51
|
Kar P, Ruiz-Perez L, Arooj M, Mancera RL. Current methods for the prediction of T-cell epitopes. Pept Sci (Hoboken) 2018. [DOI: 10.1002/pep2.24046] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Prattusha Kar
- School of Pharmacy and Biomedical Sciences; Curtin Health Innovation Research Institute and Curtin Institute for Computation, Curtin University; Perth Western Australia 6845 Australia
| | - Lanie Ruiz-Perez
- School of Pharmacy and Biomedical Sciences; Curtin Health Innovation Research Institute and Curtin Institute for Computation, Curtin University; Perth Western Australia 6845 Australia
| | - Mahreen Arooj
- School of Pharmacy and Biomedical Sciences; Curtin Health Innovation Research Institute and Curtin Institute for Computation, Curtin University; Perth Western Australia 6845 Australia
| | - Ricardo L. Mancera
- School of Pharmacy and Biomedical Sciences; Curtin Health Innovation Research Institute and Curtin Institute for Computation, Curtin University; Perth Western Australia 6845 Australia
| |
Collapse
|
52
|
Gravett AM, Trautwein N, Stevanović S, Dalgleish AG, Copier J. Gemcitabine alters the proteasome composition and immunopeptidome of tumour cells. Oncoimmunology 2018; 7:e1438107. [PMID: 29930882 PMCID: PMC5990974 DOI: 10.1080/2162402x.2018.1438107] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 01/30/2018] [Accepted: 02/02/2018] [Indexed: 01/05/2023] Open
Abstract
The antigenic makeup of tumour cells can have a profound effect on the progression of cancer and success of immunotherapies. Therefore, one strategy to improve the efficacy of cancer treatments is to augment the antigens displayed by tumours. The present study explores how the recognition of tumour cells may be altered by non-cytotoxic concentrations of gemcitabine (GEM). Testing a panel of chemotherapeutics in human cancer cell lines in vitro, it was found that GEM increased surface expression of HLA-A,B,C and that underlying this were specific increases in β-2-microglobulin and immunoproteasome subunit proteins. Furthermore, the peptide antigen repertoire displayed on HLA class I was altered, revealing a number of novel antigens, many of which that were derived from proteins involved in the DNA-damage response. Changes in the nature of the peptide antigens eluted from HLA-A,B,C after GEM treatment consisted of amino acid anchor-residue modifications and changes in peptide length which rendered peptides likely to favour alternative HLA-alleles and increased their predicted immunogenicity. Signalling through the MAPK/ERK and NFκB/RelB pathways was associated with these changes. These data may explain observations made in previous in vivo studies, advise as to which antigens should be used in future vaccination protocols and reinforce the idea that chemotherapy and immunotherapy could be used in combination.
Collapse
Affiliation(s)
- A M Gravett
- Institute for infection and immunity, St George's, University of London, London, UK
| | - N Trautwein
- Department of Immunology, Institute of Cell Biology, University of Tübingen, Tübingen, Germany
| | - S Stevanović
- Department of Immunology, Institute of Cell Biology, University of Tübingen, Tübingen, Germany
| | - A G Dalgleish
- Institute for infection and immunity, St George's, University of London, London, UK
| | - J Copier
- Institute for infection and immunity, St George's, University of London, London, UK
| |
Collapse
|
53
|
Iyer SP, Hunt CR, Pandita TK. Cross Talk between Radiation and Immunotherapy: The Twain Shall Meet. Radiat Res 2017; 189:219-224. [PMID: 29261410 DOI: 10.1667/rr14941.1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
There has been increased interest in the immune stimulatory properties of ionizing radiation based on several preclinical models and recently completed clinical studies performed in combination with checkpoint inhibitors. This is a paradigm shift in that it considers the role of radiation beyond its direct cytotoxic effects, however, the factors that promote or limit radiation-induced immunogenicity are still unclear. Here we review the role of radiation in modulating the various aspects of the tumor immune microenvironment and discuss in particular the direct effects of radiation on the DNA damage response and its immediate consequences to neighboring cells. The latter "danger response" in particular can enhance recruitment of dendritic and macrophage cells to the tumor microenvironment, which in turn can activate or diminish subsequent T-cell priming. Identification of the critical factors that modulate the interaction between radiation-induced cell damage and the immune system will allow for rational combinational therapy design and the development of biomarkers that predict effective immune responses.
Collapse
Affiliation(s)
- Swaminathan P Iyer
- a Department of Hematology, Houston Methodist Cancer Center, Weil Cornell Medical College, Houston Texas 77030
| | - Clayton R Hunt
- b Department of Radiation Oncology, The Houston Methodist Research Institute, Weil Cornell Medical College, Houston Texas 77030
| | - Tej K Pandita
- b Department of Radiation Oncology, The Houston Methodist Research Institute, Weil Cornell Medical College, Houston Texas 77030
| |
Collapse
|
54
|
The 20S immunoproteasome and constitutive proteasome bind with the same affinity to PA28αβ and equally degrade FAT10. Mol Immunol 2017; 113:22-30. [PMID: 29208314 DOI: 10.1016/j.molimm.2017.11.030] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 11/27/2017] [Accepted: 11/28/2017] [Indexed: 11/22/2022]
Abstract
The 20S immunoproteasome (IP) is an interferon(IFN)-γ - and tumor necrosis factor (TNF) -inducible variant of the 20S constitutive proteasome (CP) in which all its peptidolytically active subunits β1, β2, and β5 are replaced by their cytokine inducible homologues β1i (LMP2), β2i (MECL-1), and β5i (LMP7). These subunit replacements alter the cleavage specificity of the proteasome and the spectrum of proteasome-generated peptide ligands of MHC class I molecules. In addition to antigen processing, the IP has recently been shown to serve unique functions in the generation of pro-inflammatory T helper cell subtypes and cytokines as well as in the pathogenesis of autoimmune diseases, but the mechanistic involvement of the IP in these processes has remained elusive. In this study we investigated whether the IP differs from the CP in the interaction with two IFN-γ/TNF inducible factors: the 11S proteasome regulator PA28αβ and the ubiquitin-like modifier FAT10 (ubiquitin D). Using thermophoresis, we determined the affinity of PA28αβ for the CP and IP to be 12.2nM +/- 2.8nM and 15.3nM +/- 2.7nM, respectively, which is virtually identical. Also the activation of the peptidolytic activities of the IP and CP by PA28αβ did not differ. For FAT10 we determined the degradation kinetics in cycloheximide chase experiments in cells expressing almost exclusively IP or CP as well as in IFN-γ stimulated and unstimulated cells and found no differences between the degradation rates. Taken together, we conclude that neither differences in the binding strength to, nor activation by PA28αβ, nor a difference in the rate of FAT10-mediated degradation can account for distinct functional capabilities of the IP as compared to the CP.
Collapse
|
55
|
Koparde V, Abdul Razzaq B, Suntum T, Sabo R, Scalora A, Serrano M, Jameson-Lee M, Hall C, Kobulnicky D, Sheth N, Feltz J, Contaifer D, Wijesinghe D, Reed J, Roberts C, Qayyum R, Buck G, Neale M, Toor A. Dynamical system modeling to simulate donor T cell response to whole exome sequencing-derived recipient peptides: Understanding randomness in alloreactivity incidence following stem cell transplantation. PLoS One 2017; 12:e0187771. [PMID: 29194460 PMCID: PMC5711034 DOI: 10.1371/journal.pone.0187771] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 10/25/2017] [Indexed: 12/01/2022] Open
Abstract
Quantitative relationship between the magnitude of variation in minor histocompatibility antigens (mHA) and graft versus host disease (GVHD) pathophysiology in stem cell transplant (SCT) donor-recipient pairs (DRP) is not established. In order to elucidate this relationship, whole exome sequencing (WES) was performed on 27 HLA matched related (MRD), & 50 unrelated donors (URD), to identify nonsynonymous single nucleotide polymorphisms (SNPs). An average 2,463 SNPs were identified in MRD, and 4,287 in URD DRP (p<0.01); resulting peptide antigens that may be presented on HLA class I molecules in each DRP were derived in silico (NetMHCpan ver2.0) and the tissue expression of proteins these were derived from determined (GTex). MRD DRP had an average 3,670 HLA-binding-alloreactive peptides, putative mHA (pmHA) with an IC50 of <500 nM, and URD, had 5,386 (p<0.01). To simulate an alloreactive donor cytotoxic T cell response, the array of pmHA in each patient was considered as an operator matrix modifying a hypothetical cytotoxic T cell clonal vector matrix; each responding T cell clone’s proliferation was determined by the logistic equation of growth, accounting for HLA binding affinity and tissue expression of each alloreactive peptide. The resulting simulated organ-specific alloreactive T cell clonal growth revealed marked variability, with the T cell count differences spanning orders of magnitude between different DRP. Despite an estimated, uniform set of constants used in the model for all DRP, and a heterogeneously treated group of patients, higher total and organ-specific T cell counts were associated with cumulative incidence of moderate to severe GVHD in recipients. In conclusion, exome wide sequence differences and the variable alloreactive peptide binding to HLA in each DRP yields a large range of possible alloreactive donor T cell responses. Our findings also help understand the apparent randomness observed in the development of alloimmune responses.
Collapse
Affiliation(s)
- Vishal Koparde
- Center for the Study of Biological Complexity, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Badar Abdul Razzaq
- Bone Marrow Transplant Program, Department of Internal Medicine, Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Tara Suntum
- Bone Marrow Transplant Program, Department of Internal Medicine, Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Roy Sabo
- Department of Biostatistics, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Allison Scalora
- Bone Marrow Transplant Program, Department of Internal Medicine, Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Myrna Serrano
- Center for the Study of Biological Complexity, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Max Jameson-Lee
- Bone Marrow Transplant Program, Department of Internal Medicine, Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Charles Hall
- Bone Marrow Transplant Program, Department of Internal Medicine, Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - David Kobulnicky
- Bone Marrow Transplant Program, Department of Internal Medicine, Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Nihar Sheth
- Center for the Study of Biological Complexity, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Juliana Feltz
- Center for the Study of Biological Complexity, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Daniel Contaifer
- School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Dayanjan Wijesinghe
- School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Jason Reed
- Department of Physics, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Catherine Roberts
- Bone Marrow Transplant Program, Department of Internal Medicine, Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Rehan Qayyum
- Section of Hospital Medicine, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Gregory Buck
- Center for the Study of Biological Complexity, Virginia Commonwealth University, Richmond, Virginia, United States of America
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Michael Neale
- Departments of Psychiatry and Human & Molecular Genetics, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Amir Toor
- Bone Marrow Transplant Program, Department of Internal Medicine, Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia, United States of America
- * E-mail:
| |
Collapse
|
56
|
Zaky W, Manton C, Miller CP, Khatua S, Gopalakrishnan V, Chandra J. The ubiquitin-proteasome pathway in adult and pediatric brain tumors: biological insights and therapeutic opportunities. Cancer Metastasis Rev 2017; 36:617-633. [PMID: 29071526 DOI: 10.1007/s10555-017-9700-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Nearly 20 years ago, the concept of targeting the proteasome for cancer therapy began gaining momentum. This concept was driven by increased understanding of the biology/structure and function of the 26S proteasome, insight into the role of the proteasome in transformed cells, and the synthesis of pharmacological inhibitors with clinically favorable features. Subsequent in vitro, in vivo, and clinical testing culminated in the FDA approval of three proteasome inhibitors-bortezomib, carfilzomib, and ixazomib -for specific hematological malignancies. However, despite in vitro and in vivo studies pointing towards efficacy in solid tumors, clinical responses broadly have been evasive. For brain tumors, a malignancy in dire need of new approaches both in adult and pediatric patients, this has also been the case. Elucidation of proteasome-dependent processes in specific types of brain tumors, the evolution of newer proteasome targeting strategies, and the use of proteasome inhibitors in combination strategies will clarify how these agents can be leveraged more effectively to treat central nervous system malignancies. Since brain tumors represent a heterogeneous subset of solid tumors, and in particular, pediatric brain tumors possess distinct biology from adult brain tumors, tailoring of proteasome inhibitor-based strategies to specific subtypes of these tumors will be critical for advancing care for affected patients, and will be discussed in this review.
Collapse
Affiliation(s)
- Wafik Zaky
- Children's Cancer Hospital, Division of Pediatrics, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Christa Manton
- Children's Cancer Hospital, Division of Pediatrics, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Claudia P Miller
- Children's Cancer Hospital, Division of Pediatrics, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Soumen Khatua
- Children's Cancer Hospital, Division of Pediatrics, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Vidya Gopalakrishnan
- Children's Cancer Hospital, Division of Pediatrics, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Joya Chandra
- Children's Cancer Hospital, Division of Pediatrics, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA.
| |
Collapse
|
57
|
Winter MB, La Greca F, Arastu-Kapur S, Caiazza F, Cimermancic P, Buchholz TJ, Anderl JL, Ravalin M, Bohn MF, Sali A, O'Donoghue AJ, Craik CS. Immunoproteasome functions explained by divergence in cleavage specificity and regulation. eLife 2017; 6:e27364. [PMID: 29182146 PMCID: PMC5705213 DOI: 10.7554/elife.27364] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 11/11/2017] [Indexed: 12/16/2022] Open
Abstract
The immunoproteasome (iP) has been proposed to perform specialized roles in MHC class I antigen presentation, cytokine modulation, and T cell differentiation and has emerged as a promising therapeutic target for autoimmune disorders and cancer. However, divergence in function between the iP and the constitutive proteasome (cP) has been unclear. A global peptide library-based screening strategy revealed that the proteasomes have overlapping but distinct substrate specificities. Differing iP specificity alters the quantity of production of certain MHC I epitopes but does not appear to be preferentially suited for antigen presentation. Furthermore, iP specificity was found to have likely arisen through genetic drift from the ancestral cP. Specificity differences were exploited to develop isoform-selective substrates. Cellular profiling using these substrates revealed that divergence in regulation of the iP balances its relative contribution to proteasome capacity in immune cells, resulting in selective recovery from inhibition. These findings have implications for iP-targeted therapeutic development.
