1
|
Mi X, Li S, Ye Z, Dai Z, Ding B, Sun B, Shen Y, Xiao Z. LRMAHpan: a novel tool for multi-allelic HLA presentation prediction using Resnet-based and LSTM-based neural networks. Front Immunol 2024; 15:1478201. [PMID: 39669561 PMCID: PMC11634944 DOI: 10.3389/fimmu.2024.1478201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 10/30/2024] [Indexed: 12/14/2024] Open
Abstract
Introduction The identification of peptides eluted from HLA complexes by mass spectrometry (MS) can provide critical data for deep learning models of antigen presentation prediction and promote neoantigen vaccine design. A major challenge remains in determining which HLA allele eluted peptides correspond to. Methods To address this, we present a tool for prediction of multiple allele (MA) presentation called LRMAHpan, which integrates LSTM network and ResNet_CA network for antigen processing and presentation prediction. We trained and tested the LRMAHpan BA (binding affinity) and the LRMAHpan AP (antigen processing) models using mass spectrometry data, subsequently combined them into the LRMAHpan PS (presentation score) model. Our approach is based on a novel pHLA encoding method that enables the integration of neoantigen prediction tasks into computer vision methods. This method aggregates MA data into a multichannel matrix and incorporates peptide sequences to efficiently capture binding signals. Results LRMAHpan outperforms standard predictors such as NetMHCpan 4.1, MHCflurry 2.0, and TransPHLA in terms of positive predictive value (PPV) when applied to MA data. Additionally, it can accommodate peptides of variable lengths and predict HLA class I and II presentation. We also predicted neoantigens in a cohort of metastatic melanoma patients, identifying several shared neoantigens. Discussion Our results demonstrate that LRMAHpan significantly improves the accuracy of antigen presentation predictions.
Collapse
Affiliation(s)
- Xue Mi
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Shaohao Li
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Zheng Ye
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Zhu Dai
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Bo Ding
- Department of Obstetrics and Gynecoloty, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Bo Sun
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Yang Shen
- Department of Obstetrics and Gynecoloty, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
- Jiangsu Sports Health Research Institute, Institute of Sports and Health, Nanjing, China
| | - Zhongdang Xiao
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
- Jiangsu Sports Health Research Institute, Institute of Sports and Health, Nanjing, China
| |
Collapse
|
2
|
Krull KK, Ali SA, Krijgsveld J. Enhanced feature matching in single-cell proteomics characterizes IFN-γ response and co-existence of cell states. Nat Commun 2024; 15:8262. [PMID: 39327420 PMCID: PMC11427561 DOI: 10.1038/s41467-024-52605-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 09/17/2024] [Indexed: 09/28/2024] Open
Abstract
Proteome analysis by data-independent acquisition (DIA) has become a powerful approach to obtain deep proteome coverage, and has gained recent traction for label-free analysis of single cells. However, optimal experimental design for DIA-based single-cell proteomics has not been fully explored, and performance metrics of subsequent data analysis tools remain to be evaluated. Therefore, we here formalize and comprehensively evaluate a DIA data analysis strategy that exploits the co-analysis of low-input samples with a so-called matching enhancer (ME) of higher input, to increase sensitivity, proteome coverage, and data completeness. We assess the matching specificity of DIA-ME by a two-proteome model, and demonstrate that false discovery and false transfer are maintained at low levels when using DIA-NN software, while preserving quantification accuracy. We apply DIA-ME to investigate the proteome response of U-2 OS cells to interferon gamma (IFN-γ) in single cells, and recapitulate the time-resolved induction of IFN-γ response proteins as observed in bulk material. Moreover, we uncover co- and anti-correlating patterns of protein expression within the same cell, indicating mutually exclusive protein modules and the co-existence of different cell states. Collectively our data show that DIA-ME is a powerful, scalable, and easy-to-implement strategy for single-cell proteomics.
Collapse
Affiliation(s)
- Karl K Krull
- German Cancer Research Center (DKFZ), Heidelberg, Division of Proteomics of Stem Cells and Cancer, Heidelberg, Germany
- Heidelberg University, Faculty of Biosciences, Heidelberg, Germany
- Heidelberg University, Medical Faculty, Heidelberg, Germany
| | - Syed Azmal Ali
- German Cancer Research Center (DKFZ), Heidelberg, Division of Proteomics of Stem Cells and Cancer, Heidelberg, Germany
| | - Jeroen Krijgsveld
- German Cancer Research Center (DKFZ), Heidelberg, Division of Proteomics of Stem Cells and Cancer, Heidelberg, Germany.
- Heidelberg University, Medical Faculty, Heidelberg, Germany.
| |
Collapse
|
3
|
Boulanger DSM, Douglas LR, Duriez PJ, Kang Y, Dalchau N, James E, Elliott T. Tapasin-mediated editing of the MHC I immunopeptidome is epitope specific and dependent on peptide off-rate, abundance, and level of tapasin expression. Front Immunol 2022; 13:956603. [PMID: 36389776 PMCID: PMC9659924 DOI: 10.3389/fimmu.2022.956603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 09/27/2022] [Indexed: 11/23/2022] Open
Abstract
Tapasin, a component of the major histocompatibility complex (MHC) I peptide loading complex, edits the repertoire of peptides that is presented at the cell surface by MHC I and thereby plays a key role in shaping the hierarchy of CD8+ T-cell responses to tumors and pathogens. We have developed a system that allows us to tune the level of tapasin expression and independently regulate the expression of competing peptides of different off-rates. By quantifying the relative surface expression of peptides presented by MHC I molecules, we show that peptide editing by tapasin can be measured in terms of “tapasin bonus,” which is dependent on both peptide kinetic stability (off-rate) and peptide abundance (peptide supply). Each peptide has therefore an individual tapasin bonus fingerprint. We also show that there is an optimal level of tapasin expression for each peptide in the immunopeptidome, dependent on its off-rate and abundance. This is important, as the level of tapasin expression can vary widely during different stages of the immune response against pathogens or cancer and is often the target for immune escape.
Collapse
Affiliation(s)
- Denise S. M. Boulanger
- Centre for Cancer Immunology, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- *Correspondence: Denise S. M. Boulanger, ; Tim Elliott,
| | - Leon R. Douglas
- Cancer Research UK (CR-UK) Protein Core Facility, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Patrick J. Duriez
- Cancer Research UK (CR-UK) Protein Core Facility, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Yoyel Kang
- Centre for Cancer Immunology, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | | | - Edd James
- Centre for Cancer Immunology, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Tim Elliott
- Centre for Cancer Immunology, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- Centre for Immuno-oncology, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- *Correspondence: Denise S. M. Boulanger, ; Tim Elliott,
| |
Collapse
|
4
|
Lee W, Suresh M. Vaccine adjuvants to engage the cross-presentation pathway. Front Immunol 2022; 13:940047. [PMID: 35979365 PMCID: PMC9376467 DOI: 10.3389/fimmu.2022.940047] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 07/08/2022] [Indexed: 11/13/2022] Open
Abstract
Adjuvants are indispensable components of vaccines for stimulating optimal immune responses to non-replicating, inactivated and subunit antigens. Eliciting balanced humoral and T cell-mediated immunity is paramount to defend against diseases caused by complex intracellular pathogens, such as tuberculosis, malaria, and AIDS. However, currently used vaccines elicit strong antibody responses, but poorly stimulate CD8 cytotoxic T lymphocyte (CTL) responses. To elicit potent CTL memory, vaccines need to engage the cross-presentation pathway, and this requirement has been a crucial bottleneck in the development of subunit vaccines that engender effective T cell immunity. In this review, we focus on recent insights into DC cross-presentation and the extent to which clinically relevant vaccine adjuvants, such as aluminum-based nanoparticles, water-in oil emulsion (MF59) adjuvants, saponin-based adjuvants, and Toll-like receptor (TLR) ligands modulate DC cross-presentation efficiency. Further, we discuss the feasibility of using carbomer-based adjuvants as next generation of adjuvant platforms to elicit balanced antibody- and T-cell based immunity. Understanding of the molecular mechanism of DC cross-presentation and the mode of action of adjuvants will pave the way for rational design of vaccines for infectious diseases and cancer that require balanced antibody- and T cell-based immunity.
Collapse
|
5
|
Isakov N. Histocompatibility and Reproduction: Lessons from the Anglerfish. LIFE (BASEL, SWITZERLAND) 2022; 12:life12010113. [PMID: 35054506 PMCID: PMC8780861 DOI: 10.3390/life12010113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/06/2022] [Accepted: 01/08/2022] [Indexed: 11/16/2022]
Abstract
Reproduction in certain deep-sea anglerfishes involves the permanent attachment of dwarf males to much larger females and fusion of their tissues leading to the establishment of a shared circulatory system. This unusual phenomenon of sexual parasitism enables anglerfishes to maximize reproductive success in the vast and deep oceans, where females and males otherwise rarely meet. An even more surprising phenomenon relates to the observation that joining of genetically disparate male and female anglerfishes does not evoke a strong anti-graft immune rejection response, which occurs in vertebrates following allogeneic parabiosis. Recent studies demonstrated that the evolutionary processes that led to the unique mating strategy of anglerfishes coevolved with genetic changes that resulted in loss of functional genes encoding critical components of the adaptive immune system. These genetic alterations enabled anglerfishes to tolerate the histoincompatible tissue antigens of their mate and prevent the occurrence of reciprocal graft rejection responses. While the exact mechanisms by which anglerfishes defend themselves against pathogens have not yet been deciphered, it is speculated that during evolution, anglerfishes adopted new immune strategies that compensate for the loss of B and T lymphocyte functions and enable them to resist infection by pathogens.
Collapse
Affiliation(s)
- Noah Isakov
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben Gurion University of the Negev, P.O. Box 653, Beer Sheva 84105, Israel
| |
Collapse
|
6
|
Vollmers S, Lobermeyer A, Körner C. The New Kid on the Block: HLA-C, a Key Regulator of Natural Killer Cells in Viral Immunity. Cells 2021; 10:cells10113108. [PMID: 34831331 PMCID: PMC8620871 DOI: 10.3390/cells10113108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/04/2021] [Accepted: 11/05/2021] [Indexed: 11/01/2022] Open
Abstract
The human leukocyte antigen system (HLA) is a cluster of highly polymorphic genes essential for the proper function of the immune system, and it has been associated with a wide range of diseases. HLA class I molecules present intracellular host- and pathogen-derived peptides to effector cells of the immune system, inducing immune tolerance in healthy conditions or triggering effective immune responses in pathological situations. HLA-C is the most recently evolved HLA class I molecule, only present in humans and great apes. Differentiating from its older siblings, HLA-A and HLA-B, HLA-C exhibits distinctive features in its expression and interaction partners. HLA-C serves as a natural ligand for multiple members of the killer-cell immunoglobulin-like receptor (KIR) family, which are predominately expressed by natural killer (NK) cells. NK cells are crucial for the early control of viral infections and accumulating evidence indicates that interactions between HLA-C and its respective KIR receptors determine the outcome and progression of viral infections. In this review, we focus on the unique role of HLA-C in regulating NK cell functions and its consequences in the setting of viral infections.
Collapse
|
7
|
Mathematical modeling and stochastic simulations suggest that low-affinity peptides can bisect MHC1-mediated export of high-affinity peptides into "early"- and "late"-phases. Heliyon 2021; 7:e07466. [PMID: 34286133 PMCID: PMC8278427 DOI: 10.1016/j.heliyon.2021.e07466] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 05/23/2021] [Accepted: 06/29/2021] [Indexed: 02/01/2023] Open
Abstract
The peptide loading complex (PLC) is a multi-protein complex of the endoplasmic reticulum (ER) which optimizes major histocompatibility I (MHC1)-mediated export of intracellular high-affinity peptides. Whilst, the molecular biology of MHC1-mediated export is well supported by empirical data, the stoichiometry, kinetics and spatio-temporal profile of the participating molecular entities are a matter of considerable debate. Here, a low-affinity peptide-driven (LAPD)-model of MHC1-mediated high-affinity peptide export is formulated, implemented, analyzed and simulated. The model is parameterized in terms of the contribution of the shunt reaction to the concentration of exportable MHC1. Theoretical analyses and simulation studies of the model suggest that low-affinity peptides can bisect MHC1-mediated export of high-affinity peptides into time-dependent distinct “early”- and “late”-phases. The net exportable MHC1 (eM1β(t)) is a function of the retrograde (rM1β(t))- and anterograde (aM1β(t))-derived fractions. The “early”-phase is dominated by the contribution of the retrograde/recyclable (rM1β≈61%,aM1β≈39%) pathway to exportable MHC1, is characterized by Tapasin-mediated peptide-editing and is ATP-independent. The “late”-phase on the other hand, is characterized by de novo PLC-assembly, rapid disassembly and a significant contribution of the anterograde pathway to exportable MHC1 (rM1β≈21%,aM1β≈79%). The shunt reaction is rate limiting and may integrate peptide translocation with PLC-assembly/disassembly thereby, regulating peptide export under physiological and pathological (viral infections, dysplastic alterations) conditions.
