Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 2.503
1.
Proc Natl Acad Sci U S A ; 121(22): e2319029121, 2024 May 28.
Article En | MEDLINE | ID: mdl-38781214

The HapImmuneTM platform exploits covalent inhibitors as haptens for creating major histocompatibility complex (MHC)-presented tumor-specific neoantigens by design, combining targeted therapies with immunotherapy for the treatment of drug-resistant cancers. A HapImmune antibody, R023, recognizes multiple sotorasib-conjugated KRAS(G12C) peptides presented by different human leukocyte antigens (HLAs). This high specificity to sotorasib, coupled with broad HLA-binding capability, enables such antibodies, when reformatted as T cell engagers, to potently and selectively kill sotorasib-resistant KRAS(G12C) cancer cells expressing different HLAs upon sotorasib treatment. The loosening of HLA restriction could increase the patient population that can benefit from this therapeutic approach. To understand the molecular basis for its unconventional binding capability, we used single-particle cryogenic electron microscopy to determine the structures of R023 bound to multiple sotorasib-peptide conjugates presented by different HLAs. R023 forms a pocket for sotorasib between the VH and VL domains, binds HLAs in an unconventional, angled way, with VL making most contacts with them, and makes few contacts with the peptide moieties. This binding mode enables the antibody to accommodate different hapten-peptide conjugates and to adjust its conformation to different HLAs presenting hapten-peptides. Deep mutational scanning validated the structures and revealed distinct levels of mutation tolerance by sotorasib- and HLA-binding residues. Together, our structural information and sequence landscape analysis reveal key features for achieving MHC-restricted recognition of multiple hapten-peptide antigens, which will inform the development of next-generation therapeutic antibodies.


Peptides , Humans , Peptides/immunology , Peptides/chemistry , HLA Antigens/immunology , HLA Antigens/metabolism , Major Histocompatibility Complex/immunology , Haptens/immunology , Protein Binding , Cryoelectron Microscopy
2.
Adv Protein Chem Struct Biol ; 140: 327-345, 2024.
Article En | MEDLINE | ID: mdl-38762273

Major histocompatibility complex (MHC) tetramers stand as formidable tools within T cell biology, facilitating the exploration and comprehension of immune responses. These artificial molecules, comprising four bound MHC molecules, typically with a specified peptide and a fluorescent label, play a pivotal role in characterizing T cell subsets, monitoring clonal expansion, and unraveling T cell dynamics during responses to infections or immunotherapies. Beyond their applications in T cell biology, MHC tetramers prove valuable in investigating a spectrum of diseases such as infectious diseases, autoimmune disorders, and cancers. Their instrumental role extends to vaccine research and development. Notably, when appropriately configured, tetramers transcend T cell biology research and find utility in exploring natural killer T cells and contributing to specific T cell clonal deletions.


Major Histocompatibility Complex , Humans , Major Histocompatibility Complex/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Neoplasms/immunology , Neoplasms/therapy , Neoplasms/metabolism , Animals , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism
3.
Pharmacol Res ; 203: 107168, 2024 May.
Article En | MEDLINE | ID: mdl-38583689

Parkinson's disease (PD) is a common neurodegenerative disease characterized by progressive loss of dopaminergic neurons in the substantia nigra and the aggregation of alpha-synuclein (α-syn). The central nervous system (CNS) has previously been considered as an immune-privileged area. However, studies have shown that the immune responses are involved in PD. The major histocompatibility complex (MHC) presents antigens from antigen-presenting cells (APCs) to T lymphocytes, immune responses will be induced. MHCs are expressed in microglia, astrocytes, and dopaminergic neurons. Single nucleotide polymorphisms in MHC are related to the risk of PD. The aggregated α-syn triggers the expression of MHCs by activating glia cells. CD4+ and CD8+ T lymphocytes responses and microglia activation are detected in brains of PD patients. In addiction immune responses further increase blood-brain barrier (BBB) permeability and T cell infiltration in PD. Thus, MHCs are involved in PD through participating in immune and inflammatory responses.