Collapse
Affiliation(s)
- Michael B Winter
- Department of Pharmaceutical ChemistryUniversity of California, San FranciscoSan FranciscoUnited States
| | - Florencia La Greca
- Department of Pharmaceutical ChemistryUniversity of California, San FranciscoSan FranciscoUnited States
| | - Shirin Arastu-Kapur
- Department of Pharmaceutical ChemistryUniversity of California, San FranciscoSan FranciscoUnited States
- Onyx PharmaceuticalsInc., an Amgen subsidiarySan FranciscoUnited States
| | - Francesco Caiazza
- Department of Pharmaceutical ChemistryUniversity of California, San FranciscoSan FranciscoUnited States
| | - Peter Cimermancic
- Department of Bioengineering and Therapeutic SciencesCalifornia Institute for Quantitative Biosciences, University of California, San FranciscoSan FranciscoUnited States
| | - Tonia J Buchholz
- Onyx PharmaceuticalsInc., an Amgen subsidiarySan FranciscoUnited States
| | - Janet L Anderl
- Onyx PharmaceuticalsInc., an Amgen subsidiarySan FranciscoUnited States
| | - Matthew Ravalin
- Department of Pharmaceutical ChemistryUniversity of California, San FranciscoSan FranciscoUnited States
| | - Markus F Bohn
- Department of Pharmaceutical ChemistryUniversity of California, San FranciscoSan FranciscoUnited States
| | - Andrej Sali
- Department of Pharmaceutical ChemistryUniversity of California, San FranciscoSan FranciscoUnited States
- Department of Bioengineering and Therapeutic SciencesCalifornia Institute for Quantitative Biosciences, University of California, San FranciscoSan FranciscoUnited States
| | - Anthony J O'Donoghue
- Skaggs School of Pharmacy and Pharmaceutical SciencesUniversity of California, San DiegoSan DiegoUnited States
| | - Charles S Craik
- Department of Pharmaceutical ChemistryUniversity of California, San FranciscoSan FranciscoUnited States
| |
Collapse
|
58
|
Basler M, Mundt S, Groettrup M. The immunoproteasome subunit LMP7 is required in the murine thymus for filling up a hole in the T cell repertoire. Eur J Immunol 2017; 48:419-429. [PMID: 29067678 DOI: 10.1002/eji.201747282] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 09/22/2017] [Accepted: 10/19/2017] [Indexed: 01/09/2023]
Abstract
Cells of hematopoietic origin express high levels of the immunoproteasome, a cytokine-inducible variant of the proteasome which has been implicated in regulating inflammatory responses and antigen presentation. In the thymus, medullary thymic epithelial cells (mTECs) and cortical thymic epithelial cells (cTECs) do express different proteasome subunits exerting chymotrypsin-like activities suggesting distinct functions in thymic T cell selection. Employing the lymphocytic choriomeningitis virus (LCMV) infection model, we could show that the immunoproteasome subunit LMP7 was absolutely required for the generation of LCMV GP118-125 -specific T cells although the class I mediated presentation of GP118-125 was not dependent on LMP7. Using bone marrow chimeras and adoptive transfer of LMP7-deficient CD8+ T cells into RAG1-deficient mice we show that LMP7-deficient mice lacked GP118-125 -specific T cell precursors and that LMP7 was required in radioresistant cells - most likely thymic epithelial cells - to enable their selection. Since LMP7 is strongly expressed in negatively selecting mTECs but barely in positively selecting cTECs our data suggest that LMP7 was required to avoid excessive negative selection of GP118-125 -specific T cell precursors. Taken together, this study demonstrates that the immunoproteasome is a crucial factor for filling up holes within the cytotoxic T cell repertoire.
Collapse
Affiliation(s)
- Michael Basler
- Biotechnology Institute Thurgau (BITg) at the University of Konstanz, CH-8280, Kreuzlingen, Switzerland.,Division of Immunology, Department of Biology, University of Konstanz, D-78457, Konstanz, Germany
| | - Sarah Mundt
- Division of Immunology, Department of Biology, University of Konstanz, D-78457, Konstanz, Germany
| | - Marcus Groettrup
- Biotechnology Institute Thurgau (BITg) at the University of Konstanz, CH-8280, Kreuzlingen, Switzerland.,Division of Immunology, Department of Biology, University of Konstanz, D-78457, Konstanz, Germany
| |
Collapse
|
59
|
Platteel ACM, Liepe J, van Eden W, Mishto M, Sijts AJAM. An Unexpected Major Role for Proteasome-Catalyzed Peptide Splicing in Generation of T Cell Epitopes: Is There Relevance for Vaccine Development? Front Immunol 2017; 8:1441. [PMID: 29163514 PMCID: PMC5675849 DOI: 10.3389/fimmu.2017.01441] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 10/17/2017] [Indexed: 02/06/2023] Open
Abstract
Efficient and safe induction of CD8+ T cell responses is a desired characteristic of vaccines against intracellular pathogens. To achieve this, a new generation of safe vaccines is being developed accommodating single, dominant antigens of pathogens of interest. In particular, the selection of such antigens is challenging, since due to HLA polymorphism the ligand specificities and immunodominance hierarchies of pathogen-specific CD8+ T cell responses differ throughout the human population. A recently discovered mechanism of proteasome-mediated CD8+ T cell epitope generation, i.e., by proteasome-catalyzed peptide splicing (PCPS), expands the pool of peptides and antigens, presented by MHC class I HLA molecules. On the cell surface, one-third of the presented self-peptides are generated by PCPS, which coincides with one-fourth in terms of abundance. Spliced epitopes are targeted by CD8+ T cell responses during infection and, like non-spliced epitopes, can be identified within antigen sequences using a novel in silico strategy. The existence of spliced epitopes, by enlarging the pool of peptides available for presentation by different HLA variants, opens new opportunities for immunotherapies and vaccine design.
Collapse
Affiliation(s)
- Anouk C M Platteel
- Faculty of Veterinary Medicine, Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, Netherlands
| | - Juliane Liepe
- Max-Planck-Institute for Biophysical Chemistry, Göttingen, Germany
| | - Willem van Eden
- Faculty of Veterinary Medicine, Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, Netherlands
| | - Michele Mishto
- Institut für Biochemie, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health, Berlin, Germany.,Centre for Inflammation Biology and Cancer Immunology (CIBCI), Peter Gorer Department of Immunobiology, King's College London, London, United Kingdom
| | - Alice J A M Sijts
- Faculty of Veterinary Medicine, Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
60
|
von Brzezinski L, Säring P, Landgraf P, Cammann C, Seifert U, Dieterich DC. Low Neurotoxicity of ONX-0914 Supports the Idea of Specific Immunoproteasome Inhibition as a Side-Effect-Limiting, Therapeutic Strategy. Eur J Microbiol Immunol (Bp) 2017; 7:234-245. [PMID: 29034113 PMCID: PMC5632751 DOI: 10.1556/1886.2017.00025] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 09/05/2017] [Indexed: 01/01/2023] Open
Abstract
Application of the proteasome inhibitor Bortezomib for the treatment of haematopoietic malignancies such as multiple myeloma significantly improves the average overall survival of patients. However, one of the most severe side effects is the development of peripheral neuropathies caused by neurotoxic effects of Bortezomib limiting its therapeutic efficacy. With ONX-0914 a specific inhibitor of the β5i (LMP7)-immunosubunit containing proteasomes was developed that targets exclusively the proteasome subtypes mainly expressed in immune cells including B lymphocytes as the origin of multiple myeloma. Furthermore, immunosubunitspecific inhibitors have been shown to be promising tools for the therapy of autoimmune disorders. In the presented study, we analysed the concentration-dependent impact of both inhibitors on primary neurons regarding survival rate, morphological changes, and overall viability. Our results clearly demonstrate that ONX-0914, compared to Bortezomib, is less neurotoxic suggesting its potential as a putative antineoplastic drug and as a candidate for the treatment of autoimmune disorders affecting the peripheral and/or central nervous system.
Collapse
Affiliation(s)
- Laura von Brzezinski
- Neural Plasticity and Communication, Institute for Pharmacology and Toxicology, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Paula Säring
- Neural Plasticity and Communication, Institute for Pharmacology and Toxicology, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Peter Landgraf
- Neural Plasticity and Communication, Institute for Pharmacology and Toxicology, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Clemens Cammann
- Friedrich Loeffler Institute for Medical Microbiology, Greifswald, Germany
| | - Ulrike Seifert
- Friedrich Loeffler Institute for Medical Microbiology, Greifswald, Germany
| | - Daniela C Dieterich
- Neural Plasticity and Communication, Institute for Pharmacology and Toxicology, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany.,Center for Behavioral Brain Sciences, Magdeburg, Germany
| |
Collapse
|
61
|
Abelin JG, Keskin DB, Sarkizova S, Hartigan CR, Zhang W, Sidney J, Stevens J, Lane W, Zhang GL, Eisenhaure TM, Clauser KR, Hacohen N, Rooney MS, Carr SA, Wu CJ. Mass Spectrometry Profiling of HLA-Associated Peptidomes in Mono-allelic Cells Enables More Accurate Epitope Prediction. Immunity 2017; 46:315-326. [PMID: 28228285 DOI: 10.1016/j.immuni.2017.02.007] [Citation(s) in RCA: 451] [Impact Index Per Article: 56.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 11/29/2016] [Accepted: 12/29/2016] [Indexed: 12/11/2022]
Abstract
Identification of human leukocyte antigen (HLA)-bound peptides by liquid chromatography-tandem mass spectrometry (LC-MS/MS) is poised to provide a deep understanding of rules underlying antigen presentation. However, a key obstacle is the ambiguity that arises from the co-expression of multiple HLA alleles. Here, we have implemented a scalable mono-allelic strategy for profiling the HLA peptidome. By using cell lines expressing a single HLA allele, optimizing immunopurifications, and developing an application-specific spectral search algorithm, we identified thousands of peptides bound to 16 different HLA class I alleles. These data enabled the discovery of subdominant binding motifs and an integrative analysis quantifying the contribution of factors critical to epitope presentation, such as protein cleavage and gene expression. We trained neural-network prediction algorithms with our large dataset (>24,000 peptides) and outperformed algorithms trained on datasets of peptides with measured affinities. We thus demonstrate a strategy for systematically learning the rules of endogenous antigen presentation.
Collapse
Affiliation(s)
| | - Derin B Keskin
- Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA; Department of Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA; Harvard Medical School, Boston, MA, 02115, USA; Department of Computer Science, Metropolitan College, Boston University, Boston, MA, 02215, USA
| | - Siranush Sarkizova
- Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Biomedical Informatics, Harvard Medical School, Boston, MA, 02142, USA
| | | | - Wandi Zhang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - John Sidney
- La Jolla Institute for Allergy and Immunology, 92037, La Jolla, CA
| | - Jonathan Stevens
- Tissue Typing Laboratory, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - William Lane
- Tissue Typing Laboratory, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Guang Lan Zhang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA; Harvard Medical School, Boston, MA, 02115, USA; Department of Computer Science, Metropolitan College, Boston University, Boston, MA, 02215, USA
| | | | | | - Nir Hacohen
- Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA; Center for Cancer Immunology, Massachusetts General Hospital, Boston, MA, 02114, USA.
| | - Michael S Rooney
- Broad Institute of MIT and Harvard, Cambridge, MA, USA; Harvard/MIT Division of Health Sciences and Technology, Cambridge, Massachusetts, 02139 USA; Neon Therapeutics, Cambridge, MA, 02139, USA.
| | - Steven A Carr
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| | - Catherine J Wu
- Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA; Department of Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA; Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
62
|
Post-Translational Peptide Splicing and T Cell Responses. Trends Immunol 2017; 38:904-915. [PMID: 28830734 DOI: 10.1016/j.it.2017.07.011] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 07/10/2017] [Accepted: 07/26/2017] [Indexed: 12/21/2022]
Abstract
CD8+ T cell specificity depends on the recognition of MHC class I-epitope complexes at the cell surface. These epitopes are mainly produced via degradation of proteins by the proteasome, generating fragments of the original sequence. However, it is now clear that proteasomes can produce a significant portion of epitopes by reshuffling the antigen sequence, thus expanding the potential antigenic repertoire. MHC class I-restricted spliced epitopes have been described in tumors and infections, suggesting an unpredicted relevance of these peculiar peptides. We review current knowledge about proteasome-catalyzed peptide splicing (PCPS), the emerging rules governing this process, and the potential implications for our understanding and therapeutic use of CD8+ T cells, as well as mechanisms generating other non-canonical antigenic epitopes targeted by the T cell response.