Collapse
|
8
|
Tsoras AN, Wong KM, Paravastu AK, Champion JA. Rational Design of Antigen Incorporation Into Subunit Vaccine Biomaterials Can Enhance Antigen-Specific Immune Responses. Front Immunol 2020; 11:1547. [PMID: 32849524 PMCID: PMC7396695 DOI: 10.3389/fimmu.2020.01547] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 06/11/2020] [Indexed: 12/29/2022] Open
Abstract
Peptide subunit vaccines increase safety by reducing the risk of off-target responses and improving the specificity of the induced adaptive immune response. The immunogenicity of most soluble peptides, however, is often insufficient to produce robust and lasting immunity. Many biomaterials and delivery vehicles have been developed for peptide antigens to improve immune response while maintaining specificity. Peptide nanoclusters (PNC) are a subunit peptide vaccine material that has shown potential to increase immunogenicity of peptide antigens. PNC are comprised only of crosslinked peptide antigen and have been synthesized from several peptide antigens as small as 8 amino acids in length. However, as with many peptide vaccine biomaterials, synthesis requires adding residues to the peptide and/or engaging amino acids within the antigen epitope covalently to form a stable material. The impact of antigen modifications made to enable biomaterial incorporation or formation is rarely investigated, since the goal of most studies is to compare the soluble antigen with biomaterial form of antigen. This study investigates PNC as a platform vaccine biomaterial to evaluate how peptide modification and biomaterial formation with different crosslinking chemistries affect epitope-specific immune cell presentation and activation. Several types of PNC were synthesized by desolvation from the model peptide epitope SIINFEKL, which is derived from the immunogenic protein ovalbumin. SIINFEKL was altered to include extra residues on each end, strategically chosen to enable multiple conjugation chemistry options for incorporation into PNC. Several crosslinking methods were used to control which functional groups were used to stabilize the PNC, as well as the reducibility of the crosslinking. These variations were evaluated for immune responses and biodistribution following in vivo immunization. All modified antigen formulations still induced comparable immune responses when incorporated into PNC compared to unmodified soluble antigen alone. However, some crosslinking methods led to a significant increase in desirable immune responses while others did not, suggesting that not all PNC were processed the same. These results help guide future peptide vaccine biomaterial design, including PNC and a wide variety of conjugated and self-assembled peptide antigen materials, to maximize and tune the desired immune response.
Collapse
Affiliation(s)
| | | | | | - Julie A. Champion
- School of Chemical & Biomolecular Engineering, Atlanta, GA, United States
| |
Collapse
|
9
|
Prokhnevska N, Emerson DA, Kissick HT, Redmond WL. Immunological Complexity of the Prostate Cancer Microenvironment Influences the Response to Immunotherapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1210:121-147. [PMID: 31900908 DOI: 10.1007/978-3-030-32656-2_7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Prostate cancer is one of the most common cancers in men and a leading cause of cancer-related death. Recent advances in the treatment of advanced prostate cancer, including the use of more potent and selective inhibitors of the androgen signaling pathway, have provided significant clinical benefit for men with metastatic castration-resistant prostate cancer (mCRPC). However, most patients develop progressive lethal disease, highlighting the need for more effective treatments. One such approach is immunotherapy, which harness the power of the patient's immune system to identify and destroy cancer cells through the activation of cytotoxic CD8 T cells specific for tumor antigens. Although immunotherapy, particularly checkpoint blockade, can induce significant clinical responses in patients with solid tumors or hematological malignancies, minimal efficacy has been observed in men with mCRPC. In the current review, we discuss our current understanding of the immunological complexity of the immunosuppressive prostate cancer microenvironment, preclinical models of prostate cancer, and recent advances in immunotherapy clinical trials to improve outcomes for men with mCRPC.
Collapse
Affiliation(s)
| | - Dana A Emerson
- Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR, USA.,Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA
| | | | - William L Redmond
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA.
| |
Collapse
|
10
|
Wang H, Zhu N, Ye X, Wang L, Wang B, Shan W, Lai X, Tan Y, Fu S, Xiao H, Huang H. PTPN21-CDS long isoform inhibits the response of acute lymphoblastic leukemia cells to NK-mediated lysis via the KIR/HLA-I axis. J Cell Biochem 2020; 121:3298-3312. [PMID: 31898344 DOI: 10.1002/jcb.29601] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 12/11/2019] [Indexed: 12/11/2022]
Abstract
Protein tyrosine phosphatase non-receptor type 21 (PTPN21) is a member of the non-receptor tyrosine phosphatase family. We have found that PTPN21 is mutated in relapsed Philadelphia chromosome-negative acute lymphoblastic leukemia (ALL) after allogeneic hematopoietic stem cell transplantation. PTPN21 consists of three types of isoforms according to the length of the protein encoded. However, the roles of different isoforms in leukemic cells have not been elucidated. In the study, PTPN21 isoform constitution in five ALL cell lines were identified by transcriptome polymerase chain reaction combined with Sanger sequencing, and the relationship between PTPN21 isoforms and sensitivity to natural killer (NK) cells mediated killing in ALL cell lines were further assessed by knock-out of different isoforms of PTPN21 using CRISPR-Cas9 technique. Subsequently, we explored the functional mechanisms through RNA sequencing and confirmatory testing. The results showed that there was no significant change when all PTPN21 isoforms were knocked out in ALL cells, but the sensitivity of NALM6 cells with PTPN21-CDSlong knock-out (NALM6-PTPN21lk ) to NK-mediated killing was significantly increased. Whole transcriptome sequencing and further validation testing showed that human leukocyte antigen class I (HLA-I) molecules were significantly decreased, accompanied by a significantly downregulated expression of antigen presenting-related chaperones in NALM6-PTPN21lk cells. Our results uncovered a previously unknown mechanism that PTPN21-CDSlong and CDSshort isoforms may play opposite roles in NK-mediated killing in ALL cells, and showed that the endogenous PTPN21-CDSlong isoform inhibited ALL cells to NK cell-mediated lysis by regulating the KIR-HLA-I axis.
Collapse
Affiliation(s)
- Huafang Wang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, China.,Zhejiang Engineering Laboratory for Stem cell and Immunotherapy, Hangzhou, Zhejiang, China
| | - Ni Zhu
- Department of Hematology, The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Xiaohang Ye
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Limengmeng Wang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, China.,Zhejiang Engineering Laboratory for Stem cell and Immunotherapy, Hangzhou, Zhejiang, China
| | - Binsheng Wang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, China.,Zhejiang Engineering Laboratory for Stem cell and Immunotherapy, Hangzhou, Zhejiang, China
| | - Wei Shan
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, China.,Zhejiang Engineering Laboratory for Stem cell and Immunotherapy, Hangzhou, Zhejiang, China
| | - Xiaoyu Lai
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yamin Tan
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Shan Fu
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Haowen Xiao
- Department of Hematology, The Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, China
| | - He Huang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, China.,Zhejiang Engineering Laboratory for Stem cell and Immunotherapy, Hangzhou, Zhejiang, China
| |
Collapse
|
11
|
Tumor mechanisms of resistance to immune attack. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2019; 164:61-100. [PMID: 31383409 DOI: 10.1016/bs.pmbts.2019.03.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The immune system plays a key role in the interactions between host and tumor. Immune selection pressure is a driving force behind the sculpting and evolution of malignant cancer cells to escape this immune attack. Several common tumor cell-based mechanisms of resistance to immune attack have been identified and can be broadly categorized into three main classes: loss of antigenicity, loss of immunogenicity, and creation of an immunosuppressive microenvironment. In this review, we will discuss in detail the relevant literature associated with each class of resistance and will describe the relevance of these mechanisms to human cancer patients. To conclude, we will outline the implications these mechanisms have for the treatment of cancer using currently available therapeutic approaches. Immunotherapy has been a successful addition to current treatment approaches, but many patients either do not respond or quickly become resistant. This reflects the ability of tumors to continue to adapt to immune selection pressure at all stages of development. Additional study of immune escape mechanisms and immunotherapy resistance mechanisms will be needed to inform future treatment approaches.
Collapse
|
12
|
Natarajan K, Jiang J, May NA, Mage MG, Boyd LF, McShan AC, Sgourakis NG, Bax A, Margulies DH. The Role of Molecular Flexibility in Antigen Presentation and T Cell Receptor-Mediated Signaling. Front Immunol 2018; 9:1657. [PMID: 30065727 PMCID: PMC6056622 DOI: 10.3389/fimmu.2018.01657] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 07/04/2018] [Indexed: 01/20/2023] Open
Abstract
Antigen presentation is a cellular process that involves a number of steps, beginning with the production of peptides by proteolysis or aberrant synthesis and the delivery of peptides to cellular compartments where they are loaded on MHC class I (MHC-I) or MHC class II (MHC-II) molecules. The selective loading and editing of high-affinity immunodominant antigens is orchestrated by molecular chaperones: tapasin/TAP-binding protein, related for MHC-I and HLA-DM for MHC-II. Once peptide/MHC (pMHC) complexes are assembled, following various steps of quality control, they are delivered to the cell surface, where they are available for identification by αβ receptors on CD8+ or CD4+ T lymphocytes. In addition, recognition of cell surface peptide/MHC-I complexes by natural killer cell receptors plays a regulatory role in some aspects of the innate immune response. Many of the components of the pathways of antigen processing and presentation and of T cell receptor (TCR)-mediated signaling have been studied extensively by biochemical, genetic, immunological, and structural approaches over the past several decades. Until recently, however, dynamic aspects of the interactions of peptide with MHC, MHC with molecular chaperones, or of pMHC with TCR have been difficult to address experimentally, although computational approaches such as molecular dynamics (MD) simulations have been illuminating. Studies exploiting X-ray crystallography, cryo-electron microscopy, and multidimensional nuclear magnetic resonance (NMR) spectroscopy are beginning to reveal the importance of molecular flexibility as it pertains to peptide loading onto MHC molecules, the interactions between pMHC and TCR, and subsequent TCR-mediated signals. In addition, recent structural and dynamic insights into how molecular chaperones define peptide selection and fine-tune the MHC displayed antigen repertoire are discussed. Here, we offer a review of current knowledge that highlights experimental data obtained by X-ray crystallography and multidimensional NMR methodologies. Collectively, these findings strongly support a multifaceted role for protein plasticity and conformational dynamics throughout the antigen processing and presentation pathway in dictating antigen selection and recognition.
Collapse
Affiliation(s)
- Kannan Natarajan
- Molecular Biology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Jiansheng Jiang
- Molecular Biology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Nathan A May
- Molecular Biology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Michael G Mage
- Molecular Biology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Lisa F Boyd
- Molecular Biology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Andrew C McShan
- Department of Chemistry and Biochemistry, University of California at Santa Cruz, Santa Cruz, CA, United States
| | - Nikolaos G Sgourakis
- Department of Chemistry and Biochemistry, University of California at Santa Cruz, Santa Cruz, CA, United States
| | - Ad Bax
- Laboratory of Chemical Physics, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| | - David H Margulies
- Molecular Biology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
13
|
Transcriptome sequencing of the long-nosed bandicoot (Perameles nasuta) reveals conservation and innovation of immune genes in the marsupial order Peramelemorphia. Immunogenetics 2017; 70:327-336. [PMID: 29159447 DOI: 10.1007/s00251-017-1043-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 10/30/2017] [Indexed: 12/22/2022]
Abstract
Bandicoots are omnivorous marsupials of the order Peramelemorphia. Conservation concerns and their unique biological characteristics suggest peramelomorphs are worthy research subjects, but knowledge of their genetics and immunology has lagged behind that of other high-profile marsupials. Here, we characterise the transcriptome of the long-nose bandicoot (Perameles nasuta), the first high-throughput data set from any peramelomorph. We investigate the immune gene repertoire of the bandicoot, with a focus on key immune gene families, and compare to previously characterised marsupial and mammalian species. We find that the immune gene complement in bandicoot is often conserved with respect to other marsupials; however, the diversity of expressed transcripts in several key families, such as major histocompatibility complex, T cell receptor μ and natural killer cell receptors, appears greater in the bandicoot than other Australian marsupials, including devil and koala. This transcriptome is an important first step for future studies of bandicoots and the bilby, allowing for population level analysis and construction of bandicoot-specific immunological reagents and assays. Such studies will be critical to understanding the immunology and physiology of Peramelemorphia and to inform the conservation of these unique marsupials.