Major Histocompatibility Complex , Parkinson Disease , Humans , Parkinson Disease/immunology , Parkinson Disease/genetics , Animals , Major Histocompatibility Complex/immunology , alpha-Synuclein/immunology , alpha-Synuclein/metabolism , Blood-Brain Barrier/immunology , Blood-Brain Barrier/metabolism , Microglia/immunology , Microglia/metabolism
4.
Nature ; 618(7967): 1033-1040, 2023 Jun.
Article En | MEDLINE | ID: mdl-37316667

Most clinically applied cancer immunotherapies rely on the ability of CD8+ cytolytic T cells to directly recognize and kill tumour cells1-3. These strategies are limited by the emergence of major histocompatibility complex (MHC)-deficient tumour cells and the formation of an immunosuppressive tumour microenvironment4-6. The ability of CD4+ effector cells to contribute to antitumour immunity independently of CD8+ T cells is increasingly recognized, but strategies to unleash their full potential remain to be identified7-10. Here, we describe a mechanism whereby a small number of CD4+ T cells is sufficient to eradicate MHC-deficient tumours that escape direct CD8+ T cell targeting. The CD4+ effector T cells preferentially cluster at tumour invasive margins where they interact with MHC-II+CD11c+ antigen-presenting cells. We show that T helper type 1 cell-directed CD4+ T cells and innate immune stimulation reprogramme the tumour-associated myeloid cell network towards interferon-activated antigen-presenting and iNOS-expressing tumouricidal effector phenotypes. Together, CD4+ T cells and tumouricidal myeloid cells orchestrate the induction of remote inflammatory cell death that indirectly eradicates interferon-unresponsive and MHC-deficient tumours. These results warrant the clinical exploitation of this ability of CD4+ T cells and innate immune stimulators in a strategy to complement the direct cytolytic activity of CD8+ T cells and natural killer cells and advance cancer immunotherapies.


CD4-Positive T-Lymphocytes , Cell Death , Immunotherapy , Inflammation , Neoplasms , Tumor Microenvironment , Humans , Antigen-Presenting Cells/immunology , CD11c Antigen/immunology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Death/immunology , Histocompatibility Antigens Class II/immunology , Immunity, Innate , Inflammation/immunology , Interferons/immunology , Major Histocompatibility Complex/immunology , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/therapy , Tumor Microenvironment/immunology , Immunotherapy/methods , Killer Cells, Natural/immunology , Myeloid Cells/immunology , Th1 Cells/cytology , Th1 Cells/immunology
5.
Mol Immunol ; 142: 105-119, 2022 02.
Article En | MEDLINE | ID: mdl-34973498

In the late 1980s and early 1990s, the hunt for a transporter molecule ostensibly responsible for the translocation of peptides across the endoplasmic reticulum (ER) membrane yielded the successful discovery of transporter associated with antigen processing (TAP) protein. TAP is a heterodimer complex comprised of TAP1 and TAP2, which utilizes ATP to transport cytosolic peptides into the ER across its membrane. In the ER, together with other components it forms the peptide loading complex (PLC), which directs loading of high affinity peptides onto nascent major histocompatibility complex class I (MHC-I) molecules that are then transported to the cell surface for presentation to CD8+ T cells. TAP also plays a crucial role in transporting peptides into phagosomes and endosomes during cross-presentation in dendritic cells (DCs). Because of the critical role that TAP plays in both classical MHC-I presentation and cross-presentation, its expression and function are often compromised by numerous types of cancers and viruses to evade recognition by cytotoxic CD8 T cells. Here we review the discovery and function of TAP with a major focus on its role in cross-presentation in DCs. We discuss a recently described emergency route of noncanonical cross-presentation that is mobilized in DCs upon TAP blockade to restore CD8 T cell cross-priming. We also discuss the various strategies employed by cancer cells and viruses to target TAP expression or function to evade immunosurveillance - along with some strategies by which the repertoire of peptides presented by cells which downregulate TAP can be targeted as a therapeutic strategy to mobilize a TAP-independent CD8 T cell response. Lastly, we discuss TAP polymorphisms and the role of TAP in inherited disorders.