Collapse
|
63
|
Chang AY, Dao T, Gejman RS, Jarvis CA, Scott A, Dubrovsky L, Mathias MD, Korontsvit T, Zakhaleva V, Curcio M, Hendrickson RC, Liu C, Scheinberg DA. A therapeutic T cell receptor mimic antibody targets tumor-associated PRAME peptide/HLA-I antigens. J Clin Invest 2017; 127:2705-2718. [PMID: 28628042 PMCID: PMC5490756 DOI: 10.1172/jci92335] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 04/27/2017] [Indexed: 12/18/2022] Open
Abstract
Preferentially expressed antigen in melanoma (PRAME) is a cancer-testis antigen that is expressed in many cancers and leukemias. In healthy tissue, PRAME expression is limited to the testes and ovaries, making it a highly attractive cancer target. PRAME is an intracellular protein that cannot currently be drugged. After proteasomal processing, the PRAME300-309 peptide ALYVDSLFFL (ALY) is presented in the context of human leukocyte antigen HLA-A*02:01 molecules for recognition by the T cell receptor (TCR) of cytotoxic T cells. Here, we have described Pr20, a TCR mimic (TCRm) human IgG1 antibody that recognizes the cell-surface ALY peptide/HLA-A2 complex. Pr20 is an immunological tool and potential therapeutic agent. Pr20 bound to PRAME+HLA-A2+ cancers. An afucosylated Fc form (Pr20M) directed antibody-dependent cellular cytotoxicity against PRAME+HLA-A2+ leukemia cells and was therapeutically effective against mouse xenograft models of human leukemia. In some tumors, Pr20 binding markedly increased upon IFN-γ treatment, mediated by induction of the immunoproteasome catalytic subunit β5i. The immunoproteasome reduced internal destructive cleavages within the ALY epitope compared with the constitutive proteasome. The data provide rationale for developing TCRm antibodies as therapeutic agents for cancer, offer mechanistic insight on proteasomal regulation of tumor-associated peptide/HLA antigen complexes, and yield possible therapeutic solutions to target antigens with ultra-low surface presentation.
Collapse
Affiliation(s)
- Aaron Y. Chang
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center (MSKCC), New York, New York, USA
- Biochemistry Cell and Molecular Biology Program
| | - Tao Dao
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center (MSKCC), New York, New York, USA
| | - Ron S. Gejman
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center (MSKCC), New York, New York, USA
- Pharmacology Program, and
| | - Casey A. Jarvis
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center (MSKCC), New York, New York, USA
| | - Andrew Scott
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center (MSKCC), New York, New York, USA
- Immunology and Microbial Pathogenesis Program, Weill Cornell Medicine, Cornell University, New York, New York, USA
| | - Leonid Dubrovsky
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center (MSKCC), New York, New York, USA
| | - Melissa D. Mathias
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center (MSKCC), New York, New York, USA
| | - Tatyana Korontsvit
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center (MSKCC), New York, New York, USA
| | - Victoriya Zakhaleva
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center (MSKCC), New York, New York, USA
| | - Michael Curcio
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center (MSKCC), New York, New York, USA
| | - Ronald C. Hendrickson
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center (MSKCC), New York, New York, USA
| | - Cheng Liu
- Eureka Therapeutics, Emeryville, California, USA
| | - David A. Scheinberg
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center (MSKCC), New York, New York, USA
- Pharmacology Program, and
| |
Collapse
|
64
|
Ortega-Atienza S, Krawic C, Watts L, McCarthy C, Luczak MW, Zhitkovich A. 20S immunoproteasomes remove formaldehyde-damaged cytoplasmic proteins suppressing caspase-independent cell death. Sci Rep 2017; 7:654. [PMID: 28381880 PMCID: PMC5429636 DOI: 10.1038/s41598-017-00757-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 03/09/2017] [Indexed: 01/08/2023] Open
Abstract
Immunoproteasomes are known for their involvement in antigen presentation. However, their broad tissue presence and other evidence are indicative of nonimmune functions. We examined a role for immunoproteasomes in cellular responses to the endogenous and environmental carcinogen formaldehyde (FA) that binds to cytosolic and nuclear proteins producing proteotoxic stress and genotoxic DNA-histone crosslinks. We found that immunoproteasomes were important for suppression of a caspase-independent cell death and the long-term survival of FA-treated cells. All major genotoxic responses to FA, including replication inhibition and activation of the transcription factor p53 and the apical ATM and ATR kinases, were unaffected by immunoproteasome inactivity. Immunoproteasome inhibition enhanced activation of the cytosolic protein damage sensor HSF1, elevated levels of K48-polyubiquitinated cytoplasmic proteins and increased depletion of unconjugated ubiquitin. We further found that FA induced the disassembly of 26S immunoproteasomes, but not standard 26S proteasomes, releasing the 20S catalytic immunoproteasome. FA-treated cells also had higher amounts of small activators PA28αβ and PA28γ bound to 20S particles. Our findings highlight the significance of nonnuclear damage in FA injury and reveal a major role for immunoproteasomes in elimination of FA-damaged cytoplasmic proteins through ubiquitin-independent proteolysis.
Collapse
Affiliation(s)
- Sara Ortega-Atienza
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, 02912, USA
| | - Casey Krawic
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, 02912, USA
| | - Lauren Watts
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, 02912, USA
| | - Caitlin McCarthy
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, 02912, USA
| | - Michal W Luczak
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, 02912, USA
| | - Anatoly Zhitkovich
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, 02912, USA.
| |
Collapse
|
65
|
Immunoproteasome subunit deficiency has no influence on the canonical pathway of NF-κB activation. Mol Immunol 2017; 83:147-153. [PMID: 28157553 DOI: 10.1016/j.molimm.2017.01.019] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Revised: 01/13/2017] [Accepted: 01/20/2017] [Indexed: 01/02/2023]
Abstract
Activation of the pro-inflammatory transcription factor NF-κB requires signal-induced proteasomal degradation of the inhibitor of NF-κB (IκB) in order to allow nuclear translocation. Most cell types are capable of expressing two types of 20S proteasome core particles, the constitutive proteasome and immunoproteasome. Inducible under inflammatory conditions, the immunoproteasome is mainly characterized through an altered cleavage specificity compared to the constitutive proteasome. However, the question whether immunoproteasome subunits affect NF-κB signal transduction differently from constitutive subunits is still up for debate. To study the effect of immunoproteasomes on LPS- or TNF-α-induced NF-κB activation, we used IFN-γ stimulated peritoneal macrophages and mouse embryonic fibroblasts derived from mice deficient for the immunoproteasome subunits low molecular mass polypeptide (LMP) 2, or LMP7 and multicatalytic endopeptidase complex-like 1 (MECL-1). Along the canonical signaling pathway of NF-κB activation no differences in the extent and kinetic of IκB degradation were observed. Neither the nuclear translocation and DNA binding of NF-κB nor the production of the NF-κB dependent cytokines TNF-α, IL-6, and IL-10 differed between immunoproteasome deficient and proficient cells. Hence, we conclude that immunoproteasome subunits have no specialized function for canonical NF-κB activation.
Collapse
|
66
|
Expression of immunoproteasome genes is regulated by cell-intrinsic and -extrinsic factors in human cancers. Sci Rep 2016; 6:34019. [PMID: 27659694 PMCID: PMC5034284 DOI: 10.1038/srep34019] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 09/06/2016] [Indexed: 01/07/2023] Open
Abstract
Based on transcriptomic analyses of thousands of samples from The Cancer Genome Atlas, we report that expression of constitutive proteasome (CP) genes (PSMB5, PSMB6, PSMB7) and immunoproteasome (IP) genes (PSMB8, PSMB9, PSMB10) is increased in most cancer types. In breast cancer, expression of IP genes was determined by the abundance of tumor infiltrating lymphocytes and high expression of IP genes was associated with longer survival. In contrast, IP upregulation in acute myeloid leukemia (AML) was a cell-intrinsic feature that was not associated with longer survival. Expression of IP genes in AML was IFN-independent, correlated with the methylation status of IP genes, and was particularly high in AML with an M5 phenotype and/or MLL rearrangement. Notably, PSMB8 inhibition led to accumulation of polyubiquitinated proteins and cell death in IPhigh but not IPlow AML cells. Co-clustering analysis revealed that genes correlated with IP subunits in non-M5 AMLs were primarily implicated in immune processes. However, in M5 AML, IP genes were primarily co-regulated with genes involved in cell metabolism and proliferation, mitochondrial activity and stress responses. We conclude that M5 AML cells can upregulate IP genes in a cell-intrinsic manner in order to resist cell stress.
Collapse
|
67
|
Rock KL, Reits E, Neefjes J. Present Yourself! By MHC Class I and MHC Class II Molecules. Trends Immunol 2016; 37:724-737. [PMID: 27614798 DOI: 10.1016/j.it.2016.08.010] [Citation(s) in RCA: 527] [Impact Index Per Article: 58.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2016] [Revised: 08/09/2016] [Accepted: 08/15/2016] [Indexed: 12/01/2022]
Abstract
Since the discovery of MHC molecules, it has taken 40 years to arrive at a coherent picture of how MHC class I and MHC class II molecules really work. This is a story of the proteases and MHC-like chaperones that support the MHC class I and II molecules in presenting peptides to the immune system. We now understand that the MHC system shapes both the repertoire of presented peptides and the subsequent T cell response, with important implications ranging from transplant rejection to tumor immunotherapies. Here we present an illustrated review of the ins and outs of MHC class I and MHC class II antigen presentation.
Collapse
Affiliation(s)
- Kenneth L Rock
- Department of Pathology, UMass Medical School, Worcester, MA, USA
| | - Eric Reits
- Department of Cell Biology and Histology, Amsterdam Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Jacques Neefjes
- Department of Cell Biology, The Netherlands Cancer Institute, Amsterdam, The Netherlands; Department of Chemical Immunology, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
68
|
Koizumi S, Irie T, Hirayama S, Sakurai Y, Yashiroda H, Naguro I, Ichijo H, Hamazaki J, Murata S. The aspartyl protease DDI2 activates Nrf1 to compensate for proteasome dysfunction. eLife 2016; 5. [PMID: 27528193 PMCID: PMC5001836 DOI: 10.7554/elife.18357] [Citation(s) in RCA: 134] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 08/12/2016] [Indexed: 01/21/2023] Open
Abstract
In response to proteasome dysfunction, mammalian cells upregulate proteasome gene expression by activating Nrf1. Nrf1 is an endoplasmic reticulum-resident transcription factor that is continually retrotranslocated and degraded by the proteasome. Upon proteasome inhibition, Nrf1 escapes degradation and is cleaved to become active. However, the processing enzyme for Nrf1 remains obscure. Here we show that the aspartyl protease DNA-damage inducible 1 homolog 2 (DDI2) is required to cleave and activate Nrf1. Deletion of DDI2 reduced the cleaved form of Nrf1 and increased the full-length cytosolic form of Nrf1, resulting in poor upregulation of proteasomes in response to proteasome inhibition. These defects were restored by adding back wild-type DDI2 but not protease-defective DDI2. Our results provide a clue for blocking compensatory proteasome synthesis to improve cancer therapies targeting proteasomes. DOI:http://dx.doi.org/10.7554/eLife.18357.001 The proteasome is a machine that destroys unnecessary or damaged proteins inside cells. This role of the proteasome is essential for cell survival, and so when the proteasome is inhibited, cells produce new proteasomes to compensate. Upon proteasome inhibition, a protein called Nrf1 is activated and executes this “bounce-back” response. Some cancer treatments aim to kill cancer cells by inhibiting proteasomes, but these treatments may be unsuccessful if the bounce-back response is not also prevented. Therefore, understanding how Nrf1 is activated is an important issue. Nrf1 is produced at a structure called the endoplasmic reticulum in cells and is continually destroyed by the proteasome. On the other hand, when proteasomes are inhibited, Nrf1 accumulates and is cleaved into an active form, which moves to the cell nucleus to start producing proteasomes. However, it was not known which molecule cleaves Nrf1. Koizumi et al. set out to discover this molecule by screening the genetic material of human cells, and identified a gene that encodes a protease (an enzyme that cleaves other proteins) called DDI2. The loss of DDI2 from cells prevented Nrf1 from being cleaved and entering the nucleus, resulting in low levels of proteasome production. Further experiments showed that a mutant form of DDI2 that lacked protease activity was unable to cleave Nrf1, confirming DDI2’s role in activating Nrf1. Deleting DDI2 from cells does not completely prevent the cleavage of Nrf1, and so some other cleaving enzyme might exist; the identity of this enzyme remains to be discovered. Future work is also needed to establish exactly how DDI2 cleaves Nrf1. This could help to develop a DDI2 inhibitor for cancer treatment that could be used in combination with existing proteasome inhibitors. DOI:http://dx.doi.org/10.7554/eLife.18357.002
Collapse
Affiliation(s)
- Shun Koizumi
- Laboratory of Protein Metabolism, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Taro Irie
- Laboratory of Protein Metabolism, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Shoshiro Hirayama
- Laboratory of Protein Metabolism, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Yasuyuki Sakurai
- Laboratory of Protein Metabolism, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Hideki Yashiroda
- Laboratory of Protein Metabolism, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Isao Naguro
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Hidenori Ichijo
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Jun Hamazaki
- Laboratory of Protein Metabolism, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Shigeo Murata
- Laboratory of Protein Metabolism, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
69
|
Kammerl IE, Meiners S. Proteasome function shapes innate and adaptive immune responses. Am J Physiol Lung Cell Mol Physiol 2016; 311:L328-36. [PMID: 27343191 DOI: 10.1152/ajplung.00156.2016] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 06/17/2016] [Indexed: 11/22/2022] Open
Abstract
The proteasome system degrades more than 80% of intracellular proteins into small peptides. Accordingly, the proteasome is involved in many essential cellular functions, such as protein quality control, transcription, immune responses, cell signaling, and apoptosis. Moreover, degradation products are loaded onto major histocompatibility class I molecules to communicate the intracellular protein composition to the immune system. The standard 20S proteasome core complex contains three distinct catalytic active sites that are exchanged upon stimulation with inflammatory cytokines to form the so-called immunoproteasome. Immunoproteasomes are constitutively expressed in immune cells and have different proteolytic activities compared with standard proteasomes. They are rapidly induced in parenchymal cells upon intracellular pathogen infection and are crucial for priming effective CD8(+) T-cell-mediated immune responses against infected cells. Beyond shaping these adaptive immune reactions, immunoproteasomes also regulate the function of immune cells by degradation of inflammatory and immune mediators. Accordingly, they emerge as novel regulators of innate immune responses. The recently unraveled impairment of immunoproteasome function by environmental challenges and by genetic variations of immunoproteasome genes might represent a currently underestimated risk factor for the development and progression of lung diseases. In particular, immunoproteasome dysfunction will dampen resolution of infections, thereby promoting exacerbations, may foster autoimmunity in chronic lung diseases, and possibly contributes to immune evasion of tumor cells. Novel pharmacological tools, such as site-specific inhibitors of the immunoproteasome, as well as activity-based probes, however, hold promises as innovative therapeutic drugs for respiratory diseases and biomarker profiling, respectively.