Collapse
|
14
|
Gao S, Kang Y, Yuan R, Ma H, Xiang B, Wang Z, Dai X, Wang F, Xiao J, Liao M, Ren T. Immune Responses of Chickens Infected with Wild Bird-Origin H5N6 Avian Influenza Virus. Front Microbiol 2017; 8:1081. [PMID: 28676793 PMCID: PMC5476689 DOI: 10.3389/fmicb.2017.01081] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 05/29/2017] [Indexed: 11/13/2022] Open
Abstract
Since April 2014, new infections of H5N6 avian influenza virus (AIV) in humans and domestic poultry have caused considerable economic losses in the poultry industry and posed an enormous threat to human health worldwide. In previous research using gene sequence and phylogenetic analysis, we reported that H5N6 AIV isolated in February 2015 (ZH283) in Pallas’s sandgrouse was highly similar to that isolated in a human in December 2015 (A/Guangdong/ZQ874/2015), whereas a virus (i.e., SW8) isolated in oriental magpie-robin in 2014 was highly similar to that of A/chicken/Dongguan/2690/2013 (H5N6). However, the pathogenicity, transmissibility, and host immune-related response of chickens infected by those wild bird-origin H5N6 AIVs remain unknown. In response, we examined the viral distribution and mRNA expression profiles of immune-related genes in chickens infected with both viruses. Results showed that the H5N6 AIVs were highly pathogenic to chickens and caused not only systemic infection in multiple tissues, but also 100% mortality within 3–5 days post-infection. Additionally, ZH283 efficiently replicated in all tested tissues and transmitted among chickens more rapidly than SW8. Moreover, quantitative real-time polymerase chain reaction analysis showed that following infection with H5N6, AIVs immune-related genes remained active in a tissue-dependent manner, as well as that ZH283 induced mRNA expression profiles such as TLR3, TLR7, IL-6, TNF-α, IL-1β, IL-10, IL-8, and MHC-II to a greater extent than SW8 in the tested tissues of infected chickens. Altogether, our findings help to illuminate the pathogenesis and immunologic mechanisms of H5N6 AIVs in chickens.
Collapse
Affiliation(s)
- Shimin Gao
- College of Animal Science and Veterinary Medicine, Shanxi Agriculture UniversityTaigu, China.,College of Veterinary Medicine, Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, South China Agricultural UniversityGuangzhou, China
| | - Yinfeng Kang
- College of Veterinary Medicine, Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, South China Agricultural UniversityGuangzhou, China.,State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Experimental Research, Sun Yat-sen University Cancer CenterGuangzhou, China
| | - Runyu Yuan
- College of Veterinary Medicine, Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, South China Agricultural UniversityGuangzhou, China.,Key Laboratory for Repository and Application of Pathogenic Microbiology, Research Center for Pathogens Detection Technology of Emerging Infectious Diseases, Guangdong Provincial Center for Disease Control and PreventionGuangzhou, China
| | - Haili Ma
- College of Animal Science and Veterinary Medicine, Shanxi Agriculture UniversityTaigu, China
| | - Bin Xiang
- College of Veterinary Medicine, Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, South China Agricultural UniversityGuangzhou, China
| | - Zhaoxiong Wang
- College of Animal Science, Yangtze UniversityJingzhou, China
| | - Xu Dai
- College of Veterinary Medicine, Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, South China Agricultural UniversityGuangzhou, China
| | - Fumin Wang
- Guangdong Provincial Wildlife Rescue CenterGuangzhou, China
| | - Jiajie Xiao
- Guangdong Provincial Wildlife Rescue CenterGuangzhou, China
| | - Ming Liao
- College of Veterinary Medicine, Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, South China Agricultural UniversityGuangzhou, China
| | - Tao Ren
- College of Veterinary Medicine, Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, South China Agricultural UniversityGuangzhou, China
| |
Collapse
|
15
|
Ge F, Memon S, Xi D, Li S, Liu X, Li G, Wang L, Leng J, Khan S, Deng W. Cloning and characterization of MHC-DQA1 and MHC-DQA2 molecules from yak (<i>Bos grunniens</i>). Arch Anim Breed 2016. [DOI: 10.5194/aab-59-395-2016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Abstract. The major histocompatibility complex (MHC) plays a crucial role in the processing and presentation of antigens and in discrimination between self and non-self. The aim of this investigation was to scrutinize the structural diversity and possible duplication of the MHC-DQA genes in yak (Bos grunniens). Two cDNA sequences were amplified and designated as Bogr-DQA1 (DQA*0101) and Bogr-DQA2 (DQA*2001) with GenBank accession numbers JQ864314 and JQ864315, respectively. The nucleotide and amino acid sequence alignment between Bogr-DQA1 and Bogr-DQA2 molecules showed that these two identified MHC-DQA gene sequences had more similarity to alleles of specific DQA1 and DQA2 genes from other Ruminantia species than to each other. The result from phylogenic investigation also revealed that there was a larger genetic distance between these two genes than between homologous genes from different species. The presence of different bovine DQA putative motifs and the large genetic distance between Bogr-DQA1 and Bogr-DQA2 suggest that these sequences are non-allelic. Further, these results indicate that DQA gene duplication occurs in ruminants. This study will be helpful in knowing MHC diversity in common ruminants and will deepen our understanding of the variation of immunological functions, evolutionary constraints, and selective forces that affect MHC variation within and between species.
Collapse
|
16
|
Ma Y, Cheng L, Yuan B, Zhang Y, Zhang C, Zhang Y, Tang K, Zhuang R, Chen L, Yang K, Zhang F, Jin B. Structure and Function of HLA-A*02-Restricted Hantaan Virus Cytotoxic T-Cell Epitope That Mediates Effective Protective Responses in HLA-A2.1/K(b) Transgenic Mice. Front Immunol 2016; 7:298. [PMID: 27551282 PMCID: PMC4976285 DOI: 10.3389/fimmu.2016.00298] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 07/22/2016] [Indexed: 12/11/2022] Open
Abstract
Hantavirus infections cause severe emerging diseases in humans and are associated with high mortality rates; therefore, they have become a global public health concern. Our previous study showed that the CD8(+) T-cell epitope aa129-aa137 (FVVPILLKA, FA9) of the Hantaan virus (HTNV) nucleoprotein (NP), restricted by human leukocyte antigen (HLA)-A*02, induced specific CD8(+) T-cell responses that controlled HTNV infection in humans. However, the in vivo immunogenicity of peptide FA9 and the effect of FA9-specific CD8(+) T-cell immunity remain unclear. Here, based on a detailed structural analysis of the peptide FA9/HLA-A*0201 complex and functional investigations using HLA-A2.1/K(b) transgenic (Tg) mice, we found that the overall structure of the peptide FA9/HLA-A*0201 complex displayed a typical MHC class I fold with Val2 and Ala9 as primary anchor residues and Val3 and Leu7 as secondary anchor residues that allow peptide FA9 to bind tightly with an HLA-A*0201 molecule. Residues in the middle portion of peptide FA9 extruding out of the binding groove may be the sites that allow for recognition by T-cell receptors. Immunization with peptide FA9 in HLA-A2.1/K(b) Tg mice induced FA9-specific cytotoxic T-cell responses characterized by the induction of high expression levels of interferon-γ, tumor necrosis factor-α, granzyme B, and CD107a. In an HTNV challenge trial, significant reductions in the levels of both the antigens and the HTNV RNA loads were observed in the liver, spleen, and kidneys of Tg mice pre-vaccinated with peptide FA9. Thus, our findings highlight the ability of HTNV epitope-specific CD8(+) T-cell immunity to control HTNV and support the possibility that the HTNV-NP FA9 peptide, naturally processed in vivo in an HLA-A*02-restriction manner, may be a good candidate for the development HTNV peptide vaccines.
Collapse
Affiliation(s)
- Ying Ma
- Department of Immunology, The Fourth Military Medical University , Xi'an , China
| | - Linfeng Cheng
- Department of Microbiology, The Fourth Military Medical University , Xi'an , China
| | - Bin Yuan
- Institute of Orthopaedics of Xijing Hospital, The Fourth Military Medical University , Xi'an , China
| | - Yusi Zhang
- Department of Immunology, The Fourth Military Medical University , Xi'an , China
| | - Chunmei Zhang
- Department of Immunology, The Fourth Military Medical University , Xi'an , China
| | - Yun Zhang
- Department of Immunology, The Fourth Military Medical University , Xi'an , China
| | - Kang Tang
- Department of Immunology, The Fourth Military Medical University , Xi'an , China
| | - Ran Zhuang
- Department of Immunology, The Fourth Military Medical University , Xi'an , China
| | - Lihua Chen
- Department of Immunology, The Fourth Military Medical University , Xi'an , China
| | - Kun Yang
- Department of Immunology, The Fourth Military Medical University , Xi'an , China
| | - Fanglin Zhang
- Department of Microbiology, The Fourth Military Medical University , Xi'an , China
| | - Boquan Jin
- Department of Immunology, The Fourth Military Medical University , Xi'an , China
| |
Collapse
|
17
|
Lum R, Ahmad S, Hong SJ, Chapman DC, Kozlov G, Williams DB. Contributions of the Lectin and Polypeptide Binding Sites of Calreticulin to Its Chaperone Functions in Vitro and in Cells. J Biol Chem 2016; 291:19631-41. [PMID: 27413183 DOI: 10.1074/jbc.m116.746321] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Indexed: 11/06/2022] Open
Abstract
Calreticulin is a lectin chaperone of the endoplasmic reticulum that interacts with newly synthesized glycoproteins by binding to Glc1Man9GlcNAc2 oligosaccharides as well as to the polypeptide chain. In vitro, the latter interaction potently suppresses the aggregation of various non-glycosylated proteins. Although the lectin-oligosaccharide association is well understood, the polypeptide-based interaction is more controversial because the binding site on calreticulin has not been identified, and its significance in the biogenesis of glycoproteins in cells remains unknown. In this study, we identified the polypeptide binding site responsible for the in vitro aggregation suppression function by mutating four candidate hydrophobic surface patches. Mutations in only one patch, P19K/I21E and Y22K/F84E, impaired the ability of calreticulin to suppress the thermally induced aggregation of non-glycosylated firefly luciferase. These mutants also failed to bind several hydrophobic peptides that act as substrate mimetics and compete in the luciferase aggregation suppression assay. To assess the relative contributions of the glycan-dependent and -independent interactions in living cells, we expressed lectin-deficient, polypeptide binding-deficient, and doubly deficient calreticulin constructs in calreticulin-negative cells and monitored the effects on the biogenesis of MHC class I molecules, the solubility of mutant forms of α1-antitrypsin, and interactions with newly synthesized glycoproteins. In all cases, we observed a profound impairment in calreticulin function when its lectin site was inactivated. Remarkably, inactivation of the polypeptide binding site had little impact. These findings indicate that the lectin-based mode of client interaction is the predominant contributor to the chaperone functions of calreticulin within the endoplasmic reticulum.
Collapse
Affiliation(s)
- Ronnie Lum
- From the Departments of Biochemistry and Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada and
| | - Samar Ahmad
- From the Departments of Biochemistry and Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada and
| | - Seo Jung Hong
- From the Departments of Biochemistry and Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada and
| | - Daniel C Chapman
- From the Departments of Biochemistry and Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada and
| | - Guennadi Kozlov
- the Groupe de Recherche Axé sur la Structure des Proteines, Department of Biochemistry, McGill University, Montréal, Québec H3G 0B1, Canada
| | - David B Williams
- From the Departments of Biochemistry and Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada and
| |
Collapse
|
18
|
Castiblanco J, Anaya JM. Genetics and vaccines in the era of personalized medicine. Curr Genomics 2015; 16:47-59. [PMID: 25937813 PMCID: PMC4412964 DOI: 10.2174/1389202916666141223220551] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Revised: 12/22/2014] [Accepted: 12/23/2014] [Indexed: 12/17/2022] Open
Abstract
Vaccines represent the most successful and sustainable tactic to prevent and counteract infection. A vaccine generally improves immunity to a particular disease upon administration by inducing specific protective and efficient immune responses in all of the receiving population. The main known factors influencing the observed heterogeneity for immune re-sponses induced by vaccines are gender, age, co-morbidity, immune system, and genetic background. This review is mainly focused on the genetic status effect to vaccine immune responses and how this could contribute to the development of novel vaccine candidates that could be better directed and predicted relative to the genetic history of an individual and/or population. The text offers a brief history of vaccinology as a field, a description of the genetic status of the most relevant and studied genes and their functionality and correlation with exposure to specific vaccines; followed by an inside look into autoimmunity as a concern when designing vaccines as well as perspectives and conclusions looking towards an era of personalized and predictive vaccinology instead of a one size fits all approach.
