that these FcRn-enhancing mutations could similarly affect the delivery of therapeutic Abs to the fetus. To test this hypothesis, we measured the transplacental transfer of leronlimab, an anti-CCR5 mAb, in clinical development for preventing HIV infections, using pregnant rhesus macaques to model in utero mAb transfer. We also generated a stabilized and FcRn-enhanced form of leronlimab, termed leronlimab-PLS. Leronlimab-PLS maintained higher levels within the maternal compartment while also reaching higher mAb levels in the fetus and newborn circulation. Further, a single dose of leronlimab-PLS led to complete CCR5 receptor occupancy in mothers and newborns for almost a month after birth. These findings support the optimization of FcRn interactions in mAb therapies designed for administration during pregnancy.


Subject(s)
Fetus , Histocompatibility Antigens Class I , Macaca mulatta , Receptors, CCR5 , Receptors, Fc , Animals , Pregnancy , Receptors, Fc/genetics , Receptors, Fc/immunology , Receptors, Fc/metabolism , Female , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Fetus/immunology , Receptors, CCR5/genetics , Receptors, CCR5/immunology , Animals, Newborn , Humans , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/genetics , HIV Infections/immunology , HIV Infections/drug therapy , HIV Infections/genetics , Maternal-Fetal Exchange/immunology , Mutation , HIV Antibodies/immunology , HIV Antibodies/genetics , CCR5 Receptor Antagonists/pharmacology , Antibodies, Monoclonal, Humanized/immunology
17.
Cells ; 13(18)2024 Sep 13.
Article in English | MEDLINE | ID: mdl-39329721

ABSTRACT

Impaired tumor cell antigen presentation contributes significantly to immune evasion. This study identifies Berbamine hydrochloride (Ber), a compound derived from traditional Chinese medicine, as an effective inhibitor of autophagy that enhances antigen presentation in tumor cells. Ber increases MHC-I-mediated antigen presentation in melanoma cells, improving recognition and elimination by CD8+ T cells. Mutation of Atg4b, which blocks autophagy, also raises MHC-I levels on the cell surface, and further treatment with Ber under these conditions does not increase MHC-I, indicating Ber's role in blocking autophagy to enhance MHC-I expression. Additionally, Ber treatment leads to the accumulation of autophagosomes, with elevated levels of LC3-II and p62, suggesting a disrupted autophagic flux. Fluorescence staining and co-localization analyses reveal that Ber likely inhibits lysosomal acidification without hindering autophagosome-lysosome fusion. Importantly, Ber treatment suppresses melanoma growth in mice and enhances CD8+ T cell infiltration, supporting its therapeutic potential. Our findings demonstrate that Ber disturbs late-stage autophagic flux through abnormal lysosomal acidification, enhancing MHC-I-mediated antigen presentation and curtailing tumor immune escape.


Subject(s)
Autophagy , Benzylisoquinolines , Melanoma , Tumor Escape , Autophagy/drug effects , Animals , Mice , Cell Line, Tumor , Humans , Tumor Escape/drug effects , Benzylisoquinolines/pharmacology , Benzylisoquinolines/therapeutic use , Melanoma/drug therapy , Melanoma/pathology , Melanoma/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , Antigen Presentation/drug effects , Histocompatibility Antigens Class I/metabolism , Histocompatibility Antigens Class I/immunology , Mice, Inbred C57BL , Autophagosomes/metabolism , Autophagosomes/drug effects , Lysosomes/metabolism , Lysosomes/drug effects , Autophagy-Related Proteins/metabolism , Autophagy-Related Proteins/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Melanoma, Experimental/drug therapy , Cysteine Endopeptidases
18.
Bioinformatics ; 40(9)2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39276157

ABSTRACT

MOTIVATION: Neoantigens, derived from somatic mutations in cancer cells, can elicit anti-tumor immune responses when presented to autologous T cells by human leukocyte antigen. Identifying immunogenic neoantigens is crucial for cancer immunotherapy development. However, the accuracy of current bioinformatic methods remains unsatisfactory. Surface and structural features of peptide-HLA class I (pHLA-I) complexes offer valuable insight into the immunogenicity of neoantigens. RESULTS: We present NeoaPred, a deep-learning framework for neoantigen prediction. NeoaPred accurately constructs pHLA-I complex structures, with 82.37% of the predicted structures showing an RMSD of < 1 Å. Using these structures, NeoaPred integrates differences in surface, structural, and atom group features between the mutant peptide and its wild-type counterpart to predict a foreignness score. This foreignness score is an effective factor for neoantigen prediction, achieving an AUROC (Area Under the Receiver Operating Characteristic Curve) of 0.81 and an AUPRC (Area Under the Precision-Recall Curve) of 0.54 in the test set, outperforming existing methods. AVAILABILITY AND IMPLEMENTATION: The source code is released under an Apache v2.0 license and is available at the GitHub repository (https://github.com/Dulab2020/NeoaPred).