Collapse
Affiliation(s)
- Ilona E Kammerl
- Comprehensive Pneumology Center, University Hospital, Ludwig-Maximilians University and Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Silke Meiners
- Comprehensive Pneumology Center, University Hospital, Ludwig-Maximilians University and Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| |
Collapse
|
70
|
Concha-Benavente F, Srivastava R, Ferrone S, Ferris RL. Immunological and clinical significance of HLA class I antigen processing machinery component defects in malignant cells. Oral Oncol 2016; 58:52-8. [PMID: 27264839 DOI: 10.1016/j.oraloncology.2016.05.008] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 05/11/2016] [Indexed: 12/31/2022]
Abstract
Experimental as well as clinical studies demonstrate that the immune system plays a major role in controlling generation and progression of tumors. The cancer immunoediting theory supports the notion that tumor cell immunogenicity is dynamically shaped by the immune system, as it eliminates immunogenic tumor cells in the early stage of the disease and then edits their antigenicity. The end result is the generation of a tumor cell population able to escape from immune recognition and elimination by tumor infiltrating lymphocytes. Two major mechanisms, which affect the target cells and the effector phase of the immune response, play a crucial role in the editing process. One is represented by the downregulation of tumor antigen (TA) processing and presentation because of abnormalities in the HLA class I antigen processing machinery (APM). The other one is represented by the anergy of effector immune infiltrates in the tumor microenvironment caused by aberrant inhibitory signals triggered by immune checkpoint receptor (ICR) ligands, such as programmed death ligand-1 (PD-L1). In this review, we will focus on tumor immune escape mechanisms caused by defects in HLA class I APM component expression and/or function in different types of cancer, with emphasis on head and neck cancer (HNC). We will also discuss the immunological implications and clinical relevance of these HLA class I APM abnormalities. Finally, we will describe strategies to counteract defective TA presentation with the expectation that they will enhance tumor recognition and elimination by tumor infiltrating effector T cells.
Collapse
Affiliation(s)
| | | | - Soldano Ferrone
- Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Robert L Ferris
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA; Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA, USA; Cancer Immunology Program, University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA.
| |
Collapse
|
71
|
Kowalewski DJ, Walz S, Backert L, Schuster H, Kohlbacher O, Weisel K, Rittig SM, Kanz L, Salih HR, Rammensee HG, Stevanović S, Stickel JS. Carfilzomib alters the HLA-presented peptidome of myeloma cells and impairs presentation of peptides with aromatic C-termini. Blood Cancer J 2016; 6:e411. [PMID: 27058226 PMCID: PMC4855252 DOI: 10.1038/bcj.2016.14] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 02/02/2016] [Indexed: 02/03/2023] Open
Abstract
Recent studies suggest that multiple myeloma is an immunogenic disease, which might be effectively targeted by antigen-specific T-cell immunotherapy. As standard of care in myeloma includes proteasome inhibitor therapy, it is of great importance to characterize the effects of this treatment on HLA-restricted antigen presentation and implement only robustly presented targets for immunotherapeutic intervention. Here, we present a study that longitudinally and semi-quantitatively maps the effects of the proteasome inhibitor carfilzomib on HLA-restricted antigen presentation. The relative presentation levels of 4780 different HLA ligands were quantified in an in vitro model employing carfilzomib treatment of MM.1S and U266 myeloma cells, which revealed significant modulation of a substantial fraction of the HLA-presented peptidome. Strikingly, we detected selective down-modulation of HLA ligands with aromatic C-terminal anchor amino acids. This particularly manifested as a marked reduction in the presentation of HLA ligands through the HLA allotypes A*23:01 and A*24:02 on MM.1S cells. These findings implicate that carfilzomib mediates a direct, peptide motif-specific inhibitory effect on HLA ligand processing and presentation. As a substantial proportion of HLA allotypes present peptides with aromatic C-termini, our results may have broad implications for the implementation of antigen-specific treatment approaches in patients undergoing carfilzomib treatment.
Collapse
Affiliation(s)
- D J Kowalewski
- Department of Immunology, Institute for Cell Biology, University of Tübingen, Tübingen, Germany
| | - S Walz
- Department of Immunology, Institute for Cell Biology, University of Tübingen, Tübingen, Germany.,Department of Hematology and Oncology, University of Tübingen, Tübingen, Germany
| | - L Backert
- Department of Immunology, Institute for Cell Biology, University of Tübingen, Tübingen, Germany.,Applied Bioinformatics, Department of Computer Science, Center for Bioinformatics, University of Tübingen, Tübingen, Germany
| | - H Schuster
- Department of Immunology, Institute for Cell Biology, University of Tübingen, Tübingen, Germany
| | - O Kohlbacher
- Applied Bioinformatics, Department of Computer Science, Center for Bioinformatics, University of Tübingen, Tübingen, Germany.,Quantitative Biology Center, University of Tübingen, Tübingen, Germany.,Max Planck Institute for Developmental Biology, Tübingen, Germany
| | - K Weisel
- Department of Hematology and Oncology, University of Tübingen, Tübingen, Germany
| | - S M Rittig
- Department of Hematology and Oncology, University of Tübingen, Tübingen, Germany
| | - L Kanz
- Department of Hematology and Oncology, University of Tübingen, Tübingen, Germany
| | - H R Salih
- Department of Hematology and Oncology, University of Tübingen, Tübingen, Germany.,Clinical Cooperation Unit Translational Immunology, German Cancer Consortium (DKTK), DKFZ Partner Site, Tübingen, Germany
| | - H-G Rammensee
- Department of Immunology, Institute for Cell Biology, University of Tübingen, Tübingen, Germany.,German Cancer Consortium (DKTK), DKFZ Partner Site, Tübingen, Germany
| | - S Stevanović
- Department of Immunology, Institute for Cell Biology, University of Tübingen, Tübingen, Germany.,German Cancer Consortium (DKTK), DKFZ Partner Site, Tübingen, Germany
| | - J S Stickel
- Department of Hematology and Oncology, University of Tübingen, Tübingen, Germany
| |
Collapse
|
72
|
Campana S, De Pasquale C, Carrega P, Ferlazzo G, Bonaccorsi I. Cross-dressing: an alternative mechanism for antigen presentation. Immunol Lett 2015; 168:349-54. [DOI: 10.1016/j.imlet.2015.11.002] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Revised: 11/02/2015] [Accepted: 11/03/2015] [Indexed: 12/16/2022]
|
73
|
Sennikov SV, Shevchenko JA, Kurilin VV, Khantakova JN, Lopatnikova JA, Gavrilova EV, Maksyutov RA, Bakulina AY, Sidorov SV, Khristin AA, Maksyutov AZ. Induction of an antitumor response using dendritic cells transfected with DNA constructs encoding the HLA-A*02:01-restricted epitopes of tumor-associated antigens in culture of mononuclear cells of breast cancer patients. Immunol Res 2015; 64:171-80. [DOI: 10.1007/s12026-015-8735-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
74
|
Mundt S, Engelhardt B, Kirk CJ, Groettrup M, Basler M. Inhibition and deficiency of the immunoproteasome subunit LMP7 attenuates LCMV-induced meningitis. Eur J Immunol 2015; 46:104-13. [PMID: 26464284 DOI: 10.1002/eji.201545578] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Revised: 09/07/2015] [Accepted: 10/08/2015] [Indexed: 11/05/2022]
Abstract
In addition to antigen processing, immunoproteasomes were recently shown to exert functions influencing cytokine production by monocytes and T cells, T-helper cell differentiation, and T-cell survival. Moreover, selective inhibition of the immunoproteasome subunit LMP7 ameliorated symptoms of autoimmune diseases including CD4(+) T-cell mediated EAE. In this study, we show that LMP7 also plays a crucial role in the pathogenesis of lymphocytic choriomeningitis virus (LCMV)-induced meningitis mediated by CTLs. Mice lacking functional LMP7 display delayed and reduced clinical signs of disease accompanied by a strongly decreased inflammatory infiltration into the brain. Interestingly, we found that selective inhibition and genetic deficiency of LMP7 affect the pathogenesis of LCMV-induced meningitis in a distinct manner. Our findings support the important role of LMP7 in inflammatory disorders and suggest immunoproteasome inhibition as a novel strategy against inflammation-induced neuropathology in the CNS.
Collapse
Affiliation(s)
- Sarah Mundt
- Division of Immunology, Department of Biology, University of Konstanz, Konstanz, Germany.,Konstanz Research School Chemical Biology (KoRS-CB), University of Konstanz, Konstanz, Germany
| | | | | | - Marcus Groettrup
- Division of Immunology, Department of Biology, University of Konstanz, Konstanz, Germany.,Biotechnology Institute Thurgau at the University of Konstanz (BITg), Kreuzlingen, Switzerland
| | - Michael Basler
- Division of Immunology, Department of Biology, University of Konstanz, Konstanz, Germany.,Biotechnology Institute Thurgau at the University of Konstanz (BITg), Kreuzlingen, Switzerland
| |
Collapse
|
75
|
In-Depth, Label-Free Analysis of the Erythrocyte Cytoplasmic Proteome in Diamond Blackfan Anemia Identifies a Unique Inflammatory Signature. PLoS One 2015; 10:e0140036. [PMID: 26474164 PMCID: PMC4608755 DOI: 10.1371/journal.pone.0140036] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 09/21/2015] [Indexed: 11/19/2022] Open
Abstract
Diamond Blackfan Anemia (DBA) is a rare, congenital erythrocyte aplasia that is usually caused by haploinsufficiency of ribosomal proteins due to diverse mutations in one of several ribosomal genes. A striking feature of this disease is that a range of different mutations in ribosomal proteins results in similar disease phenotypes primarily characterized by erythrocyte abnormalities and macrocytic anemia, while most other cell types in the body are minimally affected. Previously, we analyzed the erythrocyte membrane proteomes of several DBA patients and identified several proteins that are not typically associated with this cell type and that suggested inflammatory mechanisms contribute to the pathogenesis of DBA. In this study, we evaluated the erythrocyte cytosolic proteome of DBA patients through in-depth analysis of hemoglobin-depleted erythrocyte cytosols. Simple, reproducible, hemoglobin depletion using nickel columns enabled in-depth analysis of over 1000 cytosolic erythrocyte proteins with only moderate total analysis time per proteome. Label-free quantitation and statistical analysis identified 29 proteins with significantly altered abundance levels in DBA patients compared to matched healthy control donors. Proteins that were significantly increased in DBA erythrocyte cytoplasms included three proteasome subunit beta proteins that make up the immunoproteasome and proteins induced by interferon-γ such as n-myc interactor and interferon-induced 35 kDa protein [NMI and IFI35 respectively]. Pathway analysis confirmed the presence of an inflammatory signature in erythrocytes of DBA patients and predicted key upstream regulators including mitogen activated kinase 1, interferon-γ, tumor suppressor p53, and tumor necrosis factor. These results show that erythrocytes in DBA patients are intrinsically different from those in healthy controls which may be due to an inflammatory response resulting from the inherent molecular defect of ribosomal protein haploinsufficiency or changes in the bone marrow microenvironment that leads to red cell aplasia in DBA patients.