Collapse
Affiliation(s)
- John Castiblanco
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Carrera 24 #63-C-69, Bogota, Colombia ; Doctoral Program in Biomedical Sciences, Universidad del Rosario, Bogotá,Colombia
| | - Juan-Manuel Anaya
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Carrera 24 #63-C-69, Bogota, Colombia
| |
Collapse
|
19
|
Morris KM, Kirby K, Beatty JA, Barrs VR, Cattley S, David V, O'Brien SJ, Menotti-Raymond M, Belov K. Development of MHC-Linked Microsatellite Markers in the Domestic Cat and Their Use to Evaluate MHC Diversity in Domestic Cats, Cheetahs, and Gir Lions. J Hered 2014; 105:493-505. [PMID: 24620003 PMCID: PMC4048552 DOI: 10.1093/jhered/esu017] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Accepted: 01/14/2014] [Indexed: 11/15/2022] Open
Abstract
Diversity within the major histocompatibility complex (MHC) reflects the immunological fitness of a population. MHC-linked microsatellite markers provide a simple and an inexpensive method for studying MHC diversity in large-scale studies. We have developed 6 MHC-linked microsatellite markers in the domestic cat and used these, in conjunction with 5 neutral microsatellites, to assess MHC diversity in domestic mixed breed (n = 129) and purebred Burmese (n = 61) cat populations in Australia. The MHC of outbred Australian cats is polymorphic (average allelic richness = 8.52), whereas the Burmese population has significantly lower MHC diversity (average allelic richness = 6.81; P < 0.01). The MHC-linked microsatellites along with MHC cloning and sequencing demonstrated moderate MHC diversity in cheetahs (n = 13) and extremely low diversity in Gir lions (n = 13). Our MHC-linked microsatellite markers have potential future use in diversity and disease studies in other populations and breeds of cats as well as in wild felid species.
Collapse
Affiliation(s)
- Katrina M Morris
- From the Faculty of Veterinary Science, University of Sydney, Sydney, NSW 2006, Australia (Morris, Kirby, Beatty, Barrs, and Belov); the ANGIS, University of Sydney, Sydney, NSW 2006, Australia (Cattley); the Laboratory of Genomic Diversity, National Cancer Institute, Frederick, MD 21702-1201 (David and Menotti-Raymond); the Theodosius Dobzhansky Center for Genome Bioinformatics, St. Petersburg State University, St. Petersburg, Russia (O'Brien); and the Oceanographic Center, Nova Southeastern University, Ft Lauderdale, FL 33314-7796 (O'Brien)
| | - Katherine Kirby
- From the Faculty of Veterinary Science, University of Sydney, Sydney, NSW 2006, Australia (Morris, Kirby, Beatty, Barrs, and Belov); the ANGIS, University of Sydney, Sydney, NSW 2006, Australia (Cattley); the Laboratory of Genomic Diversity, National Cancer Institute, Frederick, MD 21702-1201 (David and Menotti-Raymond); the Theodosius Dobzhansky Center for Genome Bioinformatics, St. Petersburg State University, St. Petersburg, Russia (O'Brien); and the Oceanographic Center, Nova Southeastern University, Ft Lauderdale, FL 33314-7796 (O'Brien)
| | - Julia A Beatty
- From the Faculty of Veterinary Science, University of Sydney, Sydney, NSW 2006, Australia (Morris, Kirby, Beatty, Barrs, and Belov); the ANGIS, University of Sydney, Sydney, NSW 2006, Australia (Cattley); the Laboratory of Genomic Diversity, National Cancer Institute, Frederick, MD 21702-1201 (David and Menotti-Raymond); the Theodosius Dobzhansky Center for Genome Bioinformatics, St. Petersburg State University, St. Petersburg, Russia (O'Brien); and the Oceanographic Center, Nova Southeastern University, Ft Lauderdale, FL 33314-7796 (O'Brien)
| | - Vanessa R Barrs
- From the Faculty of Veterinary Science, University of Sydney, Sydney, NSW 2006, Australia (Morris, Kirby, Beatty, Barrs, and Belov); the ANGIS, University of Sydney, Sydney, NSW 2006, Australia (Cattley); the Laboratory of Genomic Diversity, National Cancer Institute, Frederick, MD 21702-1201 (David and Menotti-Raymond); the Theodosius Dobzhansky Center for Genome Bioinformatics, St. Petersburg State University, St. Petersburg, Russia (O'Brien); and the Oceanographic Center, Nova Southeastern University, Ft Lauderdale, FL 33314-7796 (O'Brien)
| | - Sonia Cattley
- From the Faculty of Veterinary Science, University of Sydney, Sydney, NSW 2006, Australia (Morris, Kirby, Beatty, Barrs, and Belov); the ANGIS, University of Sydney, Sydney, NSW 2006, Australia (Cattley); the Laboratory of Genomic Diversity, National Cancer Institute, Frederick, MD 21702-1201 (David and Menotti-Raymond); the Theodosius Dobzhansky Center for Genome Bioinformatics, St. Petersburg State University, St. Petersburg, Russia (O'Brien); and the Oceanographic Center, Nova Southeastern University, Ft Lauderdale, FL 33314-7796 (O'Brien)
| | - Victor David
- From the Faculty of Veterinary Science, University of Sydney, Sydney, NSW 2006, Australia (Morris, Kirby, Beatty, Barrs, and Belov); the ANGIS, University of Sydney, Sydney, NSW 2006, Australia (Cattley); the Laboratory of Genomic Diversity, National Cancer Institute, Frederick, MD 21702-1201 (David and Menotti-Raymond); the Theodosius Dobzhansky Center for Genome Bioinformatics, St. Petersburg State University, St. Petersburg, Russia (O'Brien); and the Oceanographic Center, Nova Southeastern University, Ft Lauderdale, FL 33314-7796 (O'Brien)
| | - Stephen J O'Brien
- From the Faculty of Veterinary Science, University of Sydney, Sydney, NSW 2006, Australia (Morris, Kirby, Beatty, Barrs, and Belov); the ANGIS, University of Sydney, Sydney, NSW 2006, Australia (Cattley); the Laboratory of Genomic Diversity, National Cancer Institute, Frederick, MD 21702-1201 (David and Menotti-Raymond); the Theodosius Dobzhansky Center for Genome Bioinformatics, St. Petersburg State University, St. Petersburg, Russia (O'Brien); and the Oceanographic Center, Nova Southeastern University, Ft Lauderdale, FL 33314-7796 (O'Brien)
| | - Marilyn Menotti-Raymond
- From the Faculty of Veterinary Science, University of Sydney, Sydney, NSW 2006, Australia (Morris, Kirby, Beatty, Barrs, and Belov); the ANGIS, University of Sydney, Sydney, NSW 2006, Australia (Cattley); the Laboratory of Genomic Diversity, National Cancer Institute, Frederick, MD 21702-1201 (David and Menotti-Raymond); the Theodosius Dobzhansky Center for Genome Bioinformatics, St. Petersburg State University, St. Petersburg, Russia (O'Brien); and the Oceanographic Center, Nova Southeastern University, Ft Lauderdale, FL 33314-7796 (O'Brien)
| | - Katherine Belov
- From the Faculty of Veterinary Science, University of Sydney, Sydney, NSW 2006, Australia (Morris, Kirby, Beatty, Barrs, and Belov); the ANGIS, University of Sydney, Sydney, NSW 2006, Australia (Cattley); the Laboratory of Genomic Diversity, National Cancer Institute, Frederick, MD 21702-1201 (David and Menotti-Raymond); the Theodosius Dobzhansky Center for Genome Bioinformatics, St. Petersburg State University, St. Petersburg, Russia (O'Brien); and the Oceanographic Center, Nova Southeastern University, Ft Lauderdale, FL 33314-7796 (O'Brien).
| |
Collapse
|
20
|
Lee CC, Wu CC, Lin TL. Chicken melanoma differentiation-associated gene 5 (MDA5) recognizes infectious bursal disease virus infection and triggers MDA5-related innate immunity. Arch Virol 2014; 159:1671-86. [PMID: 24452668 PMCID: PMC7086882 DOI: 10.1007/s00705-014-1983-9] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Accepted: 11/09/2013] [Indexed: 12/24/2022]
Abstract
The objective of the present study was to determine if chicken melanoma differentiation-associated gene 5 (MDA5) senses infectious bursal disease virus (IBDV) infection to initiate and amplify an innate immune response in the chicken MDA5 (chMDA5) signaling pathway. Chicken embryo fibroblast DF-1 cells were infected with IBDV LP1 at a multiplicity of infection (MOI) of 0.5 or 10. In addition, knockdown and overexpression of chMDA5 were performed by transfecting DF-1 cells with chMDA5-targeting small interfering RNA (siRNA) or chMDA5-expressing DNA. The transfected cells were infected with IBDV LP1 at an MOI of 10. Cell culture supernatants and lysates were collected at 2, 8, 16 and 24 hours postinfection (hpi) for IBDV titer determination and RNA extraction, respectively. IBDV RNA loads and mRNA expression levels of chicken MDA5, interferon-β (IFN-β) promoter stimulator 1 (IPS-1), interferon regulatory factor-3 (IRF-3), IFN-β, double-stranded RNA-dependent protein kinase (PKR), 2′,5′-oligoadenylate synthetase (OAS), myxovirus resistance gene (Mx), and major histocompatibility complex class I (MHC class I) were determined by real-time RT-PCR. The IBDV titer increased up to 1.4 × 107 plaque-forming units (PFU)/mL at 24 hpi, and the IBDV RNA load reached 464 ng/μL at 24 hpi. The mRNA expression levels of chicken MDA5, IRF-3, IFN-β, PKR, OAS, Mx and MHC class I in IBDV-infected DF-1 cells exhibited significant (p < 0.05) upregulation up to 906-, 199-, 26,310-, 12-, 66,144-, 64,039- and 33-fold, respectively. Expressed chMDA5 from transfection and double-stranded RNA from IBDV infection were localized or colocalized in the cytoplasm of DF-1 cells at 16 hpi. When chMDA5 was knocked down in DF-1 cells, IBDV titers and RNA loads were significantly higher (p < 0.05) than those in DF-1 cells without chMDA5 knockdown at 24 hpi. The expression levels of chicken MDA5, IRF-3, IFN-β and MHC class I in chMDA5-knockdown DF-1 cells were significantly lower (p < 0.05) at 16 and 24 hpi. DF-1 cells overexpressing chMDA5 by transfection with chMDA5 expressing DNA had significantly lower (p < 0.05) IBDV titers and RNA loads at 16 and 24 hpi and showed significantly higher (p < 0.05) expression of chicken MDA5, IRF-3, IFN-β, PKR, OAS, Mx and MHC class I at 2 hpi. The results indicated that chicken MDA5 recognized IBDV infection and that this interaction resulted in the activation of chMDA5-related innate immune genes and upregulation of chicken MHC class I.
Collapse
Affiliation(s)
- Chih-Chun Lee
- Department of Comparative Pathobiology, Purdue University, 406, S. University St, West Lafayette, IN, 47907, USA
| | | | | |
Collapse
|
21
|
Abstract
The mammalian immune system has evolved to display peptides derived from microbial antigens to immune effector cells. Liberated from the intact antigens through distinct proteolytic mechanisms, these peptides are subsequently transported to the cell surface while bound to chaperone-like receptors known as major histocompatibility complex molecules. These complexes are then scrutinized by T-cells that express receptors with specificity for specific major histocompatibility complex-peptide complexes. In normal uninfected cells, this process of antigen processing and presentation occurs continuously, with the resultant array of self-antigen-derived peptides displayed on the surface of these cells. Changes in this cellular peptide array alert the immune system to changes in the intracellular environment that may be associated with infection, oncogenesis or other abnormal cellular processes, resulting in a cascade of events that result in the elimination of the abnormal cell. Since peptides play such an essential role in informing the immune system of infection with viral or microbial pathogens and the transformation of cells in malignancy, the tools of proteomics, in particular mass spectrometry, are ideally suited to study these immune responses at a molecular level. Recent advances in studies of immune responses that have utilized mass spectrometry and associated technologies are reviewed. The authors gaze into the future and look at current challenges and where proteomics will impact in immunology over the next 5 years.