ATP Binding Cassette Transporter, Subfamily B, Member 2/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 3/metabolism , ATP-Binding Cassette Transporters/metabolism , Antigen Presentation/immunology , Cross-Priming/immunology , Tumor Escape/immunology , ATP Binding Cassette Transporter, Subfamily B, Member 2/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 3/genetics , ATP-Binding Cassette Transporters/genetics , Dendritic Cells/immunology , Endoplasmic Reticulum/metabolism , Humans , Major Histocompatibility Complex/immunology , Neoplasms/immunology , Protein Transport/genetics , T-Lymphocytes, Cytotoxic/immunology , Viruses/immunology
6.
J Biol Chem ; 298(2): 101542, 2022 02.
Article En | MEDLINE | ID: mdl-34968463

The monomorphic antigen-presenting molecule major histocompatibility complex-I-related protein 1 (MR1) presents small-molecule metabolites to mucosal-associated invariant T (MAIT) cells. The MR1-MAIT cell axis has been implicated in a variety of infectious and noncommunicable diseases, and recent studies have begun to develop an understanding of the molecular mechanisms underlying this specialized antigen presentation pathway. However, proteins regulating MR1 folding, loading, stability, and surface expression remain to be identified. Here, we performed a gene trap screen to discover novel modulators of MR1 surface expression through insertional mutagenesis of an MR1-overexpressing clone derived from the near-haploid human cell line HAP1 (HAP1.MR1). The most significant positive regulators identified included ß2-microglobulin, a known regulator of MR1 surface expression, and ATP13A1, a P5-type ATPase in the endoplasmic reticulum (ER) not previously known to be associated with MR1-mediated antigen presentation. CRISPR/Cas9-mediated knockout of ATP13A1 in both HAP1.MR1 and THP-1 cell lines revealed a profound reduction in MR1 protein levels and a concomitant functional defect specific to MR1-mediated antigen presentation. Collectively, these data are consistent with the ER-resident ATP13A1 being a key posttranscriptional determinant of MR1 surface expression.


Antigen Presentation , Histocompatibility Antigens Class I , Major Histocompatibility Complex , Minor Histocompatibility Antigens , P-type ATPases , Histocompatibility Antigens Class I/metabolism , Humans , Major Histocompatibility Complex/immunology , Minor Histocompatibility Antigens/immunology , P-type ATPases/immunology
7.
Sci Rep ; 11(1): 19862, 2021 10 06.
Article En | MEDLINE | ID: mdl-34615944

Individual variation in the age of pubertal onset is linked to physical and mental health, yet the factors underlying this variation are poorly understood. Life history theory predicts that individuals at higher risk of mortality due to extrinsic causes such as infectious disease should sexually mature and reproduce earlier, whereas those at lower risk can delay puberty and continue to invest resources in somatic growth. We examined relationships between a genetic predictor of infectious disease resistance, heterozygosity of the major histocompatibility complex (MHC), referred to as the human leukocyte antigen (HLA) gene in humans, and self-reported pubertal timing. In a combined sample of men from Canada (n = 137) and the United States (n = 43), MHC heterozygosity predicted later self-reported pubertal development. These findings suggest a genetic trade-off between immunocompetence and sexual maturation in human males.


Alleles , Heterozygote , Major Histocompatibility Complex/genetics , Puberty/genetics , Adolescent , Adult , Disease Resistance/genetics , Disease Susceptibility/immunology , Genetic Predisposition to Disease , Humans , Major Histocompatibility Complex/immunology , Male , Puberty/immunology , Self Report , Young Adult
8.
Front Immunol ; 12: 742881, 2021.
Article En | MEDLINE | ID: mdl-34650566