Collapse
|
76
|
Lei J, Osen W, Gardyan A, Hotz-Wagenblatt A, Wei G, Gissmann L, Eichmüller S, Löchelt M. Replication-Competent Foamy Virus Vaccine Vectors as Novel Epitope Scaffolds for Immunotherapy. PLoS One 2015; 10:e0138458. [PMID: 26397953 PMCID: PMC4580568 DOI: 10.1371/journal.pone.0138458] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 08/31/2015] [Indexed: 12/19/2022] Open
Abstract
The use of whole viruses as antigen scaffolds is a recent development in vaccination that improves immunogenicity without the need for additional adjuvants. Previous studies highlighted the potential of foamy viruses (FVs) in prophylactic vaccination and gene therapy. Replication-competent FVs can trigger immune signaling and integrate into the host genome, resulting in persistent antigen expression and a robust immune response. Here, we explored feline foamy virus (FFV) proteins as scaffolds for therapeutic B and T cell epitope delivery in vitro. Infection- and cancer-related B and T cell epitopes were grafted into FFV Gag, Env, or Bet by residue replacement, either at sites of high local sequence homology between the epitope and the host protein or in regions known to tolerate sequence alterations. Modified proviruses were evaluated in vitro for protein steady state levels, particle release, and virus titer in permissive cells. Modification of Gag and Env was mostly detrimental to their function. As anticipated, modification of Bet had no impact on virion release and affected virus titers of only some recombinants. Further evaluation of Bet as an epitope carrier was performed using T cell epitopes from the model antigen chicken ovalbumin (OVA), human tyrosinase-related protein 2 (TRP-2), and oncoprotein E7 of human papillomavirus type 16 (HPV16E7). Transfection of murine cells with constructs encoding Bet-epitope chimeric proteins led to efficient MHC-I-restricted epitope presentation as confirmed by interferon-gamma enzyme-linked immunospot assays using epitope-specific cytotoxic T lymphocyte (CTL) lines. FFV infection-mediated transduction of cells with epitope-carrying Bet also induced T-cell responses, albeit with reduced efficacy, in a process independent from the presence of free peptides. We show that primate FV Bet is also a promising T cell epitope carrier for clinical translation. The data demonstrate the utility of replication-competent and -attenuated FVs as antigen carriers in immunotherapy.
Collapse
Affiliation(s)
- Janet Lei
- Division of Molecular Diagnostics of Oncogenic Infections, Research Program Infection and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Wolfram Osen
- Division of Translational Immunology, Research Program Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Adriane Gardyan
- Division of Translational Immunology, Research Program Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Agnes Hotz-Wagenblatt
- Bioinformatics Core Facility, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Guochao Wei
- Division of Molecular Diagnostics of Oncogenic Infections, Research Program Infection and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Lutz Gissmann
- Division of Molecular Diagnostics of Oncogenic Infections, Research Program Infection and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stefan Eichmüller
- Division of Translational Immunology, Research Program Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Martin Löchelt
- Division of Molecular Diagnostics of Oncogenic Infections, Research Program Infection and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
77
|
Oyarzun P, Kobe B. Computer-aided design of T-cell epitope-based vaccines: addressing population coverage. Int J Immunogenet 2015. [PMID: 26211755 DOI: 10.1111/iji.12214] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Epitope-based vaccines (EVs) make use of short antigen-derived peptides corresponding to immune epitopes, which are administered to trigger a protective humoral and/or cellular immune response. EVs potentially allow for precise control over the immune response activation by focusing on the most relevant - immunogenic and conserved - antigen regions. Experimental screening of large sets of peptides is time-consuming and costly; therefore, in silico methods that facilitate T-cell epitope mapping of protein antigens are paramount for EV development. The prediction of T-cell epitopes focuses on the peptide presentation process by proteins encoded by the major histocompatibility complex (MHC). Because different MHCs have different specificities and T-cell epitope repertoires, individuals are likely to respond to a different set of peptides from a given pathogen in genetically heterogeneous human populations. In addition, protective immune responses are only expected if T-cell epitopes are restricted by MHC proteins expressed at high frequencies in the target population. Therefore, without careful consideration of the specificity and prevalence of the MHC proteins, EVs could fail to adequately cover the target population. This article reviews state-of-the-art algorithms and computational tools to guide EV design through all the stages of the process: epitope prediction, epitope selection and vaccine assembly, while optimizing vaccine immunogenicity and coping with genetic variation in humans and pathogens.
Collapse
Affiliation(s)
- P Oyarzun
- Biotechnology Centre, Facultad de Ingeniería y Tecnología, Universidad San Sebastián, Concepción, Chile
| | - B Kobe
- School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
78
|
Sasaki K, Takada K, Ohte Y, Kondo H, Sorimachi H, Tanaka K, Takahama Y, Murata S. Thymoproteasomes produce unique peptide motifs for positive selection of CD8(+) T cells. Nat Commun 2015; 6:7484. [PMID: 26099460 PMCID: PMC4557289 DOI: 10.1038/ncomms8484] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 05/13/2015] [Indexed: 11/09/2022] Open
Abstract
Positive selection in the thymus provides low-affinity T-cell receptor (TCR) engagement to support the development of potentially useful self-major histocompatibility complex class I (MHC-I)-restricted T cells. Optimal positive selection of CD8(+) T cells requires cortical thymic epithelial cells that express β5t-containing thymoproteasomes (tCPs). However, how tCPs govern positive selection is unclear. Here we show that the tCPs produce unique cleavage motifs in digested peptides and in MHC-I-associated peptides. Interestingly, MHC-I-associated peptides carrying these tCP-dependent motifs are enriched with low-affinity TCR ligands that efficiently induce the positive selection of functionally competent CD8(+) T cells in antigen-specific TCR-transgenic models. These results suggest that tCPs contribute to the positive selection of CD8(+) T cells by preferentially producing low-affinity TCR ligand peptides.
Collapse
Affiliation(s)
- Katsuhiro Sasaki
- Laboratory of Protein Metabolism, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan
| | - Kensuke Takada
- Division of Experimental Immunology, Institute for Genome Research, University of Tokushima, Tokushima 770-8503, Japan
| | - Yuki Ohte
- Laboratory of Protein Metabolism, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan
| | - Hiroyuki Kondo
- Division of Experimental Immunology, Institute for Genome Research, University of Tokushima, Tokushima 770-8503, Japan
| | - Hiroyuki Sorimachi
- Calpain Project, Department of Advanced Science for Biomolecules, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| | - Keiji Tanaka
- Laboratory of Protein Metabolism, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| | - Yousuke Takahama
- Division of Experimental Immunology, Institute for Genome Research, University of Tokushima, Tokushima 770-8503, Japan
| | - Shigeo Murata
- Laboratory of Protein Metabolism, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan
| |
Collapse
|
79
|
Human Tumor Antigens and Cancer Immunotherapy. BIOMED RESEARCH INTERNATIONAL 2015; 2015:948501. [PMID: 26161423 PMCID: PMC4487697 DOI: 10.1155/2015/948501] [Citation(s) in RCA: 146] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 03/03/2015] [Indexed: 01/21/2023]
Abstract
With the recent developments of adoptive T cell therapies and the use of new monoclonal antibodies against the immune checkpoints, immunotherapy is at a turning point. Key players for the success of these therapies are the cytolytic T lymphocytes, which are a subset of T cells able to recognize and kill tumor cells. Here, I review the nature of the antigenic peptides recognized by these T cells and the processes involved in their presentation. I discuss the importance of understanding how each antigenic peptide is processed in the context of immunotherapy and vaccine delivery.
Collapse
|
80
|
Huber EM, de Bruin G, Heinemeyer W, Paniagua Soriano G, Overkleeft HS, Groll M. Systematic Analyses of Substrate Preferences of 20S Proteasomes Using Peptidic Epoxyketone Inhibitors. J Am Chem Soc 2015; 137:7835-42. [DOI: 10.1021/jacs.5b03688] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Eva M. Huber
- Center
for Integrated Protein Science at the Department Chemie, Lehrstuhl
für Biochemie, Technische Universität München, 85748 Garching, Germany
| | - Gerjan de Bruin
- Gorlaeus
Laboratories, Leiden Institute of Chemistry and Netherlands Proteomics Center, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Wolfgang Heinemeyer
- Center
for Integrated Protein Science at the Department Chemie, Lehrstuhl
für Biochemie, Technische Universität München, 85748 Garching, Germany
| | - Guillem Paniagua Soriano
- Gorlaeus
Laboratories, Leiden Institute of Chemistry and Netherlands Proteomics Center, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Herman S. Overkleeft
- Gorlaeus
Laboratories, Leiden Institute of Chemistry and Netherlands Proteomics Center, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Michael Groll
- Center
for Integrated Protein Science at the Department Chemie, Lehrstuhl
für Biochemie, Technische Universität München, 85748 Garching, Germany
| |
Collapse
|
81
|
Abstract
All living organisms require protein degradation to terminate biological processes and remove damaged proteins. One such machine is the 20S proteasome, a specialized barrel-shaped and compartmentalized multicatalytic protease. The activity of the 20S proteasome generally requires the binding of regulators/proteasome activators (PAs), which control the entrance of substrates. These include the PA700 (19S complex), which assembles with the 20S and forms the 26S proteasome and allows the efficient degradation of proteins usually labeled by ubiquitin tags, PA200 and PA28, which are involved in proteolysis through ubiquitin-independent mechanisms and PI31, which was initially identified as a 20S inhibitor in vitro. Unlike 20S proteasome, shown to be present in all Eukaryotes and Archaea, the evolutionary history of PAs remained fragmentary. Here, we made a comprehensive survey and phylogenetic analyses of the four types of regulators in 17 clades covering most of the eukaryotic supergroups. We found remarkable conservation of each PA700 subunit in all eukaryotes, indicating that the current complex PA700 structure was already set up in the last eukaryotic common ancestor (LECA). Also present in LECA, PA200, PA28, and PI31 showed a more contrasted evolutionary picture, because many lineages have subsequently lost one or two of them. The paramount conservation of PA700 composition in all eukaryotes and the dynamic evolution of PA200, PA28, and PI31 are discussed in the light of current knowledge on their physiological roles.
Collapse
Affiliation(s)
- Philippe Fort
- CNRS, CRBM, UMR5237, Montpellier, France Université de Montpellier, France
| | - Andrey V Kajava
- CNRS, CRBM, UMR5237, Montpellier, France Université de Montpellier, France Institut de Biologie Computationnelle, Montpellier, France
| | - Fredéric Delsuc
- Université de Montpellier, France CNRS, IRD, Institut des Sciences de l'Evolution, UMR 5554, Montpellier, France
| | - Olivier Coux
- CNRS, CRBM, UMR5237, Montpellier, France Université de Montpellier, France
| |
Collapse
|
82
|
Sekhavati MH, Heravi RM, Tahmoorespur M, Yousefi S, Abbassi-Daloii T, Akbari R. Cloning, molecular analysis and epitopics prediction of a new chaperone GroEL Brucella melitensis antigen. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2015; 18:499-505. [PMID: 26124937 PMCID: PMC4475659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 02/04/2015] [Indexed: 11/02/2022]
Abstract
OBJECTIVES Brucellosis is a well-known domestic animal infectious disease, which is caused by Brucella bacterium. GroEL antigen increases Brucella survival and is one of the major antigens that stimulates the immune system. Hence, the objective of the present study was cloning and bioinformatics analysis of GroEL gene. MATERIALS AND METHODS The full-length open reading frame of this gene was amplified by specific primers and cloned into pTZ57R/T vector. Also, the sequence of this gene in the Brucella melitensis strain Rev 1 was submitted to the NCBI gene bank for the first time. Several prediction software applications were also used to predict B and T-cell epitopes, secondary and tertiary structures, antigenicity ability and enzymatic degradation sites. The used software applications validated experimental results. RESULTS The results of phylogenetic analysis showed that the GroEL sequence had near homology with other species instead of other Brucella spp. The bioinformatics tools used in the current study were validated by the results of four different experimental epitope predictions. Bioinformatics analysis identified eight B and seven T-cell epitopes. CONCLUSION According to the antigenic ability and proteasomal cleavage sites, four (150-160, 270-285,351-361 and 385-395) common epitopes were predicted for GroEL gene. Bioinformatics analysis showed that these regions had proper epitope characterization and so may be useful for stimulation of cell-mediated and humoral immunity system.
Collapse
Affiliation(s)
| | - Reza Majidzadeh Heravi
- Faculty of Agriculture, Ferdowsi University of Mashhad, Mashhad, Iran,*Corresponding author: Reza Majidzadeh Heravi. Faculty of Agriculture, Ferdowsi University of Mashhad, Mashhad, Iran. Tel: +98-51-38805747; Fax: +98-51-38796845;
| | | | - Soheil Yousefi
- Faculty of Agriculture, Ferdowsi University of Mashhad, Mashhad, Iran
| | | | - Rahebe Akbari
- Faculty of Agriculture, Ferdowsi University of Mashhad, Mashhad, Iran
| |
Collapse
|
83
|
The capture proteasome assay: A method to measure proteasome activity in vitro. Anal Biochem 2015; 482:7-15. [PMID: 25912419 DOI: 10.1016/j.ab.2015.04.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Revised: 04/04/2015] [Accepted: 04/08/2015] [Indexed: 11/21/2022]
Abstract
Because of its crucial role in various cellular processes, the proteasome is the focus of intensive research for the development of proteasome inhibitors to treat cancer and autoimmune diseases. Here, we describe a new and easy assay to measure the different proteasome activities in vitro (chymotrypsin-like, caspase-like, and trypsin-like) based on proteasome capture on antibody-coated plates, namely the capture proteasome assay (CAPA). Applying the CAPA to lysates from cells expressing standard proteasome, immunoproteasome, or intermediate proteasomes β5i or β1i-β5i, we can monitor the activity of the four proteasome subtypes. The CAPA provided similar results as the standard whole-cell proteasome-Glo assay without the problem of contaminating proteases requiring inhibitors. However, the profile of trypsin-like activity differed between the two assays. This could be partly explained by the presence of MgSO4 in the proteasome-Glo buffer, which inhibits the trypsin-like activity of the proteasome. The CAPA does not need MgSO4 and, therefore, provides a more precise measurement of the trypsin-like activity. The CAPA provides a quick and accurate method to measure proteasome activity in vitro in a very specific manner and should be useful for the development of proteasome inhibitors.