Collapse
Affiliation(s)
- Nicholas A Williamson
- The University of Melbourne, Department of Biochemistry & Molecular Biology, The Bio21 Molecular Science & Biotechnology Institute, 3010, Victoria, Australia.
| | | |
Collapse
|
22
|
Ashraf S, Nitschke K, Warshow UM, Brooks CR, Kim AY, Lauer GM, Hydes TJ, Cramp ME, Alexander G, Little AM, Thimme R, Neumann-Haefelin C, Khakoo SI. Synergism of tapasin and human leukocyte antigens in resolving hepatitis C virus infection. Hepatology 2013; 58:881-9. [PMID: 23532923 PMCID: PMC3759612 DOI: 10.1002/hep.26415] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Accepted: 03/21/2013] [Indexed: 01/15/2023]
Abstract
UNLABELLED CD8+ T-cell responses to hepatitis C virus (HCV) are important in generating a successful immune response and spontaneously clearing infection. Human leukocyte antigen (HLA) class I presents viral peptides to CD8+ T cells to permit detection of infected cells, and tapasin is an important component of the peptide loading complex for HLA class I. We sought to determine if tapasin polymorphisms affected the outcome of HCV infection. Patients with resolved or chronic HCV infection were genotyped for the known G/C coding polymorphism in exon 4 of the tapasin gene. In a European, but not a US, Caucasian population, the tapasin G allele was significantly associated with the outcome of HCV infection, being found in 82.5% of resolvers versus 71.3% of persistently infected individuals (P = 0.02, odds ratio [OR] = 1.90 95% confidence interval [CI] = 1.11-3.23). This was more marked at the HLA-B locus at which heterozygosity of both tapasin and HLA-B was protective (P < 0.03). Individuals with an HLA-B allele with an aspartate at residue 114 and the tapasin G allele were more likely to spontaneously resolve HCV infection (P < 0.00003, OR = 3.2 95% CI = 1.6-6.6). Additionally, individuals with chronic HCV and the combination of an HLA-B allele with an aspartate at residue 114 and the tapasin G allele also had stronger CD8+ T-cell responses (P = 0.02, OR = 2.58, 95% CI-1.05-6.5). CONCLUSION Tapasin alleles contribute to the outcome of HCV infection by synergizing with polymorphisms at HLA-B in a population-specific manner. This polymorphism may be relevant for peptide vaccination strategies against HCV infection.
Collapse
Affiliation(s)
- Shirin Ashraf
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, UK
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Agranovich A, Maman Y, Louzoun Y. Viral proteome size and CD8+ T cell epitope density are correlated: the effect of complexity on selection. INFECTION GENETICS AND EVOLUTION 2013; 20:71-7. [PMID: 23954420 PMCID: PMC7106205 DOI: 10.1016/j.meegid.2013.07.030] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Revised: 07/29/2013] [Accepted: 07/31/2013] [Indexed: 12/29/2022]
Abstract
We analyze the relation between viral complexity and their adaptation to the host immune system. Viruses with few proteins and low number of nucleotides remove more CD8+ T cell epitopes. Within a virus, short proteins (with fewer amino acids) adapt better than long ones. The relation between total size and adaptation is host specific. Complexity limits genetic adaptation in the high-mutation rate strong selection regime.
The relation between the complexity of organisms and proteins and their evolution rates has been discussed in the context of multiple generic models. The main robust claim from most such models is the negative relation between complexity and the accumulation rate of mutations. Viruses accumulate escape mutations in their epitopes to avoid detection and destruction of their host cell by CD8+ T cells. The extreme regime of immune escape, namely, strong selection and high mutation rate, provide an opportunity to extend and validate the existing models of relation between complexity and evolution rate as proposed by Fisher and Kimura. Using epitope prediction algorithms to compute the epitopes presented on the most frequent human HLA alleles in over 100 fully sequenced human viruses, and over 900 non-human viruses, we here study the correlation between viruses/proteins complexity (as measured by the number of proteins in the virus and the length of each protein, respectively) and the rate of accumulation of escape mutation. The latter is evaluated by measuring the normalized epitope density of viral proteins. If the virus/protein complexity prevents the accumulation of escape mutations, the epitope density is expected to be positively correlated with both the number of proteins in the virus and the length of proteins. We show that such correlations are indeed observed for most human viruses. For non-human viruses the correlations were much less significant, indicating that the correlation is indeed induced by human HLA molecules.
Collapse
Affiliation(s)
- Alexandra Agranovich
- Department of Mathematics and Gonda Brain Research Center, Bar-Ilan University, Ramat Gan 52900, Israel
| | - Yaakov Maman
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Yoram Louzoun
- Department of Mathematics and Gonda Brain Research Center, Bar-Ilan University, Ramat Gan 52900, Israel
- Corresponding author. Tel.: +972 3 5317610; fax: +972 3 7384057.
| |
Collapse
|
24
|
Kim HJ, Pourmand N. HLA typing from RNA-seq data using hierarchical read weighting [corrected]. PLoS One 2013; 8:e67885. [PMID: 23840783 PMCID: PMC3696101 DOI: 10.1371/journal.pone.0067885] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Accepted: 05/23/2013] [Indexed: 11/18/2022] Open
Abstract
Correctly matching the HLA haplotypes of donor and recipient is essential to the success of allogenic hematopoietic stem cell transplantation. Current HLA typing methods rely on targeted testing of recognized antigens or sequences. Despite advances in Next Generation Sequencing, general high throughput transcriptome sequencing is currently underutilized for HLA haplotyping due to the central difficulty in aligning sequences within this highly variable region. Here we present the method, HLAforest, that can accurately predict HLA haplotype by hierarchically weighting reads and using an iterative, greedy, top down pruning technique. HLAforest correctly predicts >99% of allele group level (2 digit) haplotypes and 93% of peptide-level (4 digit) haplotypes of the most diverse HLA genes in simulations with read lengths and error rates modeling currently available sequencing technology. The method is very robust to sequencing error and can predict 99% of allele-group level haplotypes with substitution rates as high as 8.8%. When applied to data generated from a trio of cell lines, HLAforest corroborated PCR-based HLA haplotyping methods and accurately predicted 16/18 (89%) major class I genes for a daughter-father-mother trio at the peptide level. Major class II genes were predicted with 100% concordance between the daughter-father-mother trio. In fifty HapMap samples with paired end reads just 37 nucleotides long, HLAforest predicted 96.5% of allele group level HLA haplotypes correctly and 83% of peptide level haplotypes correctly. In sixteen RNAseq samples with limited coverage across HLA genes, HLAforest predicted 97.7% of allele group level haplotypes and 85% of peptide level haplotypes correctly.
Collapse
Affiliation(s)
- Hyunsung John Kim
- Biomolecular Engineering Department, Baskin School of Engineering, University of California, Santa Cruz, Santa Cruz, California, United States of America
- * E-mail: (HJK); (NP)
| | - Nader Pourmand
- Biomolecular Engineering Department, Baskin School of Engineering, University of California, Santa Cruz, Santa Cruz, California, United States of America
- * E-mail: (HJK); (NP)
| |
Collapse
|
25
|
Föll D, Hinrichs J, Tischer S, Battermann A, Schambach A, Figueiredo C, Immenschuh S, Blasczyk R, Eiz-Vesper B. Closing the gap: discrimination of the expression profile of HLA questionable alleles by a cytokine-induced secretion approach using HLA-A*32:11Q. ACTA ACUST UNITED AC 2012; 79:340-50. [PMID: 22489943 DOI: 10.1111/j.1399-0039.2012.01864.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Matching of human leukocyte antigen (HLA) alleles between donors and recipients plays a major role in hematopoietic stem cell transplantation (HSCT). Null or questionably expressed HLA allelic variants are a major issue in HLA matching, because the aberrant expression of such alleles can have a major impact on the outcome of HSCT and/or its complications such as graft-versus-host disease. The goal of this study was to investigate the potential of a recently developed cytokine-induced secretion assay to differentiate the expression levels of HLA-A*32:11Q (questionable) into a null (N) or low (L) expression variant. An amino acid mutation at position 164 of HLA-A*32:11Q disrupts the disulfide bridge in the α2 domain. HLA-A*32:11Q is not detectable by standard microlymphocytotoxicity assay. To this end, we cloned soluble HLA-A*32:11Q and a reference allele (HLA-A*32:01) into expression vectors and transfected/transduced HEK293 and K562 cells. Allele-expressing K562 cells were simultaneously transfected/transduced with a β2-microglobulin (B2M)-encoding vector to ensure the intact HLA structure with B2M. After treatment with proinflammatory cytokines, secreted soluble HLA molecules were determined by enzyme-linked immunosorbent assay in the supernatant and intracellular accumulation of the recombinant proteins by flow cytometry. HLA-A*32:11Q was nearly undetectable in untreated transfectants. Cytokine treatment increased the secretion of HLA-A*32:11Q to detectable levels and resulted in intracellular accumulation of the allele. There was no difference in mRNA transcription between the A*32 alleles. On the basis of these results, we recommend reclassification of HLA-A*32:11Q as a low expression (L) variant.
Collapse
Affiliation(s)
- D Föll
- Institute for Transfusion Medicine, Hannover Medical School, Hannover, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Equine herpesvirus type 4 UL56 and UL49.5 proteins downregulate cell surface major histocompatibility complex class I expression independently of each other. J Virol 2012; 86:8059-71. [PMID: 22623773 DOI: 10.1128/jvi.00891-12] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Major histocompatibility complex class I (MHC-I) molecules are critically important in the host defense against various pathogens through presentation of viral peptides to cytotoxic T lymphocytes (CTLs), a process resulting in the destruction of virus-infected cells. Herpesviruses interfere with CTL-mediated elimination of infected cells by various mechanisms, including inhibition of peptide transport and loading, perturbation of MHC-I trafficking, and rerouting and proteolysis of cell surface MHC-I. In this study, we show that equine herpesvirus type 4 (EHV-4) modulates MHC-I cell surface expression through two different mechanisms. First, EHV-4 can lead to a significant downregulation of MHC-I expression at the cell surface through the product of ORF1, a protein expressed with early kinetics from a gene that is homologous to herpes simplex virus 1 UL56. The EHV-4 UL56 protein reduces cell surface MHC-I as early as 4 h after infection. Second, EHV-4 can interfere with MHC-I antigen presentation, starting at 6 h after infection, by inhibition of the transporter associated with antigen processing (TAP) through its UL49.5 protein. Although pUL49.5 has no immediate effect on overall surface MHC-I levels in infected cells, it blocks the supply of antigenic peptides to the endoplasmic reticulum (ER) and transport of peptide-loaded MHC-I to the cell surface. Taken together, our results show that EHV-4 encodes at least two viral immune evasion proteins: pUL56 reduces MHC-I molecules on the cell surface at early times after infection, and pUL49.5 interferes with MHC-I antigen presentation by blocking peptide transport in the ER.
Collapse
|
27
|
Van Hateren A, James E, Bailey A, Phillips A, Dalchau N, Elliott T. The cell biology of major histocompatibility complex class I assembly: towards a molecular understanding. ACTA ACUST UNITED AC 2011; 76:259-75. [PMID: 21050182 DOI: 10.1111/j.1399-0039.2010.01550.x] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Major histocompatibility complex class I (MHC I) proteins protect the host from intracellular pathogens and cellular abnormalities through the binding of peptide fragments derived primarily from intracellular proteins. These peptide-MHC complexes are displayed at the cell surface for inspection by cytotoxic T lymphocytes. Here we reveal how MHC I molecules achieve this feat in the face of numerous levels of quality control. Among these is the chaperone tapasin, which governs peptide selection in the endoplasmic reticulum as part of the peptide-loading complex, and we propose key amino acid interactions central to the peptide selection mechanism. We discuss how the aminopeptidase ERAAP fine-tunes the peptide repertoire available to assembling MHC I molecules, before focusing on the journey of MHC I molecules through the secretory pathway, where calreticulin provides additional regulation of MHC I expression. Lastly we discuss how these processes culminate to influence immune responses.