Despite the high number of individuals infected by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) who develop coronavirus disease 2019 (COVID-19) symptoms worldwide, many exposed individuals remain asymptomatic and/or uninfected and seronegative. This could be explained by a combination of environmental (exposure), immunological (previous infection), epigenetic, and genetic factors. Aiming to identify genetic factors involved in immune response in symptomatic COVID-19 as compared to asymptomatic exposed individuals, we analyzed 83 Brazilian couples where one individual was infected and symptomatic while the partner remained asymptomatic and serum-negative for at least 6 months despite sharing the same bedroom during the infection. We refer to these as "discordant couples". We performed whole-exome sequencing followed by a state-of-the-art method to call genotypes and haplotypes across the highly polymorphic major histocompatibility complex (MHC) region. The discordant partners had comparable ages and genetic ancestry, but women were overrepresented (65%) in the asymptomatic group. In the antigen-presentation pathway, we observed an association between HLA-DRB1 alleles encoding Lys at residue 71 (mostly DRB1*03:01 and DRB1*04:01) and DOB*01:02 with symptomatic infections and HLA-A alleles encoding 144Q/151R with asymptomatic seronegative women. Among the genes related to immune modulation, we detected variants in MICA and MICB associated with symptomatic infections. These variants are related to higher expression of soluble MICA and low expression of MICB. Thus, quantitative differences in these molecules that modulate natural killer (NK) activity could contribute to susceptibility to COVID-19 by downregulating NK cell cytotoxic activity in infected individuals but not in the asymptomatic partners.


Asymptomatic Infections , COVID-19 , Histocompatibility Antigens , Major Histocompatibility Complex , SARS-CoV-2 , Adult , Aged , Brazil , COVID-19/genetics , COVID-19/immunology , Female , Genetic Predisposition to Disease , Genotype , Histocompatibility Antigens/genetics , Histocompatibility Antigens/immunology , Humans , Major Histocompatibility Complex/genetics , Major Histocompatibility Complex/immunology , Male , Middle Aged , Exome Sequencing
9.
Sci Rep ; 11(1): 19056, 2021 09 24.
Article En | MEDLINE | ID: mdl-34561502

Hairy cell leukaemia (HCL) is a rare CD20+ B cell malignancy characterised by rare "hairy" B cells and extensive bone marrow (BM) infiltration. Frontline treatment with the purine analogue cladribine (CDA) results in a highly variable response duration. We hypothesised that analysis of the BM tumour microenvironment would identify prognostic biomarkers of response to CDA. HCL BM immunology pre and post CDA treatment and healthy controls were analysed using Digital Spatial Profiling to assess the expression of 57 proteins using an immunology panel. A bioinformatics pipeline was developed to accommodate the more complex experimental design of a spatially resolved study. Treatment with CDA was associated with the reduction in expression of HCL tumour markers (CD20, CD11c) and increased expression of myeloid markers (CD14, CD68, CD66b, ARG1). Expression of HLA-DR, STING, CTLA4, VISTA, OX40L were dysregulated pre- and post-CDA. Duration of response to treatment was associated with greater reduction in tumour burden and infiltration by CD8 T cells into the BM post-CDA. This is the first study to provide a high multiplex analysis of HCL BM microenvironment demonstrating significant immune dysregulation and identify biomarkers of response to CDA. With validation in future studies, prospective application of these biomarkers could allow early identification and increased monitoring in patients at increased relapse risk post CDA.


Biomarkers, Tumor/metabolism , Leukemia, Hairy Cell/pathology , Tumor Microenvironment , Adult , Case-Control Studies , Female , Humans , Leukemia, Hairy Cell/immunology , Leukemia, Hairy Cell/metabolism , Major Histocompatibility Complex/immunology , Male , Middle Aged , Prognosis
10.
Sci Rep ; 11(1): 17234, 2021 08 26.
Article En | MEDLINE | ID: mdl-34446788

Over the past two decades, there has been a great interest in the study of HLA-E-restricted αß T cells during bacterial and viral infections, including recently SARS-CoV-2 infection. Phenotyping of these specific HLA-E-restricted T cells requires new tools such as tetramers for rapid cell staining or sorting, as well as for the identification of new peptides capable to bind to the HLA-E pocket. To this aim, we have developed an optimal photosensitive peptide to generate stable HLA-E/pUV complexes allowing high-throughput production of new HLA-E/peptide complexes by peptide exchange. We characterized the UV exchange by ELISA and improved the peptide exchange readout using size exclusion chromatography. This novel approach for complex quantification is indeed very important to perform tetramerization of MHC/peptide complexes with the high quality required for detection of specific T cells. Our approach allows the rapid screening of peptides capable of binding to the non-classical human HLA-E allele, paving the way for the development of new therapeutic approaches based on the detection of HLA-E-restricted T cells.