Subject(s)
Antigens, Neoplasm , Deep Learning , Peptides , Humans , Antigens, Neoplasm/immunology , Antigens, Neoplasm/chemistry , Peptides/chemistry , Peptides/immunology , HLA Antigens/immunology , HLA Antigens/chemistry , Computational Biology/methods , Histocompatibility Antigens Class I/chemistry , Histocompatibility Antigens Class I/immunology , Software , Neoplasms/immunology
19.
BMC Bioinformatics ; 25(1): 310, 2024 Sep 27.
Article in English | MEDLINE | ID: mdl-39333860

ABSTRACT

BACKGROUND: Antigen presentation is a central step in initiating and shaping the adaptive immune response. To activate CD8+ T cells, pathogen-derived peptides are presented on the cell surface of antigen-presenting cells bound to major histocompatibility complex (MHC) class I molecules. CD8+ T cells that recognize these complexes with their T cell receptor are activated and ideally eliminate infected cells. Prediction of putative peptides binding to MHC class I (MHC-I) is crucial for understanding pathogen recognition in specific immune responses and for supporting drug and vaccine design. There are reliable databases for epitope prediction algorithms available however they primarily focus on the prediction of epitopes in single immunogenic proteins. RESULTS: We have developed the tool DiscovEpi to establish an interface between whole proteomes and epitope prediction. The tool allows the automated identification of all potential MHC-I-binding peptides within a proteome and calculates the epitope density and average binding score for every protein, a protein-centric approach. DiscovEpi provides a convenient interface between automated multiple sequence extraction by organism and cell compartment from the database UniProt for subsequent epitope prediction via NetMHCpan. Furthermore, it allows ranking of proteins by their predicted immunogenicity on the one hand and comparison of different proteomes on the other. By applying the tool, we predict a higher immunogenic potential of membrane-associated proteins of SARS-CoV-2 compared to those of influenza A based on the presented metrics epitope density and binding score. This could be confirmed visually by comparing the epitope maps of the influenza A strain and SARS-CoV-2. CONCLUSION: Automated prediction of whole proteomes and the subsequent visualization of the location of putative epitopes on sequence-level facilitate the search for putative immunogenic proteins or protein regions and support the study of adaptive immune responses and vaccine design.


Subject(s)
Histocompatibility Antigens Class I , Proteome , Proteome/metabolism , Proteome/immunology , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Histocompatibility Antigens Class I/chemistry , Humans , COVID-19/immunology , COVID-19/metabolism , COVID-19/virology , SARS-CoV-2/immunology , Software , Epitopes/chemistry , Epitopes/immunology , Databases, Protein , Algorithms
20.
Nat Commun ; 15(1): 7547, 2024 Aug 30.
Article in English | MEDLINE | ID: mdl-39214998

ABSTRACT

Most COVID-19 vaccines elicit immunity against the SARS-CoV-2 Spike protein. However, Spike protein mutations in emerging strains and immune evasion by the SARS-CoV-2 virus demonstrates the need to develop more broadly targeting vaccines. To facilitate this, we use mass spectrometry to identify immunopeptides derived from seven relatively conserved structural and non-structural SARS-CoV-2 proteins (N, E, Nsp1/4/5/8/9). We use two different B-lymphoblastoid cell lines to map Human Leukocyte Antigen (HLA) class I and class II immunopeptidomes covering some of the prevalent HLA types across the global human population. We employ DNA plasmid transfection and direct antigen delivery approaches to sample different antigens and find 248 unique HLA class I and HLA class II bound peptides with 71 derived from N, 12 from E, 28 from Nsp1, 19 from Nsp4, 73 from Nsp8 and 45 peptides derived from Nsp9. Over half of the viral peptides are unpublished. T cell reactivity tested against 56 of the detected peptides shows CD8+ and CD4+ T cell responses against several peptides from the N, E, and Nsp9 proteins. Results from this study will aid the development of next-generation COVID vaccines targeting epitopes from across a number of SARS-CoV-2 proteins.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , SARS-CoV-2/immunology , SARS-CoV-2/genetics , COVID-19/immunology , COVID-19/virology , Haplotypes , Peptides/immunology , Peptides/chemistry , Epitopes, T-Lymphocyte/immunology , HLA Antigens/immunology , HLA Antigens/genetics , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/genetics , Histocompatibility Antigens Class II/immunology , COVID-19 Vaccines/immunology , Histocompatibility Antigens Class I/immunology , CD8-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Antigens, Viral/immunology , Antigens, Viral/genetics , Cell Line
SELECTION OF CITATIONS
SEARCH DETAIL