Collapse
Affiliation(s)
- A Van Hateren
- Faculty of Medicine, Southampton General Hospital, University of Southampton, Southampton, UK
| | | | | | | | | | | |
Collapse
|
28
|
Immune-induced evolutionary selection focused on a single reading frame in overlapping hepatitis B virus proteins. J Virol 2011; 85:4558-66. [PMID: 21307195 DOI: 10.1128/jvi.02142-10] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Viruses employ various means to evade immune detection. Reduction of CD8(+) T cell epitopes is one of the common strategies used for this purpose. Hepatitis B virus (HBV), a member of the Hepadnaviridae family, has four open reading frames, with about 50% overlap between the genes they encode. We computed the CD8(+) T cell epitope density within HBV proteins and the mutations within the epitopes. Our results suggest that HBV accumulates escape mutations that reduce the number of epitopes. These mutations are not equally distributed among genes and reading frames. While the highly expressed core and X proteins are selected to have low epitope density, polymerase, which is expressed at low levels, does not undergo the same selection. In overlapping regions, mutations in one protein-coding sequence also affect the other protein-coding sequence. We show that mutations lead to the removal of epitopes in X and surface proteins even at the expense of the addition of epitopes in polymerase. The total escape mutation rate for overlapping regions is lower than that for nonoverlapping regions. The lower epitope replacement rate for overlapping regions slows the evolutionary escape rate of these regions but leads to the accumulation of mutations more robust in the transfer between hosts, such as mutations preventing proteasomal cleavage into epitopes.
Collapse
|
29
|
Zhang Y, Liu Y, Lu N, Shan NN, Zheng GX, Zhao SM, Zou X, Gao YJ, Li YJ, Wang YS. Expression of the genes encoding human leucocyte antigens-A, -B, -DP, -DQ and -G in gastric cancer patients. J Int Med Res 2010; 38:949-56. [PMID: 20819431 DOI: 10.1177/147323001003800321] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
This study compared the expression of the genes encoding human leucocyte antigens (HLA)-A, -B, -DP, -DR and -G in peripheral blood mononuclear cells (PBMCs) in gastric cancer patients and healthy controls. Using reverse transcription-polymerase chain reaction, levels of classical HLA-A, -B, -DP and -DR and non-classical HLA-G mRNA were studied in 43 gastric cancer patients and 22 controls. In addition, the levels of HLA-A,B,C and -G antigens on the surface of PBMCs were measured in 30 gastric cancer patients and 15 controls using flow cytometry. The mean fluorescence intensity of HLA-A,B,C antigen in the gastric cancer group was significantly lower than in controls. The HLA-G antigen was mainly present on CD4(+)CD8(-) T-lymphocytes. The percentage of CD4(+)CD8(-) T-lymphocytes positive for HLA-G antigen was significantly lower in the gastric cancer group compared with the healthy controls. Levels of HLA-A, -B and -G mRNA in the gastric cancer group were significantly lower than in controls. The HLA-G mRNA levels were significantly lower in gastric cancer of histological grades III and IV than in grades I and II. These data may provide a novel diagnostic and research tool for gastric cancer.
Collapse
Affiliation(s)
- Y Zhang
- Department of Clinical Laboratory Medicine, Qilu Hospital of Shandong University, Jinan, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Jin YC, Wei P, Wei XX, Zhao ZY, Li Y. Rapid detection of BF haplotypes by a semi-nested polymerase chain reaction, which causes resistance/susceptibility to Marek's disease in chicken. Scand J Immunol 2010; 72:94-7. [PMID: 20618767 DOI: 10.1111/j.1365-3083.2010.02415.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
A semi-nested polymerase chain reaction (snPCR) assay was developed for the rapid detection of resistant/susceptible BF haplotypes to Marek's disease (MD) using the cDNA samples from peripheral blood leucocytes, liver, spleen and heart from Xiayan homozygous chickens: A(11), C(23), D(8) and D(12) (resistant to MD), A(5) and B(21) (susceptible to MD). The snPCR was utilized to span alternative splicing-out of the sequence encoding the second segment of the cytoplasmic part of the mature BF molecules (exon 7). This alternative exon 7 splice variant was detected in BF*A(5) and BF*B(21) (susceptible to MD), but not in the MD-resistant BF*A(11), BF*C(23), BF*D(8) and BF*D(12) haplotypes, suggesting a potential role of exon 7 for the detection of resistant/susceptible BF haplotypes to MD.
Collapse
Affiliation(s)
- Y-C Jin
- Poultry Science and Health, Guangxi University, Nanning, China
| | | | | | | | | |
Collapse
|
31
|
Improved transplantation outcome by epigenetic changes. Transpl Immunol 2010; 23:104-10. [DOI: 10.1016/j.trim.2010.05.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2009] [Revised: 05/01/2010] [Accepted: 05/04/2010] [Indexed: 01/31/2023]
|
32
|
Fu H, Liu C, Flutter B, Tao H, Gao B. Calreticulin maintains the low threshold of peptide required for efficient antigen presentation. Mol Immunol 2009; 46:3198-206. [PMID: 19748124 DOI: 10.1016/j.molimm.2009.08.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2009] [Revised: 08/11/2009] [Accepted: 08/12/2009] [Indexed: 11/26/2022]
Abstract
Calreticulin (CRT) plays a critical role in MHC class I antigen processing and elicits peptide-specific CD8(+) T cell responses against tumours when administered with peptides. However, how CRT contributes to class I antigen processing and the mechanism of its adjuvant effect in anti-tumour responses, remain to be elucidated. Here we show that reduced class I expression in CRT deficient cells can be restored by the direct delivery of peptides into the ER or by incubation at low temperature. CRT deficient cells exhibited a TAP-deficient phenotype in terms of class I assembly, without loss of TAP expression or functionality. Furthermore, a higher concentration of antigen in the cytosol is required for specific T cell stimulation, suggesting that CRT has a functional role in the maintenance of the low peptide concentration threshold required in the ER for efficient antigen presentation. In the absence of CRT, ERp57 is up-regulated, which indicates that they collaborate with each other in class I antigen processing.
Collapse
Affiliation(s)
- Hongmei Fu
- Rheumatology Unit, UCL Institute of Child Health, University College London, London, UK
| | | | | | | | | |
Collapse
|
33
|
Lee S, Park B, Kang K, Ahn K. Redox-regulated export of the major histocompatibility complex class I-peptide complexes from the endoplasmic reticulum. Mol Biol Cell 2009; 20:3285-94. [PMID: 19477919 DOI: 10.1091/mbc.e09-03-0238] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
In contrast to the fairly well-characterized mechanism of assembly of MHC class I-peptide complexes, the disassembly mechanism by which peptide-loaded MHC class I molecules are released from the peptide-loading complex and exit the endoplasmic reticulum (ER) is poorly understood. Optimal peptide binding by MHC class I molecules is assumed to be sufficient for triggering exit of peptide-filled MHC class I molecules from the ER. We now show that protein disulfide isomerase (PDI) controls MHC class I disassembly by regulating dissociation of the tapasin-ERp57 disulfide conjugate. PDI acts as a peptide-dependent molecular switch; in the peptide-bound state, it binds to tapasin and ERp57 and induces dissociation of the tapasin-ERp57 conjugate. In the peptide-free state, PDI is incompetent to bind to tapasin or ERp57 and fails to dissociate the tapasin-ERp57 conjugates, resulting in ER retention of MHC class I molecules. Thus, our results indicate that even after optimal peptide loading, MHC class I disassembly does not occur by default but, rather, is a regulated process involving PDI-mediated interactions within the peptide-loading complex.
Collapse
Affiliation(s)
- Sungwook Lee
- National Creative Research Center for Antigen Presentation, Department of Biological Sciences, Seoul National University, Seoul 151-747, Korea
| | | | | | | |
Collapse
|
34
|
Almani M, Raffaeli S, Vider-Shalit T, Tsaban L, Fishbain V, Louzoun Y. Human self-protein CD8+ T-cell epitopes are both positively and negatively selected. Eur J Immunol 2009; 39:1056-65. [PMID: 19291702 DOI: 10.1002/eji.200838353] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The cellular immune system recognizes self-epitopes in the context of MHC-I molecules. The immunological general view presumes that these self-epitopes are just a background, both positively and negatively selecting T cells. We here estimate the number of epitopes in each human protein for many frequent HLA alleles, and a score representing over or under presentation of epitopes on these proteins. We further show that there is a clear selection for the presentation of specific self-protein types. Proteins presenting many epitopes include, for example, autoimmune regulator (AIRE) upregulated tissue-specific antigens, immune system receptors and proteins with a high expression level. On the other hand, proteins that may be considered less "useful" for the immune system, such as low expression level proteins, are under-presented. We combine our epitope estimate with single nucleotide polymorphism (SNP) measures to show that this selection can be directly observed through the fraction of non-synonymous SNP (replacement fraction), which is significantly higher inside epitopes than outside.
Collapse
Affiliation(s)
- Michal Almani
- Math Department, Bar Ilan University, Ramat Gan, Israel
| | | | | | | | | | | |
Collapse
|
35
|
Kim Y, Kang K, Kim I, Lee YJ, Oh C, Ryoo J, Jeong E, Ahn K. Molecular mechanisms of MHC class I-antigen processing: redox considerations. Antioxid Redox Signal 2009; 11:907-36. [PMID: 19178136 DOI: 10.1089/ars.2008.2316] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Major histocompatibility complex (MHC) class I molecules present antigenic peptides to the cell surface for screening by CD8(+) T cells. A number of ER-resident chaperones assist the assembly of peptides onto MHC class I molecules, a process that can be divided into several steps. Early folding of the MHC class I heavy chain is followed by its association with beta(2)-microglobulin (beta(2)m). The MHC class I heavy chain-beta(2)m heterodimer is incorporated into the peptide-loading complex, leading to peptide loading, release of the peptide-filled MHC class I molecules from the peptide-loading complex, and exit of the complete MHC class I complex from the ER. Because proper antigen presentation is vital for normal immune responses, the assembly of MHC class I molecules requires tight regulation. Emerging evidence indicates that thiol-based redox regulation plays critical roles in MHC class I-restricted antigen processing and presentation, establishing an unexpected link between redox biology and antigen processing. We review the influences of redox regulation on antigen processing and presentation. Because redox signaling pathways are a rich source of validated drug targets, newly discovered redox biology-mediated mechanisms of antigen processing may facilitate the development of more selective and therapeutic drugs or vaccines against immune diseases.
Collapse
Affiliation(s)
- Youngkyun Kim
- National Creative Research Center for Antigen Presentation, Department of Biological Sciences, Seoul National University, Seoul, South Korea
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Adams SC, Xing Z, Li J, Cardona CJ. Immune-related gene expression in response to H11N9 low pathogenic avian influenza virus infection in chicken and Pekin duck peripheral blood mononuclear cells. Mol Immunol 2009; 46:1744-9. [PMID: 19250679 DOI: 10.1016/j.molimm.2009.01.025] [Citation(s) in RCA: 99] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2008] [Accepted: 01/29/2009] [Indexed: 01/08/2023]
Abstract
The duck and chicken are important hosts of avian influenza virus (AIV) with distinctive responses to infection. Frequently, AIV infections in ducks are asymptomatic and long-lasting in contrast to the clinically apparent and transient infections observed in chickens. These differences may be due in part to the host response to AIV infection. Using real-time quantitative PCR, we examined the expression of immune-related genes in response to low pathogenic AIV H11N9 infection in peripheral blood mononuclear cells (PBMC) isolated from the blood of chickens and Pekin ducks. While chicken PBMC expressed IL-1beta and IL-6 at high levels similar to mammalian species, duck PBMC expression levels were minimal or unchanged. Similarly, duck IFN-beta expression was nearly unaffected, whereas chicken expression was highly upregulated. Chicken IFN-gamma was expressed to higher levels than duck IFN-gamma, while IFN-alpha was expressed similarly by both species. IL-2 was elevated early in infection in duck PBMC, but returned to baseline levels by the end of the experiment; in contrast, IL-2 was weakly induced in chicken PBMC at late time points. TLR-7 and MHC class I molecule expressions were conserved between species, whereas duck MHC class II expression was downregulated and chicken expression was unchanged. These results show distinct PBMC expression patterns of pro-inflammatory cytokines and IFNs between species. The differences in pro-inflammatory cytokine and IFN expression reflect the asymptomatic and lasting infection observed in ducks and the tendency towards clinical signs and rapid clearance seen in chickens. These results highlight important differences in the host response to AIV of two species thought to be critical in the genesis and maintenance of epidemic strains of AIV.
Collapse
Affiliation(s)
- Sean C Adams
- Department of Population Health and Reproduction, University of California, Davis, Davis, CA 95616, USA
| | | | | | | |
Collapse
|
37
|
Montagnaro S, Longo M, Pacilio M, Indovina P, Roberti A, De Martino L, Iovane G, Pagnini U. Feline herpesvirus-1 down-regulates MHC class I expression in an homologous cell system. J Cell Biochem 2009; 106:179-85. [PMID: 19009565 DOI: 10.1002/jcb.21986] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Cytotoxic T lymphocytes (CTLs) are an essential component of the immune defense against many virus infections. CTLs recognize viral peptides in the context of the major histocompatibility complex (MHC) class I molecules on the surface of infected cells. Many viruses have evolved mechanisms to interfere with MHC class I expression as a means of evading the host immune response. In the present research we have studied the effect of in vitro Feline Herpesvirus 1 (FeHV-1) infection on MHC class I expression. The results of this study demonstrate that FeHV-1 down regulates surface expression of MHC class I molecules on infected cells, presumably to evade cytotoxic T-cell recognition and, perhaps, attenuate induction of immunity. Sensitivity to UV irradiation and insensitivity to a viral DNA synthesis inhibitor, like phosphonacetic acid, revealed that immediate early or early viral gene(s) are responsible. Use of the protein translation inhibitor cycloheximide confirmed that an early gene is primarily responsible.