Epitopes, T-Lymphocyte/chemistry , Histocompatibility Antigens Class I/chemistry , Major Histocompatibility Complex/immunology , Peptides/chemistry , Amino Acid Sequence , Epitopes, T-Lymphocyte/immunology , High-Throughput Screening Assays , Histocompatibility Antigens Class I/immunology , Humans , Immunologic Techniques , Photochemical Processes , Protein Binding , Protein Conformation , T-Lymphocytes, Cytotoxic/immunology , HLA-E Antigens
11.
Mol Immunol ; 139: 76-86, 2021 11.
Article En | MEDLINE | ID: mdl-34455212

The activation of T cells is triggered by the interactions of T cell receptors (TCRs) with their epitopes, which are peptides presented by major histocompatibility complex (MHC) on the surfaces of antigen presenting cells (APC). While each TCR can only recognize a specific subset from a large repertoire of peptide-MHC (pMHC) complexes, it is very often that peptides in this subset share little sequence similarity. This is known as the specificity and cross-reactivity of T cells, respectively. The binding affinities between different types of TCRs and pMHC are the major driving force to shape this specificity and cross-reactivity in T cell recognition. The binding affinities, furthermore, are determined by the sequence and structural properties at the interfaces between TCRs and pMHC. Fortunately, a wealth of data on binding and structures of TCR-pMHC interactions becomes publicly accessible in online resources, which offers us the opportunity to develop a random forest classifier for predicting the binding affinities between TCR and pMHC based on the structure of their complexes. Specifically, the structure and sequence of a given complex were projected onto a high-dimensional feature space as the input of the classifier, which was then trained by a large-scale benchmark dataset. Based on the cross-validation results, we found that our machine learning model can predict if the binding affinity of a given TCR-pMHC complex is stronger or weaker than a predefined threshold with an overall accuracy approximately around 75 %. The significance of our prediction was estimated by statistical analysis. Moreover, more than 60 % of binding affinities in the ATLAS database can be successfully classified into groups within the range of 2 kcal/mol. Additionally, we show that TCR-pMHC complexes with strong binding affinity prefer hydrophobic interactions between amino acids with large aromatic rings instead of electrostatic interactions. Our results therefore provide insights to design engineered TCRs which enhance the specificity for their targeted epitopes. Taken together, this method can serve as a useful addition to a suite of existing approaches which study binding between TCR and pMHC.


Lymphocyte Activation/immunology , Machine Learning , Major Histocompatibility Complex/immunology , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Humans , Protein Binding
12.
Nat Methods ; 18(8): 881-892, 2021 08.
Article En | MEDLINE | ID: mdl-34282327

T cells express T cell receptors (TCRs) composed of somatically recombined TCRα and TCRß chains, which mediate recognition of major histocompatibility complex (MHC)-antigen complexes and drive the antigen-specific adaptive immune response to pathogens and cancer. The TCR repertoire in each individual is highly diverse, which allows for recognition of a wide array of foreign antigens, but also presents a challenge in analyzing this response using conventional methods. Recent studies have developed high-throughput sequencing technologies to identify TCR sequences, analyze their antigen specificities using experimental and computational tools, and pair TCRs with transcriptional and epigenetic cell state phenotypes in single cells. In this Review, we highlight these technological advances and describe how they have been applied to discover fundamental insights into T cell-mediated immunity.