Collapse
|
84
|
Reguzova A, Antonets D, Karpenko L, Ilyichev A, Maksyutov R, Bazhan S. Design and evaluation of optimized artificial HIV-1 poly-T cell-epitope immunogens. PLoS One 2015; 10:e0116412. [PMID: 25786238 PMCID: PMC4364888 DOI: 10.1371/journal.pone.0116412] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2014] [Accepted: 12/09/2014] [Indexed: 11/18/2022] Open
Abstract
A successful HIV vaccine in addition to induction of antibody responses should elicit effective T cell responses. Here we described possible strategies for rational design of T-cell vaccine capable to induce high levels of both CD4+ and CD8+ T- cell responses. We developed artificial HIV-1 polyepitope T-cell immunogens based on the conserved natural CD8+ and CD4+ T cell epitopes from different HIV-1 strains and restricted by the most frequent major human leukocyte antigen (HLA) alleles. Designed immunogens contain optimized core polyepitope sequence and additional "signal" sequences which increase epitope processing and presentation to CD8+ and CD4+ T-lymphocytes: N-terminal ubiquitin, N-terminal signal peptide and C-terminal tyrosine motif of LAMP-1 protein. As a result we engineered three T cell immunogens - TCI-N, TCI-N2, and TCI-N3, with different combinations of signal sequences. All designed immunogens were able to elicit HIV-specific CD4+ and CD8+ T cell responses following immunization. Attachment of either ubiquitin or ER-signal/LAMP-1 sequences increased both CD4+ and CD8+ mediated HIV-specific T cell responses in comparison with polyepitope immunogen without any additional signal sequences. Moreover, TCI-N3 polyepitope immunogen with ubiquitin generated highest magnitude of HIV-specific CD4+ and CD8+ T cell responses in our study. Obtained data suggests that attachment of signal sequences targeting polyepitope immunogens to either MHC class I or MHC class II presentation pathways may improve immunogenicity of T-cell vaccines. These results support the strategy of the rational T cell immunogen design and contribute to the development of effective HIV-1 vaccine.
Collapse
Affiliation(s)
- Alena Reguzova
- State Research Center of Virology and Biotechnology “Vector”, Koltsovo, Novosibirsk region, 630559, Russia
| | - Denis Antonets
- State Research Center of Virology and Biotechnology “Vector”, Koltsovo, Novosibirsk region, 630559, Russia
| | - Larisa Karpenko
- State Research Center of Virology and Biotechnology “Vector”, Koltsovo, Novosibirsk region, 630559, Russia
| | - Alexander Ilyichev
- State Research Center of Virology and Biotechnology “Vector”, Koltsovo, Novosibirsk region, 630559, Russia
| | - Rinat Maksyutov
- State Research Center of Virology and Biotechnology “Vector”, Koltsovo, Novosibirsk region, 630559, Russia
| | - Sergei Bazhan
- State Research Center of Virology and Biotechnology “Vector”, Koltsovo, Novosibirsk region, 630559, Russia
| |
Collapse
|
85
|
McCarthy MK, Weinberg JB. The immunoproteasome and viral infection: a complex regulator of inflammation. Front Microbiol 2015; 6:21. [PMID: 25688236 PMCID: PMC4310299 DOI: 10.3389/fmicb.2015.00021] [Citation(s) in RCA: 135] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 01/08/2015] [Indexed: 11/13/2022] Open
Abstract
During viral infection, proper regulation of immune responses is necessary to ensure successful viral clearance with minimal host tissue damage. Proteasomes play a crucial role in the generation of antigenic peptides for presentation on MHC class I molecules, and thus activation of CD8 T cells, as well as activation of the NF-κB pathway. A specialized type of proteasome called the immunoproteasome is constitutively expressed in hematopoietic cells and induced in non-immune cells during viral infection by interferon signaling. The immunoproteasome regulates CD8 T cell responses to many viral epitopes during infection. Accumulating evidence suggests that the immunoproteasome may also contribute to regulation of proinflammatory cytokine production, activation of the NF-κB pathway, and management of oxidative stress. Many viruses have mechanisms of interfering with immunoproteasome function, including prevention of transcriptional upregulation of immunoproteasome components as well as direct interaction of viral proteins with immunoproteasome subunits. A better understanding of the role of the immunoproteasome in different cell types, tissues, and hosts has the potential to improve vaccine design and facilitate the development of effective treatment strategies for viral infections.
Collapse
Affiliation(s)
- Mary K McCarthy
- Department of Microbiology and Immunology, University of Michigan Ann Arbor, MI, USA
| | - Jason B Weinberg
- Department of Microbiology and Immunology, University of Michigan Ann Arbor, MI, USA ; Department of Pediatrics and Communicable Diseases, University of Michigan Ann Arbor, MI, USA
| |
Collapse
|
86
|
Fabre B, Lambour T, Garrigues L, Amalric F, Vigneron N, Menneteau T, Stella A, Monsarrat B, Van den Eynde B, Burlet-Schiltz O, Bousquet-Dubouch MP. Deciphering preferential interactions within supramolecular protein complexes: the proteasome case. Mol Syst Biol 2015; 11:771. [PMID: 25561571 PMCID: PMC4332148 DOI: 10.15252/msb.20145497] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
In eukaryotic cells, intracellular protein breakdown is mainly performed by the ubiquitin-proteasome system. Proteasomes are supramolecular protein complexes formed by the association of multiple sub-complexes and interacting proteins. Therefore, they exhibit a very high heterogeneity whose function is still not well understood. Here, using a newly developed method based on the combination of affinity purification and protein correlation profiling associated with high-resolution mass spectrometry, we comprehensively characterized proteasome heterogeneity and identified previously unknown preferential associations within proteasome sub-complexes. In particular, we showed for the first time that the two main proteasome subtypes, standard proteasome and immunoproteasome, interact with a different subset of important regulators. This trend was observed in very diverse human cell types and was confirmed by changing the relative proportions of both 20S proteasome forms using interferon-γ. The new method developed here constitutes an innovative and powerful strategy that could be broadly applied for unraveling the dynamic and heterogeneous nature of other biologically relevant supramolecular protein complexes.
Collapse
Affiliation(s)
- Bertrand Fabre
- CNRS IPBS (Institut de Pharmacologie et de Biologie Structurale), Toulouse, France Université de Toulouse UPS IPBS, Toulouse, France
| | - Thomas Lambour
- CNRS IPBS (Institut de Pharmacologie et de Biologie Structurale), Toulouse, France Université de Toulouse UPS IPBS, Toulouse, France
| | - Luc Garrigues
- CNRS IPBS (Institut de Pharmacologie et de Biologie Structurale), Toulouse, France Université de Toulouse UPS IPBS, Toulouse, France
| | - François Amalric
- CNRS IPBS (Institut de Pharmacologie et de Biologie Structurale), Toulouse, France Université de Toulouse UPS IPBS, Toulouse, France
| | - Nathalie Vigneron
- Ludwig Institute for Cancer Research, Brussels, Belgium WELBIO (Walloon Excellence in Life Sciences and Biotechnology), Brussels, Belgium de Duve Institute Université catholique de Louvain, Brussels, Belgium
| | - Thomas Menneteau
- CNRS IPBS (Institut de Pharmacologie et de Biologie Structurale), Toulouse, France Université de Toulouse UPS IPBS, Toulouse, France
| | - Alexandre Stella
- CNRS IPBS (Institut de Pharmacologie et de Biologie Structurale), Toulouse, France Université de Toulouse UPS IPBS, Toulouse, France
| | - Bernard Monsarrat
- CNRS IPBS (Institut de Pharmacologie et de Biologie Structurale), Toulouse, France Université de Toulouse UPS IPBS, Toulouse, France
| | - Benoît Van den Eynde
- Ludwig Institute for Cancer Research, Brussels, Belgium WELBIO (Walloon Excellence in Life Sciences and Biotechnology), Brussels, Belgium de Duve Institute Université catholique de Louvain, Brussels, Belgium
| | - Odile Burlet-Schiltz
- CNRS IPBS (Institut de Pharmacologie et de Biologie Structurale), Toulouse, France Université de Toulouse UPS IPBS, Toulouse, France
| | - Marie-Pierre Bousquet-Dubouch
- CNRS IPBS (Institut de Pharmacologie et de Biologie Structurale), Toulouse, France Université de Toulouse UPS IPBS, Toulouse, France
| |
Collapse
|
87
|
Meiners S, Keller IE, Semren N, Caniard A. Regulation of the proteasome: evaluating the lung proteasome as a new therapeutic target. Antioxid Redox Signal 2014; 21:2364-82. [PMID: 24437504 DOI: 10.1089/ars.2013.5798] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
SIGNIFICANCE Lung diseases are on the second rank worldwide with respect to morbidity and mortality. For most respiratory diseases, no effective therapies exist. Whereas the proteasome has been successfully evaluated as a novel target for therapeutic interventions in cancer, neurodegenerative, and cardiac disorders, there is a profound lack of knowledge on the regulation of proteasome activity in chronic and acute lung diseases. RECENT ADVANCES There are various means of how the amount of active proteasome complexes in the cell can be regulated such as transcriptional regulation of proteasomal subunit expression, association with different regulators, assembly and half-life of proteasomes and regulatory complexes, as well as post-translational modifications. It also becomes increasingly evident that proteasome activity is fine-tuned and depends on the state of the cell. We propose here that 20S proteasomes and their regulators can be regarded as dynamic building blocks, which assemble or disassemble in response to cellular needs. The composition of proteasome complexes in a cell may vary depending on tissue, cell type and compartment, stage of development, or pathological context. CRITICAL ISSUES AND FUTURE DIRECTIONS Dissecting the expression and regulation of the various catalytic forms of 20S proteasomes, such as constitutive, immuno-, and mixed proteasomes, together with their associated regulatory complexes will not only greatly enhance our understanding of proteasome function in lung pathogenesis but will also pave the way to develop new classes of drugs that inhibit or activate proteasome function in a defined setting for treatment of lung diseases.
Collapse
Affiliation(s)
- Silke Meiners
- Comprehensive Pneumology Center (CPC), University Hospital , Ludwig-Maximilians University, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | | | | | | |
Collapse
|
88
|
Role of peptide processing predictions in T cell epitope identification: contribution of different prediction programs. Immunogenetics 2014; 67:85-93. [PMID: 25475908 PMCID: PMC4297296 DOI: 10.1007/s00251-014-0815-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Accepted: 11/10/2014] [Indexed: 10/27/2022]
Abstract
Proteolysis is the general term to describe the process of protein degradation into peptides. Proteasomes are the main actors in cellular proteolysis, and their activity can be measured in in vitro digestion experiments. However, in vivo proteolysis can be different than what is measured in these experiments if other proteases participate or if proteasomal activity is different in vivo. The in vivo proteolysis can be measured only indirectly, by the analysis of peptides presented on MHC-I molecules. MHC-I presented peptides are protected from further degradation, thus enabling an indirect view on the underlying in vivo proteolysis. The ligands presented on different MHC-I molecules enable different views on this process; in combination, they might give a complete picture. Based on in vitro proteasome-only digestions and MHC-I ligand data, different proteolysis predictors have been developed. With new in vitro digestion and MHC-I ligand data sets, we benchmarked how well these predictors capture in vitro proteasome-only activity and in vivo whole-cell proteolysis, respectively. Even though the in vitro proteasome digestion patterns were best captured by methods trained on such data (ProteaSMM and NetChop 20S), the in vivo whole-cell proteolysis was best predicted by a method trained on MHC-I ligand data (NetChop Cterm). Follow-up analysis showed that the likely source of this difference is the activity from proteases other than the proteasome, such as TPPII. This non-proteasomal in vivo activity is captured by NetChop Cterm and should be taken into account in MHC-I ligand predictions.
Collapse
|
89
|
Mishto M, Liepe J, Textoris-Taube K, Keller C, Henklein P, Weberruß M, Dahlmann B, Enenkel C, Voigt A, Kuckelkorn U, Stumpf MPH, Kloetzel PM. Proteasome isoforms exhibit only quantitative differences in cleavage and epitope generation. Eur J Immunol 2014; 44:3508-21. [PMID: 25231383 DOI: 10.1002/eji.201444902] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Revised: 08/01/2014] [Accepted: 09/12/2014] [Indexed: 11/09/2022]
Abstract
Immunoproteasomes are considered to be optimised to process Ags and to alter the peptide repertoire by generating a qualitatively different set of MHC class I epitopes. Whether the immunoproteasome at the biochemical level, influence the quality rather than the quantity of the immuno-genic peptide pool is still unclear. Here, we quantified the cleavage-site usage by human standard- and immunoproteasomes, and proteasomes from immuno-subunit-deficient mice, as well as the peptides generated from model polypeptides. We show in this study that the different proteasome isoforms can exert significant quantitative differences in the cleavage-site usage and MHC class I restricted epitope production. However, independent of the proteasome isoform and substrates studied, no evidence was obtained for the abolishment of the specific cleavage-site usage, or for differences in the quality of the peptides generated. Thus, we conclude that the observed differences in MHC class I restricted Ag presentation between standard- and immunoproteasomes are due to quantitative differences in the proteasome-generated antigenic peptides.