Collapse
Affiliation(s)
- S Montagnaro
- Department of Pathology and animal Health, School of Veterinary Medicine, University of Naples Federico II, Via Delpino no 1, 80137 Naples, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Beta2-microglobulin-dependent bacterial clearance and survival during murine Klebsiella pneumoniae bacteremia. Infect Immun 2008; 77:360-6. [PMID: 18981251 DOI: 10.1128/iai.00909-08] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Klebsiella pneumoniae is a leading cause of both community-acquired and nosocomial gram-negative bacterial pneumonia. A significant clinical complication of Klebsiella pulmonary infections is peripheral blood dissemination, resulting in a systemic infection concurrent with the localized pulmonary infection. We report here on the critical importance of beta(2)-microglobulin expression during murine K. pneumoniae bacteremia. Beta(2)-microglobulin knockout mice displayed significantly increased mortality upon intravenous inoculation that correlated with increased bacterial burden in the blood, liver, and spleen. As beta(2)-microglobulin knockout mice lack both CD8(+) T cells and invariant NK T cells, mouse models specifically deficient in either cell population were examined to see if this would account for the increased mortality noted in beta(2)-microglobulin knockout mice. Surprisingly, neither CD8 T-cell-deficient (TAP-1 knockout; in vivo anti-CD8 antibody treatment) nor invariant NK (iNK) T-cell-deficient (CD1d knockout, J alpha281 knockout) mice were more susceptible to K. pneumoniae bacteremia. Combined, these studies clearly indicate the importance of a beta(2)-microglobulin-dependent but CD8 T-cell- and iNK T-cell-independent mechanism critical for survival during K. pneumoniae bacteremia.
Collapse
|
39
|
CD40 induces antigen transporter and immunoproteasome gene expression in carcinomas via the coordinated action of NF-kappaB and of NF-kappaB-mediated de novo synthesis of IRF-1. Mol Cell Biol 2008; 28:6208-22. [PMID: 18694960 DOI: 10.1128/mcb.00611-08] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Cancer cells may evade immune surveillance as a result of defective antigen processing and presentation. In this study, we demonstrate that CD40 ligation overcomes this defect through the coordinated action of the transcription factors NF-kappaB and interferon regulatory factor 1 (IRF-1). We show that unlike interferon signaling, which triggers the STAT1-mediated transcriptional activation of IRF-1, the ligation of CD40 in carcinomas induces the rapid upregulation of IRF-1 in a STAT1-independent but NF-kappaB-dependent manner. The transcriptional activation of IRF-1 is controlled largely by the recruitment of p65 (RelA) NF-kappaB to the IRF-1 promoter following the engagement of a TAK1/IkappaB kinase beta/IkappaBalpha signaling pathway downstream of CD40. NF-kappaB and de novo-synthesized IRF-1 converge to regulate the expression of genes involved in antigen processing and transport, as evident from the sequential recruitment of NF-kappaB and IRF-1 to the promoters of the genes encoding transporter for antigen processing 1 (TAP1), TAP2, tapasin, and low-molecular-mass polypeptides LMP2 and LMP10. Moreover, the RNA interference-mediated knockdown of IRF-1 reduced, whereas the inhibition of NF-kappaB abolished, the effects of CD40 on TAP1 and LMP2 upregulation in carcinoma cells. Collectively, these data reveal a novel "feed-forward" mechanism induced by NF-kappaB which ensures that acutely synthesized IRF-1 operates in concert with NF-kappaB to amplify the immunoproteasome and antigen-processing functions of CD40.
Collapse
|
40
|
Chessman D, Kostenko L, Lethborg T, Purcell AW, Williamson NA, Chen Z, Kjer-Nielsen L, Mifsud NA, Tait BD, Holdsworth R, Almeida CA, Nolan D, Macdonald WA, Archbold JK, Kellerher AD, Marriott D, Mallal S, Bharadwaj M, Rossjohn J, McCluskey J. Human leukocyte antigen class I-restricted activation of CD8+ T cells provides the immunogenetic basis of a systemic drug hypersensitivity. Immunity 2008; 28:822-32. [PMID: 18549801 DOI: 10.1016/j.immuni.2008.04.020] [Citation(s) in RCA: 236] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2007] [Revised: 03/25/2008] [Accepted: 04/01/2008] [Indexed: 10/22/2022]
Abstract
The basis for strong immunogenetic associations between particular human leukocyte antigen (HLA) class I allotypes and inflammatory conditions like Behçet's disease (HLA-B51) and ankylosing spondylitis (HLA-B27) remain mysterious. Recently, however, even stronger HLA associations are reported in drug hypersensitivities to the reverse-transcriptase inhibitor abacavir (HLA-B57), the gout prophylactic allopurinol (HLA-B58), and the antiepileptic carbamazepine (HLA-B*1502), providing a defined disease trigger and suggesting a general mechanism for these associations. We show that systemic reactions to abacavir were driven by drug-specific activation of cytokine-producing, cytotoxic CD8+ T cells. Recognition of abacavir required the transporter associated with antigen presentation and tapasin, was fixation sensitive, and was uniquely restricted by HLA-B*5701 and not closely related HLA allotypes with polymorphisms in the antigen-binding cleft. Hence, the strong association of HLA-B*5701 with abacavir hypersensitivity reflects specificity through creation of a unique ligand as well as HLA-restricted antigen presentation, suggesting a basis for the strong HLA class I-association with certain inflammatory disorders.
Collapse
Affiliation(s)
- Diana Chessman
- Department of Microbiology & Immunology, The University of Melbourne, Parkville, Victoria 3010, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Epel M, Carmi I, Soueid-Baumgarten S, Oh S, Bera T, Pastan I, Berzofsky J, Reiter Y. Targeting TARP, a novel breast and prostate tumor-associated antigen, with T cell receptor-like human recombinant antibodies. Eur J Immunol 2008; 38:1706-20. [PMID: 18446790 PMCID: PMC2682370 DOI: 10.1002/eji.200737524] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
MHC class I molecules are important components of immune surveillance. There are no available methods to directly visualize and determine the quantity and distribution of MHC/peptide complexes on individual cells or to detect such complexes on antigen-presenting cells in tissues. MHC-restricted recombinant antibodies with the same specificity of T cell receptors (TCR) may become a valuable tool to address these questions. They may also serve as valuable targeting molecules that mimic the specificity of cytotoxic T cells. We isolated by phage display a panel of human recombinant Fab antibodies with peptide-specific, MHC-restricted TCR-like reactivity directed toward HLA-A2-restricted T cell epitopes derived from a novel antigen termed TCRgamma alternative reading frame protein (TARP) which is expressed on prostate and breast cancer cells. We have characterized one of these recombinant antibodies and demonstrated its capacity to directly detect specific HLA-A2/TARP T cell epitopes on antigen-presenting cells that have complexes formed by naturally occurring active intracellular processing of the antigen, as well as on the surface of tumor cells. Moreover, by genetic fusion we armed the TCR-like antibody with a potent toxin and demonstrated that it can serve as a targeting moiety killing tumor cells in a peptide-specific, MHC-restricted manner similar to cytotoxic T lymphocytes.
Collapse
MESH Headings
- ADP Ribose Transferases/administration & dosage
- ADP Ribose Transferases/genetics
- ADP Ribose Transferases/pharmacology
- ADP Ribose Transferases/therapeutic use
- Amino Acid Substitution
- Animals
- Antibodies, Monoclonal/biosynthesis
- Antibodies, Monoclonal/immunology
- Antigens, Neoplasm/immunology
- Antineoplastic Agents/administration & dosage
- Antineoplastic Agents/pharmacokinetics
- Antineoplastic Agents/therapeutic use
- Bacterial Toxins/administration & dosage
- Bacterial Toxins/genetics
- Bacterial Toxins/pharmacology
- Bacterial Toxins/therapeutic use
- Breast Neoplasms/immunology
- Breast Neoplasms/pathology
- Cell Line, Tumor
- Epitopes, T-Lymphocyte/biosynthesis
- Epitopes, T-Lymphocyte/genetics
- Epitopes, T-Lymphocyte/immunology
- Exotoxins/administration & dosage
- Exotoxins/genetics
- Exotoxins/pharmacology
- Exotoxins/therapeutic use
- Female
- HLA-A2 Antigen/genetics
- HLA-A2 Antigen/immunology
- Humans
- Immunoglobulin Fab Fragments/biosynthesis
- Immunoglobulin Fab Fragments/genetics
- Immunoglobulin Fab Fragments/immunology
- Immunotoxins/immunology
- Immunotoxins/pharmacology
- Immunotoxins/therapeutic use
- Male
- Mammary Neoplasms, Experimental/immunology
- Mammary Neoplasms, Experimental/pathology
- Mammary Neoplasms, Experimental/prevention & control
- Mice
- Mice, Nude
- Molecular Sequence Data
- Nuclear Proteins/genetics
- Nuclear Proteins/immunology
- Peptide Fragments/biosynthesis
- Peptide Fragments/genetics
- Peptide Fragments/immunology
- Prostatic Neoplasms/immunology
- Prostatic Neoplasms/pathology
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Recombinant Proteins/biosynthesis
- Recombinant Proteins/immunology
- Virulence Factors/administration & dosage
- Virulence Factors/genetics
- Virulence Factors/pharmacology
- Virulence Factors/therapeutic use
- Xenograft Model Antitumor Assays
- Pseudomonas aeruginosa Exotoxin A
Collapse
Affiliation(s)
- Malka Epel
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Huang S, Gilfillan S, Kim S, Thompson B, Wang X, Sant AJ, Fremont DH, Lantz O, Hansen TH. MR1 uses an endocytic pathway to activate mucosal-associated invariant T cells. ACTA ACUST UNITED AC 2008; 205:1201-11. [PMID: 18443227 PMCID: PMC2373850 DOI: 10.1084/jem.20072579] [Citation(s) in RCA: 119] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Like CD1d-restricted iNKT cells, mucosal-associated invariant T cells (MAITs) are "innate" T cells that express a canonical TCRalpha chain, have a memory phenotype, and rapidly secrete cytokines upon TCR ligation. Unlike iNKT cells, MAIT cells require the class Ib molecule MHC-related protein I (MR1), B cells, and gut flora for development and/or expansion, and they preferentially reside in the gut lamina propria. Evidence strongly suggests that MAIT cell activation is ligand-dependent, but the nature of MR1 ligand is unknown. In this study, we define a mechanism of endogenous antigen presentation by MR1 to MAIT cells. MAIT cell activation was dependent neither on a proteasome-processed ligand nor on the chaperoning by the MHC class I peptide loading complex. However, MAIT cell activation was enhanced by overexpression of MHC class II chaperones Ii and DM and was strikingly diminished by silencing endogenous Ii. Furthermore, inhibiting the acidification of the endocytic compartments reduced MR1 surface expression and ablated MAIT cell activation. The importance of the late endosome for MR1 antigen presentation was further corroborated by the localization of MR1 molecules in the multivesicular endosomes. These findings demonstrate that MR1 traffics through endocytic compartments, thereby allowing MAIT cells to sample both endocytosed and endogenous antigens.