Antigens/immunology , High-Throughput Nucleotide Sequencing/methods , Major Histocompatibility Complex/immunology , Receptors, Antigen, T-Cell/immunology , Single-Cell Analysis/methods , T-Lymphocytes/immunology , Adaptive Immunity , Animals , Humans
13.
Nat Commun ; 12(1): 2746, 2021 05 12.
Article En | MEDLINE | ID: mdl-33980853

Despite the clinical success of blocking its interactions, how PD-1 inhibits T-cell activation is incompletely understood, as exemplified by its potency far exceeding what might be predicted from its affinity for PD-1 ligand-1 (PD-L1). This may be partially attributed to PD-1's targeting the proximal signaling of the T-cell receptor (TCR) and co-stimulatory receptor CD28 via activating Src homology region 2 domain-containing phosphatases (SHPs). Here, we report PD-1 signaling regulates the initial TCR antigen recognition manifested in a smaller spreading area, fewer molecular bonds formed, and shorter bond lifetime of T cell interaction with peptide-major histocompatibility complex (pMHC) in the presence than absence of PD-L1 in a manner dependent on SHPs and Leukocyte C-terminal Src kinase. Our results identify a PD-1 inhibitory mechanism that disrupts the cooperative TCR-pMHC-CD8 trimolecular interaction, which prevents CD8 from augmenting antigen recognition, explaining PD-1's potent inhibitory function and its value as a target for clinical intervention.


CD8 Antigens/immunology , Programmed Cell Death 1 Receptor/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , Animals , B7-H1 Antigen/immunology , CD8 Antigens/metabolism , Calcium/metabolism , Humans , Lymphocyte Activation , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Major Histocompatibility Complex/immunology , Mice , Mice, Transgenic , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/metabolism , Protein Binding , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , T-Lymphocytes/metabolism
14.
Mol Cells ; 44(5): 328-334, 2021 May 31.
Article En | MEDLINE | ID: mdl-33972472

The advent of the major histocompatibility complex (MHC) multimer technology has led to a breakthrough in the quantification and analysis of antigen-specific T cells. In particular, this technology has dramatically advanced the measurement and analysis of CD8 T cells and is being applied more widely. In addition, the scope of application of MHC multimer technology is gradually expanding to other T cells such as CD4 T cells, natural killer T cells, and mucosal-associated invariant T cells. MHC multimer technology acts by complementing the T-cell receptor-MHC/peptide complex affinity, which is relatively low compared to antigen-antibody affinity, through a multivalent interaction. The application of MHC multimer technology has expanded to include various functions such as quantification and analysis of antigen-specific T cells, cell sorting, depletion, stimulation to replace antigen-presenting cells, and single-cell classification through DNA barcodes. This review aims to provide the latest knowledge of MHC multimer technology, which is constantly evolving, broaden understanding of this technology, and promote its widespread use.


Allergy and Immunology/standards , Major Histocompatibility Complex/immunology , Humans
15.
Front Immunol ; 12: 664624, 2021.
Article En | MEDLINE | ID: mdl-33995395

Melanized focal changes in white skeletal muscle of farmed Atlantic salmon, "black spots", is a quality problem affecting on average 20% of slaughtered fish. The spots appear initially as "red spots" characterized by hemorrhages and acute inflammation and progress into black spots characterized by chronic inflammation and abundant pigmented cells. Piscine orthoreovirus 1 (PRV-1) was previously found to be associated with macrophages and melano-macrophages in red and black spots. Here we have addressed the inflammatory microenvironment of red and black spots by studying the polarization status of the macrophages and cell mediated immune responses in spots, in both PRV-1 infected and non-infected fish. Samples that had been collected at regular intervals through the seawater production phase in a commercial farm were analyzed by multiplex fluorescent in situ hybridization (FISH) and RT-qPCR methods. Detection of abundant inducible nitric oxide synthase (iNOS2) expressing M1-polarized macrophages in red spots demonstrated a pro-inflammatory microenvironment. There was an almost perfect co-localization with the iNOS2 expression and PRV-1 infection. Black spots, on the other side, had few iNOS2 expressing cells, but a relatively high number of arginase-2 expressing anti-inflammatory M2-polarized macrophages containing melanin. The numerous M2-polarized melano-macrophages in black spots indicate an ongoing healing phase. Co-localization of PRV-1 and cells expressing CD8+ and MHC-I suggests a targeted immune response taking place in the spots. Altogether, this study indicates that PRV-1 induces a pro-inflammatory environment that is important for the pathogenesis of the spots. We do not have indication that infection of PRV-1 is the initial causative agent of this condition.