Collapse
Affiliation(s)
- Michele Mishto
- Institut für Biochemie, Charité - Universitätsmedizin Berlin, Berlin, Germany; Centro Interdipartimentale di Ricerca sul Cancro "Giorgio Prodi,", University of Bologna, Bologna, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
90
|
Proteasome subtypes and regulators in the processing of antigenic peptides presented by class I molecules of the major histocompatibility complex. Biomolecules 2014; 4:994-1025. [PMID: 25412285 PMCID: PMC4279167 DOI: 10.3390/biom4040994] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Revised: 10/02/2014] [Accepted: 10/29/2014] [Indexed: 02/07/2023] Open
Abstract
The proteasome is responsible for the breakdown of cellular proteins. Proteins targeted for degradation are allowed inside the proteasome particle, where they are cleaved into small peptides and released in the cytosol to be degraded into amino acids. In vertebrates, some of these peptides escape degradation in the cytosol, are loaded onto class I molecules of the major histocompatibility complex (MHC) and displayed at the cell surface for scrutiny by the immune system. The proteasome therefore plays a key role for the immune system: it provides a continued sampling of intracellular proteins, so that CD8-positive T-lymphocytes can kill cells expressing viral or tumoral proteins. Consequently, the repertoire of peptides displayed by MHC class I molecules at the cell surface depends on proteasome activity, which may vary according to the presence of proteasome subtypes and regulators. Besides standard proteasomes, cells may contain immunoproteasomes, intermediate proteasomes and thymoproteasomes. Cells may also contain regulators of proteasome activity, such as the 19S, PA28 and PA200 regulators. Here, we review the effects of these proteasome subtypes and regulators on the production of antigenic peptides. We also discuss an unexpected function of the proteasome discovered through the study of antigenic peptides: its ability to splice peptides.
Collapse
|
91
|
Mel’nikova VI, Khegai II, Popova NA, Lifantseva NV, Ivanova LN, Zakharova LA. Features of the immune proteasome expression in ascite Zajdela hepatoma after implantation into Brattleboro rats with the hereditary defect of arginine-vasopressin synthesis. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2014. [DOI: 10.1134/s1068162014060107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
92
|
Ferreira de Lima Neto D, Bonafe CFS, Arns CW. Influence of high hydrostatic pressure on epitope mapping of tobacco mosaic virus coat protein. Viral Immunol 2014; 27:60-74. [PMID: 24605789 DOI: 10.1089/vim.2013.0088] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
In this study, we investigated the effect of high hydrostatic pressure (HHP) on tobacco mosaic virus (TMV), a model virus in immunology and one of the most studied viruses to date. Exposure to HHP significantly altered the recognition epitopes when compared to sera from mice immunized with native virus. These alterations were studied further by combining HHP with urea or low temperature and then inoculating the altered virions into Balb-C mice. The antibody titers and cross-reactivity of the resulting sera were determined by ELISA. The antigenicity of the viral particles was maintained, as assessed by using polyclonal antibodies against native virus. The antigenicity of canonical epitopes was maintained, although binding intensities varied among the treatments. The patterns of recognition determined by epitope mapping were cross checked with the prediction algorithms for the TMVcp amino acid sequence to infer which alterations had occurred. These findings suggest that different cleavage sites were exposed after the treatments and this was confirmed by epitope mapping using sera from mice immunized with virus previously exposed to HHP.
Collapse
Affiliation(s)
- Daniel Ferreira de Lima Neto
- 1 Laboratório de Virologia Animal, Departamentos de 1Genética, Evolução e Bioagentes, e Universidade Estadual de Campinas (UNICAMP) , Cidade Universitária Zeferino Vaz, Campinas, SP, Brazil
| | | | | |
Collapse
|
93
|
Ammendola M, Leporini C, Marech I, Gadaleta CD, Scognamillo G, Sacco R, Sammarco G, De Sarro G, Russo E, Ranieri G. Targeting mast cells tryptase in tumor microenvironment: a potential antiangiogenetic strategy. BIOMED RESEARCH INTERNATIONAL 2014; 2014:154702. [PMID: 25295247 PMCID: PMC4177740 DOI: 10.1155/2014/154702] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Revised: 08/14/2014] [Accepted: 08/19/2014] [Indexed: 12/13/2022]
Abstract
Angiogenesis is a complex process finely regulated by the balance between angiogenesis stimulators and inhibitors. As a result of proangiogenic factors overexpression, it plays a crucial role in cancer development. Although initially mast cells (MCs) role has been defined in hypersensitivity reactions and in immunity, it has been discovered that MCs have a crucial interplay on the regulatory function between inflammatory and tumor cells through the release of classical proangiogenic factors (e.g., vascular endothelial growth factor) and nonclassical proangiogenic mediators granule-associated (mainly tryptase). In fact, in several animal and human malignancies, MCs density is highly correlated with tumor angiogenesis. In particular, tryptase, an agonist of the proteinase-activated receptor-2 (PAR-2), represents one of the most powerful angiogenic mediators released by human MCs after c-Kit receptor activation. This protease, acting on PAR-2 by its proteolytic activity, has angiogenic activity stimulating both human vascular endothelial and tumor cell proliferation in paracrine manner, helping tumor cell invasion and metastasis. Based on literature data it is shown that tryptase may represent a promising target in cancer treatment due to its proangiogenic activity. Here we focused on molecular mechanisms of three tryptase inhibitors (gabexate mesylate, nafamostat mesylate, and tranilast) in order to consider their prospective role in cancer therapy.
Collapse
Affiliation(s)
- Michele Ammendola
- Department of Medical and Surgery Sciences, Clinical Surgery Unit, University “Magna Graecia” Medical School, Viale Europa, Germaneto, 88100 Catanzaro, Italy
| | - Christian Leporini
- Department of Health Science, Clinical Pharmacology and Pharmacovigilance Unit and Pharmacovigilance's Centre Calabria Region, University of Catanzaro “Magna Graecia” Medical School, Viale Europa, Germaneto, 88100 Catanzaro, Italy
| | - Ilaria Marech
- Diagnostic and Interventional Radiology Unit with Integrated Section of Translational Medical Oncology, Istituto Tumori “Giovanni Paolo II,” Viale Orazio Flacco 65, 70124 Bari, Italy
| | - Cosmo Damiano Gadaleta
- Diagnostic and Interventional Radiology Unit with Integrated Section of Translational Medical Oncology, Istituto Tumori “Giovanni Paolo II,” Viale Orazio Flacco 65, 70124 Bari, Italy
| | - Giovanni Scognamillo
- Radiotherapy Unit, Istituto Tumori “Giovanni Paolo II,” Viale Orazio Flacco 65, 70124 Bari, Italy
| | - Rosario Sacco
- Department of Medical and Surgery Sciences, Clinical Surgery Unit, University “Magna Graecia” Medical School, Viale Europa, Germaneto, 88100 Catanzaro, Italy
| | - Giuseppe Sammarco
- Department of Medical and Surgery Sciences, Clinical Surgery Unit, University “Magna Graecia” Medical School, Viale Europa, Germaneto, 88100 Catanzaro, Italy
| | - Giovambattista De Sarro
- Department of Health Science, Clinical Pharmacology and Pharmacovigilance Unit and Pharmacovigilance's Centre Calabria Region, University of Catanzaro “Magna Graecia” Medical School, Viale Europa, Germaneto, 88100 Catanzaro, Italy
| | - Emilio Russo
- Department of Health Science, Clinical Pharmacology and Pharmacovigilance Unit and Pharmacovigilance's Centre Calabria Region, University of Catanzaro “Magna Graecia” Medical School, Viale Europa, Germaneto, 88100 Catanzaro, Italy
| | - Girolamo Ranieri
- Diagnostic and Interventional Radiology Unit with Integrated Section of Translational Medical Oncology, Istituto Tumori “Giovanni Paolo II,” Viale Orazio Flacco 65, 70124 Bari, Italy
| |
Collapse
|
94
|
de Verteuil DA, Rouette A, Hardy MP, Lavallée S, Trofimov A, Gaucher É, Perreault C. Immunoproteasomes Shape the Transcriptome and Regulate the Function of Dendritic Cells. THE JOURNAL OF IMMUNOLOGY 2014; 193:1121-32. [DOI: 10.4049/jimmunol.1400871] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
95
|
Steers NJ, Currier JR, Jobe O, Tovanabutra S, Ratto-Kim S, Marovich MA, Kim JH, Michael NL, Alving CR, Rao M. Designing the epitope flanking regions for optimal generation of CTL epitopes. Vaccine 2014; 32:3509-16. [DOI: 10.1016/j.vaccine.2014.04.039] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Revised: 01/15/2014] [Accepted: 04/17/2014] [Indexed: 12/25/2022]
|
96
|
Lu YF, Sheng H, Zhang Y, Li ZY. Computational prediction of cleavage using proteasomal in vitro digestion and MHC I ligand data. J Zhejiang Univ Sci B 2014; 14:816-28. [PMID: 24009202 DOI: 10.1631/jzus.b1200299] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Proteasomes are responsible for the production of the majority of cytotoxic T lymphocyte (CTL) epitopes. Hence, it is important to identify correctly which peptides will be generated by proteasomes from an unknown protein. However, the pool of proteasome cleavage data used in the prediction algorithms, whether from major histocompatibility complex (MHC) I ligand or in vitro digestion data, is not identical to in vivo proteasomal digestion products. Therefore, the accuracy and reliability of these models still need to be improved. In this paper, three types of proteasomal cleavage data, constitutive proteasome (cCP), immunoproteasome (iCP) in vitro cleavage, and MHC I ligand data, were used for training cleave-site predictive methods based on the kernel-function stabilized matrix method (KSMM). The predictive accuracies of the KSMM+pair coefficients were 75.0%, 72.3%, and 83.1% for cCP, iCP, and MHC I ligand data, respectively, which were comparable to the results from support vector machine (SVM). The three proteasomal cleavage methods were combined in turn with MHC I-peptide binding predictions to model MHC I-peptide processing and the presentation pathway. These integrations markedly improved MHC I peptide identification, increasing area under the receiver operator characteristics (ROC) curve (AUC) values from 0.82 to 0.91. The results suggested that both MHC I ligand and proteasomal in vitro degradation data can give an exact simulation of in vivo processed digestion. The information extracted from cCP and iCP in vitro cleavage data demonstrated that both cCP and iCP are selective in their usage of peptide bonds for cleavage.
Collapse
Affiliation(s)
- Yu-feng Lu
- School of Mathematical Sciences, Dalian University of Technology, Dalian 116023, China; College of Science, Hebei University of Science and Technology, Shijiazhuang 050018, China; School of Information Science and Technology, Dalian Maritime University, Dalian 116026, China
| | | | | | | |
Collapse
|
97
|
Payne RP, Branch S, Kløverpris H, Matthews PC, Koofhethile CK, Strong T, Adland E, Leitman E, Frater J, Ndung'u T, Hunter E, Haubrich R, Mothe B, Edwards A, Riddell L, Chen F, Harrigan PR, Brumme ZL, Mallal S, John M, Jooste JP, Shapiro R, Deeks SG, Walker BD, Brander C, Landis C, Carlson JM, Prado JG, Goulder PJR. Differential escape patterns within the dominant HLA-B*57:03-restricted HIV Gag epitope reflect distinct clade-specific functional constraints. J Virol 2014; 88:4668-78. [PMID: 24501417 PMCID: PMC3993828 DOI: 10.1128/jvi.03303-13] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Accepted: 02/02/2014] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED HLA-B*57:01 and HLA-B*57:03, the most prevalent HLA-B*57 subtypes in Caucasian and African populations, respectively, are the HLA alleles most protective against HIV disease progression. Understanding the mechanisms underlying this immune control is of critical importance, yet they remain unclear. Unexplained differences are observed in the impact of the dominant cytotoxic T lymphocyte (CTL) response restricted by HLA-B*57:01 and HLA-B*57:03 in chronic infection on the Gag epitope KAFSPEVIPMF (KF11; Gag 162 to 172). We previously showed that the HLA-B*57:03-KF11 response is associated with a >1-log-lower viral setpoint in C clade virus infection and that this response selects escape mutants within the epitope. We first examined the relationship of KF11 responses in B clade virus-infected subjects with HLA-B*57:01 to immune control and observed that a detectable KF11 response was associated with a >1-log-higher viral load (P = 0.02). No evidence of HLA-B*57:01-KF11-associated selection pressure was identified in previous comprehensive analyses of >1,800 B clade virus-infected subjects. We then studied a B clade virus-infected cohort in Barbados, where HLA-B*57:03 is highly prevalent. In contrast to findings for B clade virus-infected subjects expressing HLA-B*57:01, we observed strong selection pressure driven by the HLA-B*57:03-KF11 response for the escape mutation S173T. This mutation reduces recognition of virus-infected cells by HLA-B*57:03-KF11 CTLs and is associated with a >1-log increase in viral load in HLA-B*57:03-positive subjects (P = 0.009). We demonstrate functional constraints imposed by HIV clade relating to the residue at Gag 173 that explain the differential clade-specific escape patterns in HLA-B*57:03 subjects. Further studies are needed to evaluate the role of the KF11 response in HLA-B*57:01-associated HIV disease protection. IMPORTANCE HLA-B*57 is the HLA class I molecule that affords the greatest protection against disease progression in HIV infection. Understanding the key mechanism(s) underlying immunosuppression of HIV is of importance in guiding therapeutic and vaccine-related approaches to improve the levels of HIV control occurring in nature. Numerous mechanisms have been proposed to explain the HLA associations with differential HIV disease outcome, but no consensus exists. These studies focus on two subtypes of HLA-B*57 prevalent in Caucasian and African populations, HLA-B*57:01 and HLA-B*57:03, respectively. These alleles appear equally protective against HIV disease progression. The CTL epitopes presented are in many cases identical, and the dominant response in chronic infection in each case is to the Gag epitope KF11. However, there the similarity ends. This study sought to better understand the reasons for these differences and what they teach us about which immune responses contribute to immune control of HIV infection.