Collapse
Affiliation(s)
- Shouxiong Huang
- Department of Pathology and Immunology, Washington University, St. Louis, MO 63110, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Murakami T, Sato A, Chun NAL, Hara M, Naito Y, Kobayashi Y, Kano Y, Ohtsuki M, Furukawa Y, Kobayashi E. Transcriptional modulation using HDACi depsipeptide promotes immune cell-mediated tumor destruction of murine B16 melanoma. J Invest Dermatol 2008; 128:1506-16. [PMID: 18185535 DOI: 10.1038/sj.jid.5701216] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
With melanoma, as with many other malignancies, aberrant transcriptional repression is a hallmark of refractory cancer. To restore gene expression, use of a histone deacetylase inhibitor (HDACi) is expected to be effective. Our recent DNA micro-array analysis showed that the HDACi depsipeptide (FK228) significantly enhances gp100 antigen expression. Herein, we demonstrate that depsipeptide promotes tumor-specific T-cell-mediated killing of B16/F10 murine melanoma cells. First, by a quantitative assay of caspase-3/7 activity, a sublethal dose of depsipeptide was determined (ED50: 5 nM), in which p21(Waf1/Cip1) and Fas were sufficiently evoked concomitantly with histone H3 acetylation. Second, the sublethal dose of depsipeptide treatment with either a recombinant Fas ligand or tumor-specific T cells synergistically enhanced apoptotic cell death in B16/F10 cells in vitro. Furthermore, we found that depsipeptide increased levels of perforin in T cells. Finally, in vivo metastatic growth of B16/F10 in the lung was significantly inhibited by a combination of depsipeptide treatment and immune cell adoptive transfer from immunized mice using irradiated B16 cells and gp100-specific (Pmel-1) TCR transgenic mice (P<0.05, vs cell transfer alone). Consequently, employment of a transcriptional modulation strategy using HDACis might prove to be a useful pretreatment for human melanoma immunotherapy.
Collapse
Affiliation(s)
- Takashi Murakami
- Division of Organ Replacement Research, Center for Molecular Medicine, Jichi Medical University, Shimotuke, Tochigi, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Newcomb EW, Demaria S, Lukyanov Y, Shao Y, Schnee T, Kawashima N, Lan L, Dewyngaert JK, Zagzag D, McBride WH, Formenti SC. The combination of ionizing radiation and peripheral vaccination produces long-term survival of mice bearing established invasive GL261 gliomas. Clin Cancer Res 2007; 12:4730-7. [PMID: 16899624 DOI: 10.1158/1078-0432.ccr-06-0593] [Citation(s) in RCA: 112] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE High-grade glioma treatment includes ionizing radiation therapy. The high invasiveness of glioma cells precludes their eradication and is responsible for the dismal prognosis. Recently, we reported the down-regulation of MHC class I (MHC-I) products in invading tumor cells in human and mouse GL261 gliomas. Here, we tested the hypothesis that whole-brain radiotherapy (WBRT) up-regulates MHC-I expression on GL261 tumors and enhances the effectiveness of immunotherapy. EXPERIMENTAL DESIGN MHC-I molecule expression on GL261 cells was analyzed in vitro and in vivo by flow cytometry and immunohistochemistry, respectively. To test the response of established GL261 gliomas to treatment, mice with measurable (at CT imaging) brain tumors were randomly assigned to four groups receiving (a) no treatment, (b) WBRT in two fractions of 4 Gy, (c) vaccination with irradiated GL261 cells secreting granulocyte-macrophage colony-stimulating factor, or (d) WBRT and vaccination. Endpoints were tumor response and survival. RESULTS An ionizing radiation dose of 4 Gy maximally up-regulated MHC-I molecules on GL261 cells in vitro. In vivo, WBRT induced the expression of the beta2-microglobulin light chain subunit of the MHC class I complex on glioma cells invading normal brain and increased CD4+ and CD8+ T cell infiltration. However, the survival advantage obtained with WBRT or vaccination alone was minimal. In contrast, WBRT in combination with vaccination increased long-term survival to 40% to 80%, compared with 0% to 10% in the other groups (P < 0.002). Surviving animals showed antitumor immunity by rejecting challenge tumors. CONCLUSION Ionizing radiation can be successfully combined with peripheral vaccination for the treatment of established high-grade gliomas.
Collapse
Affiliation(s)
- Elizabeth W Newcomb
- Department of Pathology, New York University School of Medicine, New York, New York 10016, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
The clinical spectrum and immunopathogenesis of inflammatory myopathies are summarized with an update on possible triggering factors, cell degeneration, and emerging new therapies.
Collapse
Affiliation(s)
- Marinos C Dalakas
- Neuromuscular Diseases Section, US National Institute of Neurological Disorders and Stroke, US National Institutes of Health, Building 10, Room 4N248, Bethesda, MD 20892, USA.
| |
Collapse
|
46
|
Prpić Massari L, Kastelan M, Laskarin G, Zamolo G, Massari D, Rukavina D. Analysis of perforin expression in peripheral blood and lesions in severe and mild psoriasis. J Dermatol Sci 2007; 47:29-36. [PMID: 17412565 DOI: 10.1016/j.jdermsci.2007.02.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2006] [Revised: 02/21/2007] [Accepted: 02/22/2007] [Indexed: 11/24/2022]
Abstract
BACKGROUND Perforin is a membrane-disrupting protein that allows the entry of granzymes into a target cell inducing degradation of target substances in the cytoplasm and nucleus thus leading to programmed cell death or apoptosis. Recent work demonstrated a possible involvement of perforin mediated cytotoxicity in immunopathogenesis of psoriasis. OBJECTIVES To investigate a difference in systemic (peripheral blood) and local (lesions) expression and distribution of perforin in psoriatic patients with severe and mild disease. METHODS Flow cytometry was used for simultaneous detection of intracellular (perforin) and cell surface antigens in peripheral blood lymphocytes. The expression of perforin in skin lesions was evaluated by immunohistochemistry. RESULTS Significant increase of perforin expression in T lymphocytes, especially cytotoxic CD8+ cells was found in severe psoriasis compared to mild disease (p<0.01 and p<0.05, respectively). There was also an increase of CD56+P+ NK cells (p<0.05) in severe compared to mild psoriasis. The psoriatic plaque of both, severe and mild disease were abundant with perforin showing no significant difference on local level. CONCLUSION Based on our results we suggest the association between perforin expression and disease severity.
Collapse
Affiliation(s)
- Larisa Prpić Massari
- Department of Dermatovenerology, Clinical Hospital Centre Rijeka, University of Rijeka, Kresimirova 42, HR-51000, Rijeka, Croatia.
| | | | | | | | | | | |
Collapse
|
47
|
Papadopoulos M, Momburg F. Multiple residues in the transmembrane helix and connecting peptide of mouse tapasin stabilize the transporter associated with the antigen-processing TAP2 subunit. J Biol Chem 2007; 282:9401-9410. [PMID: 17244610 DOI: 10.1074/jbc.m610429200] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The type I endoplasmic reticulum (ER) glycoprotein tapasin (Tpn) is essential for loading of major histocompatibility complex class I (MHC-I) molecules with an optimal spectrum of antigenic peptides and for stable expression of the heterodimeric, polytopic TAP peptide transporter. In a detailed mutational analysis, the transmembrane domain (TMD) and ER-luminal connecting peptide (CP) of mouse Tpn were analyzed for their capacity to stabilize the TAP2 subunit. Replacement of the TMD of Tpn by TMDs from calnexin or the Tpn-related protein, respectively, completely abolished TAP2 stabilization after transfection of Tpn-deficient cells, whereas TMDs derived from distantly related Tpn molecules (chicken and fish) were functional. A detailed mutational analysis of the TMD and adjacent residues in the ER-luminal CP of mouse Tpn was performed to elucidate amino acids that control the stabilization of TAP2. Single amino acid substitutions, including a conserved Lys residue in the center of the putative TMD, did not affect TAP2 expression levels. Mutation of this Lys plus four additional residues, predicted to be neighbors in an assumed alpha-helical TMD arrangement, abrogated the TAP2-stabilizing capacity of Tpn. In the presence of a wild-type TMD, also the substitution of a highly conserved Glu residue in the CP of Tpn strongly affected TAP2 stabilization. Defective TAP2 stabilization resulted in impaired cell surface expression of MHC-I molecules. This study thus defines a novel, spatially arranged motif in the TMD of Tpn essential for stable expression of the TAP2 protein and a novel protein interaction mode involving an ER-luminal Glu residue close to the membrane.
Collapse
Affiliation(s)
- Martina Papadopoulos
- Department of Molecular Immunology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Frank Momburg
- Department of Molecular Immunology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| |
Collapse
|
48
|
Ladasky JJ, Boyle S, Seth M, Li H, Pentcheva T, Abe F, Steinberg SJ, Edidin M. Bap31 enhances the endoplasmic reticulum export and quality control of human class I MHC molecules. THE JOURNAL OF IMMUNOLOGY 2006; 177:6172-81. [PMID: 17056546 PMCID: PMC1978250 DOI: 10.4049/jimmunol.177.9.6172] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The assembly of class I MHC molecules and their export from the endoplasmic reticulum (ER) is governed by chaperones and accessory proteins. We present evidence that the putative cargo receptor protein Bap31 participates in the transport and the quality control of human class I molecules. Transfection of the human adenocarcinoma cell line HeLa with yellow fluorescent protein-Bap31 chimeras increased surface levels of class I in a dose-dependent manner, by as much as 3.7-fold. The increase in surface class I resulted from an increase in the rate of export of newly synthesized class I molecules to the cell surface and from an increase in the stability of the exported molecules. We propose that Bap31 performs quality control on class I molecules in two distinct phases: first, by exporting peptide-loaded class I molecules to the ER/Golgi intermediate compartment, and second, by retrieving class I molecules that have lost peptides in the acidic post-ER environment. This function of Bap31 is conditional or redundant, because we find that Bap31 deficiency does not reduce surface class I levels. Overexpression of the Bap31 homolog, Bap29, decreases surface class levels in HeLa, indicating that it does not substitute for Bap31.
Collapse
Affiliation(s)
- John J Ladasky
- Department of Biology, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, USA
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Vider-Shalit T, Raffaeli S, Louzoun Y. Virus-epitope vaccine design: informatic matching the HLA-I polymorphism to the virus genome. Mol Immunol 2006; 44:1253-61. [PMID: 16930710 DOI: 10.1016/j.molimm.2006.06.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2006] [Revised: 06/07/2006] [Accepted: 06/08/2006] [Indexed: 12/01/2022]
Abstract
Attempts to develop peptide vaccines, based on a limited number of peptides face two problems: HLA polymorphism and the high mutation rate of viral epitopes. We have developed a new genomic method that ensures maximal coverage and thus maximal applicability of the peptide vaccine. The same method also promises a large number of epitopes per HLA to prevent escape via mutations. Our design can be applied swiftly in order to face rapidly emerging viral diseases. We use a genomic scan of all candidate peptides and join them optimally. For a given virus, we use algorithms computing: peptide cleavage probability, transfer through TAP and MHC binding for a large number of HLA alleles. The resulting peptide libraries are pruned for peptides that are not conserved or are too similar to self peptides. We then use a genetic algorithm to produce an optimal protein composed of peptides from this list properly ordered for cleavage. The selected peptides represent an optimal combination to cover all HLA alleles and all viral proteins. We have applied this method to HCV and found that some HCV proteins (mainly envelope proteins) represent much less peptide than expected. A more detailed analysis of the peptide variability shows a balance between the attempts of the immune system to detect less mutating peptides, and the attempts of viruses to mutate peptides and avoid detection by the immune system. In order to show the applicability of our method, we have further used it on HIV-I, Influenza H3N2 and the Avian Flu Viruses.
Collapse
|
50
|
Hirv K, Pannicke U, Mytilineos J, Schwarz K. Disulfide Bridge Disruption in the α2 Domain of the HLA Class I Molecule Leads to Low Expression of the Corresponding Antigen. Hum Immunol 2006; 67:589-96. [PMID: 16916654 DOI: 10.1016/j.humimm.2006.04.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2005] [Indexed: 11/30/2022]
Abstract
Using sequence-based typing, we have identified a novel human leukocyte antigen (HLA)-A*30 allele, HLA-A*3014L, with a low expression pattern. The sequence of HLA-A*3014L is identical to that of HLA-A*3001 except for a G to C substitution in exon 3 at nucleotide position 563, resulting in an amino acid difference at position 164 (Cys to Ser). Due to the cysteine substitution, a disulfide bridge in the alpha2 domain of the HLA class I heavy chain cannot be formed. By using the standard microlymphocytotoxicity test, the HLA-A30 antigen cannot be detected. By flow cytometric analysis of the cell-surface expression at either 37 degrees C or 30 degrees C, a temperature-sensitive expression pattern of the HLA-A*3014L antigen was observed. Only by incubating the cells at 30 degrees C, which increases the stability of HLA class I heavy chains, was a weak but clearly detectable HLA-A*3014L expression found. The mRNA expression level of the HLA-A*3014L allele was not affected by the nucleotide substitution. The intrachain disulfide bond formation in the alpha2 domain is essential for the normal expression of the HLA molecules. Reduced protein expression is probably caused by incorrect HLA class I heavy chain folding and HLA class I complex assembly.
Collapse
Affiliation(s)
- Kaimo Hirv
- Department of Transplantation Immunology, Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, Germany.
| | | | | | | |
Collapse
|