Cellular Microenvironment , Fish Diseases/etiology , Fish Diseases/metabolism , Macrophages/immunology , Macrophages/virology , Orthoreovirus/physiology , Reoviridae Infections/veterinary , Salmo salar , Animals , Biomarkers , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Fish Diseases/pathology , Fluorescent Antibody Technique , Immunohistochemistry , Macrophage Activation/genetics , Macrophage Activation/immunology , Macrophages/pathology , Major Histocompatibility Complex/genetics , Major Histocompatibility Complex/immunology
16.
Cells ; 10(5)2021 04 26.
Article En | MEDLINE | ID: mdl-33926103

T cells are sensitive to 1 to 10 foreign-peptide-MHC complexes among a vast majority of self-peptide-MHC complexes, and discriminate selectively between peptide-MHC complexes that differ not much in their binding affinity to T-cell receptors (TCRs). Quantitative models that aim to explain this sensitivity and selectivity largely focus on single TCR/peptide-MHC complexes, but T cell adhesion involves a multitude of different complexes. In this article, we demonstrate in a three-dimensional computational model of T-cell adhesion that the cooperative stabilization of close-contact zones is sensitive to one to three foreign-peptide-MHC complexes and occurs at a rather sharp threshold affinity of these complexes, which implies selectivity. In these close-contact zones with lateral extensions of hundred to several hundred nanometers, few TCR/foreign-peptide-MHC complexes and many TCR/self-peptide-MHC complexes are segregated from LFA-1/ICAM-1 complexes that form at larger membrane separations. Previous high-resolution microscopy experiments indicate that the sensitivity and selectivity in the formation of closed-contact zones reported here are relevant for T-cell recognition, because the stabilization of close-contact zones by foreign, agonist peptide-MHC complexes precedes T-cell signaling and activation in the experiments.


Major Histocompatibility Complex/immunology , T-Lymphocytes/cytology , Cell Adhesion , Cell Membrane/metabolism , Computer Simulation , Diffusion , Humans , Lymphocyte Activation , Models, Theoretical , Peptides/chemistry , Probability , Protein Binding , Receptors, Antigen, T-Cell/metabolism , Thermodynamics
17.
Drug Deliv ; 28(1): 752-766, 2021 Dec.
Article En | MEDLINE | ID: mdl-33860719

Macrophages are a major component of the immunoresponse. Diversity and plasticity are two of the hallmarks of macrophages, which allow them to act as proinflammatory, anti-inflammatory, and homeostatic agents. Research has found that cancer and many inflammatory or autoimmune disorders are correlated with activation and tissue infiltration of macrophages. Recent developments in macrophage nanomedicine-based disease treatment are proving to be timely owing to the increasing inadequacy of traditional treatment. Here, we review the role of macrophages in nanomedicine-based disease treatment. First, we present a brief background on macrophages and nanomedicine. Then, we delve into applications of macrophages as a target for disease treatment and delivery systems and summarize the applications of macrophage-derived extracellular vesicles. Finally, we provide an outlook on the clinical utility of macrophages in nanomedicine-based disease treatment.


Drug Delivery Systems/methods , Macrophages/physiology , Nanomedicine/methods , Biological Availability , Chemistry, Pharmaceutical , Extracellular Vesicles/metabolism , Inflammation Mediators/metabolism , Major Histocompatibility Complex/immunology , Neoplasms/drug therapy , Neoplasms/immunology
18.
STAR Protoc ; 2(1): 100385, 2021 03 19.
Article En | MEDLINE | ID: mdl-33778779

Major histocompatibility complexes (MHC) play a critical role in immunity by presenting peptides on the cell surface for T cell recognition. Identification of these peptides can be valuable to develop vaccines or immunotherapeutic strategies for infectious diseases and cancers. Mass spectrometry is the only tool available for unbiased identification of the immunopeptidome. Here, we describe a protocol for purification and identification of MHC class I peptides, including in-house purification of anti-MHC-antibody from hybridoma cells and the LC-MS/MS analysis of MHC-I bound peptides.