Collapse
Affiliation(s)
- R. P. Payne
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - S. Branch
- Ladymeade Reference Unit, Queen Elizabeth Hospital, Bridgetown, Barbados
| | - H. Kløverpris
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
- K-RITH, University of Kwa-Zulu Natal, Duran, South Africa
| | - P. C. Matthews
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - C. K. Koofhethile
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - T. Strong
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - E. Adland
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - E. Leitman
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - J. Frater
- Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
- Oxford NIHR Biomedical Research Centre, Oxford, United Kingdom
| | - T. Ndung'u
- HIV Pathogenesis Program, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
| | - E. Hunter
- Emory Vaccine Center, Yerkes National Primate Research Centre, Emory University, Atlanta, Georgia, USA
- Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | - R. Haubrich
- Antiviral Research Center, University of California San Diego, San Diego, California, USA
| | - B. Mothe
- IrsiCaixa AIDS Research Institute-HIVCAT, Hospital Germans Trias i Pujol, Badalona, Spain
| | - A. Edwards
- Oxford Department of Genitourinary Medicine, The Churchill Hospital, Oxford, United Kingdom
| | - L. Riddell
- Department of Genitourinary Medicine, Northamptonshire Healthcare National Health Service Trust, Northampton General Hospital, Cliftonville, Northampton, United Kingdom
| | - F. Chen
- Department of Sexual Health, Royal Berkshire Hospital, Reading, United Kingdom
| | - P. R. Harrigan
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, British Columbia, Canada
| | - Z. L. Brumme
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, British Columbia, Canada
- Faculty of Health Sciences, Simon Fraser University, Burnaby, British Columbia, Canada
| | - S. Mallal
- Centre for Clinical Immunology and Biomedical Statistics, Institute for Immunology and Infectious Diseases, Murdoch University, Murdoch, Western Australia, Australia
- Department of Clinical Immunology, Royal Perth Hospital, Perth, Western Australia, Australia
| | - M. John
- Centre for Clinical Immunology and Biomedical Statistics, Institute for Immunology and Infectious Diseases, Murdoch University, Murdoch, Western Australia, Australia
- Department of Clinical Immunology, Royal Perth Hospital, Perth, Western Australia, Australia
| | - J. P. Jooste
- Paediatric Department, Kimberley Hospital, Northern Cape, South Africa
| | - R. Shapiro
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, Massachusetts, USA
| | - S. G. Deeks
- Department of Medicine, University of California, San Francisco, California, USA
| | - B. D. Walker
- HIV Pathogenesis Program, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
- Ragon Institute of MGH, MIT, and Harvard, AIDS Research Center, Charlestown, Massachusetts, USA
| | - C. Brander
- IrsiCaixa AIDS Research Institute-HIVCAT, Hospital Germans Trias i Pujol, Badalona, Spain
- Institució Catalana de Recerca i Estudis Avancats (ICREA), Barcelona, Spain
| | - C. Landis
- Ladymeade Reference Unit, Queen Elizabeth Hospital, Bridgetown, Barbados
| | - J. M. Carlson
- Microsoft Research, eScience Group, Los Angeles, California, USA
| | - J. G. Prado
- IrsiCaixa AIDS Research Institute-HIVCAT, Hospital Germans Trias i Pujol, Badalona, Spain
| | - P. J. R. Goulder
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
- HIV Pathogenesis Program, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
98
|
Tenzer S, Crawford H, Pymm P, Gifford R, Sreenu VB, Weimershaus M, de Oliveira T, Burgevin A, Gerstoft J, Akkad N, Lunn D, Fugger L, Bell J, Schild H, van Endert P, Iversen AKN. HIV-1 adaptation to antigen processing results in population-level immune evasion and affects subtype diversification. Cell Rep 2014; 7:448-463. [PMID: 24726370 PMCID: PMC4005910 DOI: 10.1016/j.celrep.2014.03.031] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Revised: 12/04/2013] [Accepted: 03/11/2014] [Indexed: 02/01/2023] Open
Abstract
The recent HIV-1 vaccine failures highlight the need to better understand virus-host interactions. One key question is why CD8(+) T cell responses to two HIV-Gag regions are uniquely associated with delayed disease progression only in patients expressing a few rare HLA class I variants when these regions encode epitopes presented by ~30 more common HLA variants. By combining epitope processing and computational analyses of the two HIV subtypes responsible for ~60% of worldwide infections, we identified a hitherto unrecognized adaptation to the antigen-processing machinery through substitutions at subtype-specific motifs. Multiple HLA variants presenting epitopes situated next to a given subtype-specific motif drive selection at this subtype-specific position, and epitope abundances correlate inversely with the HLA frequency distribution in affected populations. This adaptation reflects the sum of intrapatient adaptations, is predictable, facilitates viral subtype diversification, and increases global HIV diversity. Because low epitope abundance is associated with infrequent and weak T cell responses, this most likely results in both population-level immune evasion and inadequate responses in most people vaccinated with natural HIV-1 sequence constructs. Our results suggest that artificial sequence modifications at subtype-specific positions in vitro could refocus and reverse the poor immunogenicity of HIV proteins.
Collapse
Affiliation(s)
- Stefan Tenzer
- Institute of Immunology, University Medical Center of the Johannes-Gutenberg University of Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany
| | - Hayley Crawford
- Medical Research Council Human Immunology Unit, Weatherall Institute of Molecular Medicine, Oxford University, John Radcliffe Hospital, Headley Way, Oxford OX3 9DS, UK; Division of Clinical Neurology, Nuffield Department of Clinical Neurosciences, Weatherall Institute of Molecular Medicine, Oxford University, John Radcliffe Hospital, Headley Way, Oxford OX3 9DS, UK
| | - Phillip Pymm
- Medical Research Council Human Immunology Unit, Weatherall Institute of Molecular Medicine, Oxford University, John Radcliffe Hospital, Headley Way, Oxford OX3 9DS, UK; Division of Clinical Neurology, Nuffield Department of Clinical Neurosciences, Weatherall Institute of Molecular Medicine, Oxford University, John Radcliffe Hospital, Headley Way, Oxford OX3 9DS, UK
| | - Robert Gifford
- Aaron Diamond AIDS Research Center, 455 First Avenue, New York, NY 10016, USA
| | - Vattipally B Sreenu
- Medical Research Council Human Immunology Unit, Weatherall Institute of Molecular Medicine, Oxford University, John Radcliffe Hospital, Headley Way, Oxford OX3 9DS, UK
| | - Mirjana Weimershaus
- Institut National de la Santé et de la Recherche Médicale, Unité 1151, Centre National de la Recherche Scientifique, UMR8253, Université Paris Descartes, Sorbonne Paris Cité, Hôpital Necker, 149 rue de Sèvres, 75015 Paris, France
| | - Tulio de Oliveira
- Africa Centre for Health and Population Studies, School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, KwaZulu-Natal 3935, South Africa; Research Department of Infection, University College London, Cruciform Building, 90 Gower Street, London WC1E 6BT, UK
| | - Anne Burgevin
- Institut National de la Santé et de la Recherche Médicale, Unité 1151, Centre National de la Recherche Scientifique, UMR8253, Université Paris Descartes, Sorbonne Paris Cité, Hôpital Necker, 149 rue de Sèvres, 75015 Paris, France
| | - Jan Gerstoft
- Department of Infectious Diseases, Rigshospitalet, The National University Hospital, Blegdamsvej 9, 2100 Kbh Ø Copenhagen, Denmark
| | - Nadja Akkad
- Institute of Immunology, University Medical Center of the Johannes-Gutenberg University of Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany
| | - Daniel Lunn
- Department of Statistics, University of Oxford, 1 South Parks Road, Oxford OX1 3TG, UK
| | - Lars Fugger
- Medical Research Council Human Immunology Unit, Weatherall Institute of Molecular Medicine, Oxford University, John Radcliffe Hospital, Headley Way, Oxford OX3 9DS, UK; Division of Clinical Neurology, Nuffield Department of Clinical Neurosciences, Weatherall Institute of Molecular Medicine, Oxford University, John Radcliffe Hospital, Headley Way, Oxford OX3 9DS, UK
| | - John Bell
- Office of the Regius Professor of Medicine, The Richard Doll Building, University of Oxford, Old Road Campus, Roosevelt Drive 1, Oxford OX3 7LF, UK
| | - Hansjörg Schild
- Institute of Immunology, University Medical Center of the Johannes-Gutenberg University of Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany
| | - Peter van Endert
- Institut National de la Santé et de la Recherche Médicale, Unité 1151, Centre National de la Recherche Scientifique, UMR8253, Université Paris Descartes, Sorbonne Paris Cité, Hôpital Necker, 149 rue de Sèvres, 75015 Paris, France
| | - Astrid K N Iversen
- Medical Research Council Human Immunology Unit, Weatherall Institute of Molecular Medicine, Oxford University, John Radcliffe Hospital, Headley Way, Oxford OX3 9DS, UK; Division of Clinical Neurology, Nuffield Department of Clinical Neurosciences, Weatherall Institute of Molecular Medicine, Oxford University, John Radcliffe Hospital, Headley Way, Oxford OX3 9DS, UK.
| |
Collapse
|
99
|
Basler M, Mundt S, Muchamuel T, Moll C, Jiang J, Groettrup M, Kirk CJ. Inhibition of the immunoproteasome ameliorates experimental autoimmune encephalomyelitis. EMBO Mol Med 2014; 6:226-38. [PMID: 24399752 PMCID: PMC3927957 DOI: 10.1002/emmm.201303543] [Citation(s) in RCA: 139] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic demyelinating immune mediated disease of the central nervous system. The immunoproteasome is a distinct class of proteasomes found predominantly in monocytes and lymphocytes. Recently, we demonstrated a novel function of immunoproteasomes in cytokine production and T cell differentiation. In this study, we investigated the therapeutic efficacy of an inhibitor of the immunoproteasome (ONX 0914) in two different mouse models of MS. ONX 0914 attenuated disease progression after active and passive induction of experimental autoimmune encephalomyelitis (EAE), both in MOG35–55 and PLP139–151-induced EAE. Isolation of lymphocytes from the brain or spinal cord revealed a strong reduction of cytokine-producing CD4+ cells in ONX 0914 treated mice. Additionally, ONX 0914 treatment prevented disease exacerbation in a relapsing-remitting model. An analysis of draining lymph nodes after induction of EAE revealed that the differentiation to Th17 or Th1 cells was strongly impaired in ONX 0914 treated mice. These results implicate the immunoproteasome in the development of EAE and suggest that immunoproteasome inhibitors are promising drugs for the treatment of MS.
Collapse
Affiliation(s)
- Michael Basler
- Biotechnology Institute Thurgau (BITg) at the University of Konstanz, Kreuzlingen, Switzerland
| | | | | | | | | | | | | |
Collapse
|
100
|
Karpenko LI, Bazhan SI, Antonets DV, Belyakov IM. Novel approaches in polyepitope T-cell vaccine development against HIV-1. Expert Rev Vaccines 2013; 13:155-73. [PMID: 24308576 DOI: 10.1586/14760584.2014.861748] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
RV144 clinical trial was modestly effective in preventing HIV infection. New alternative approaches are needed to design improved HIV-1 vaccines and their delivery strategies. One of these approaches is construction of synthetic polyepitope HIV-1 immunogen using protective T- and B-cell epitopes that can induce broadly neutralizing antibodies and responses of cytotoxic (CD8(+) CTL) and helpers (CD4(+) Th) T-lymphocytes. This approach seems to be promising for designing of new generation of vaccines against HIV-1, enables in theory to cope with HIV-1 antigenic variability, focuses immune responses on protective determinants and enables to exclude from the vaccine compound that can induce autoantibodies or antibodies enhancing HIV-1 infectivity. Herein, the authors will focus on construction and rational design of polyepitope T-cell HIV-1 immunogens and their delivery, including: advantages and disadvantages of existing T-cell epitope prediction methods; features of organization of polyepitope immunogens, which can generate high-level CD8(+) and CD4(+) T-lymphocyte responses; the strategies to optimize efficient processing, presentation and immunogenicity of polyepitope constructs; original software to design polyepitope immunogens; and delivery vectors as well as mucosal strategies of vaccination. This new knowledge may bring us a one step closer to developing an effective T-cell vaccine against HIV-1, other chronic viral infections and cancer.
Collapse
Affiliation(s)
- Larisa I Karpenko
- State Research Center of Virology and Biotechnology "Vector", Koltsovo, Novosibirsk region, 630559, Russia
| | | | | | | |
Collapse
|