Histocompatibility Antigens Class I/isolation & purification , Peptides/immunology , Peptides/isolation & purification , Cell Line , Cell Membrane/immunology , Chromatography, Liquid/methods , HLA Antigens/immunology , HLA Antigens/isolation & purification , Histocompatibility Antigens Class I/chemistry , Histocompatibility Antigens Class I/immunology , Humans , Major Histocompatibility Complex/immunology , Peptides/chemistry , Proteins/isolation & purification , Proteomics/methods , Tandem Mass Spectrometry/methods
19.
Comput Biol Chem ; 92: 107459, 2021 Jun.
Article En | MEDLINE | ID: mdl-33636637

Zika virus (ZIKV) infection is a global health concern due to its association with microcephaly and neurological complications. The development of a T-cell vaccine is important to combat this disease. In this study, we propose ZIKV major histocompatibility complex I (MHC-I) epitopes based on in silico screening consensus followed by molecular docking, PRODIGY, and molecular dynamics (MD) simulation analyses. The effects of the reported mutations on peptide-MHC-I (pMHC-I) complexes were also evaluated. In general, our data indicate an allele-specific peptide-binding human leukocyte antigen (HLA) and potential epitopes. For HLA-B44, we showed that the absence of acidic residue Glu at P2, due to the loss of the electrostatic interaction with Lys45, has a negative impact on the pMHC-I complex stability and explains the low free energy estimated for the immunodominant peptide E-4 (IGVSNRDFV). Our MD data also suggest the deleterious effects of acidic residue Asp at P1 on the pMHC-I stability of HLA-B8 due to destabilization of the α-helix and ß-strand. Free energy estimation further indicated that the mutation from Val to Ala at P9 of peptide E-247 (DAHAKRQTV), which was found exclusively in microcephaly samples, did not reduce HLA-B8 affinity. In contrast, the mutation from Thr to Pro at P2 of the peptide NS5-832 (VTKWTDIPY) decreased the interaction energy, number of intermolecular interactions, and adversely affected its binding mode with HLA-A1. Overall, our findings are important with regard to the design of T-cell peptide vaccines and for understanding how ZIKV escapes recognition by CD8 + T-cells.


Epitopes/immunology , Major Histocompatibility Complex/immunology , Molecular Dynamics Simulation , Peptides/immunology , Viral Proteins/genetics , Zika Virus/chemistry , Alleles , Epitopes/genetics , Major Histocompatibility Complex/genetics , Mutation , Peptides/genetics , Viral Proteins/immunology , Zika Virus/immunology
20.
Biol Chem ; 402(6): 693-702, 2021 05 26.
Article En | MEDLINE | ID: mdl-33544464

Mesenchymal stem cells (MSCs) play an important role in tissue engineering applications aiming at the regeneration or substitution of damaged tissues. In this context, off-the-shelf allogeneic MSCs would represent an attractive universal cell source. However, immune rejection is a major limitation for the clinical use of allogeneic MSCs. Immune rejection is mediated by the expression of major histocompatibility complexes (MHC)-I and -II on the donor cells. In this study, we eliminated MHC-I and/or MHC-II expression in human MSCs by using the CRISPR/Cas9 technology and investigated the effect of the individual or combined knockout of MHC-I and MHC-II on MSC survival after transplantation into immunocompetent mice. Elimination of MHC-I and/or MHC-II expression did not affect mesenchymal marker gene expression, viability, proliferation and the differentiation potential of MSCs in vitro. However, cell survival of transplanted MSCs was significantly elevated in MHC-I and MHC-II deficient MSCs. A direct side-by-side comparison does not reveal any significant difference in the immunogenicity of MHC-I and MHC-II knockout MSCs. Moreover, double knockout of MHC-I and MHC-II did not further increase in vivo cell survival of transplanted MSCs. Our results demonstrate that knockout of MHC-I and/or MHC-II represents an effective strategy to prevent immune rejection of allogeneic MSCs.


Major Histocompatibility Complex/immunology , Mesenchymal Stem Cells/immunology , CRISPR-Cas Systems/genetics , CRISPR-Cas Systems/immunology , Cell Proliferation , Cell Survival , Cells, Cultured , Flow Cytometry , Gene Editing , Humans , Major Histocompatibility Complex/genetics , Mesenchymal Stem Cells/cytology
...