1
|
Mehanna N, Pradhan A, Kaur R, Kontopoulos T, Rosati B, Carlson D, Cheung NKV, Xu H, Bean J, Hsu KC, Le Luduec JB, Vorkas CK. CD8α marks a Mycobacterium tuberculosis-reactive human NK cell population with high activation potential. Sci Rep 2025; 15:15095. [PMID: 40301594 PMCID: PMC12041513 DOI: 10.1038/s41598-025-98367-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 04/10/2025] [Indexed: 05/01/2025] Open
Abstract
Natural Killer (NK) cells can recognize and kill Mycobacterium tuberculosis (Mtb)-infected cells in vitro, however their role after natural human exposure has not been well-studied. To identify Mtb-responsive NK cell populations, we analyzed the peripheral blood of healthy household contacts of active Tuberculosis (TB) cases and source community donors in an endemic region of Port-au-Prince, Haiti by flow cytometry. We observed higher CD8α expression on NK cells in putative resistors (Interferon γ release assay negative; IGRA- contacts) with a loss of CD8α surface expression during household-associated exposure and active TB disease. In vitro assays and CITE-seq analysis of CD8α+ NK cells demonstrated enhanced maturity, cytotoxic gene expression, and response to cytokine stimulation relative to CD8α- NK cells. CD8α+ NK cells also displayed dynamic surface expression dependent on MHC class I in contrast to conventional CD8+ T cells. Together, these results support a specialized role for CD8α+ NK cell populations during Mtb infection correlating with disease resistance.
Collapse
Affiliation(s)
- Nezar Mehanna
- Renaissance School of Medicine at Stony Brook University, 101 Nicolls Road, Health Sciences Center 15060-I, Stony Brook, NY, 11794, USA
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, NY, 11794, USA
- Center for Infectious Diseases, Stony Brook University, Stony Brook, NY, 11794, USA
- Division of Infectious Diseases, Department of Medicine, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Atul Pradhan
- Renaissance School of Medicine at Stony Brook University, 101 Nicolls Road, Health Sciences Center 15060-I, Stony Brook, NY, 11794, USA
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, NY, 11794, USA
- Center for Infectious Diseases, Stony Brook University, Stony Brook, NY, 11794, USA
- Division of Infectious Diseases, Department of Medicine, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Rimanpreet Kaur
- Renaissance School of Medicine at Stony Brook University, 101 Nicolls Road, Health Sciences Center 15060-I, Stony Brook, NY, 11794, USA
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, NY, 11794, USA
- Center for Infectious Diseases, Stony Brook University, Stony Brook, NY, 11794, USA
- Division of Infectious Diseases, Department of Medicine, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Theodota Kontopoulos
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Barbara Rosati
- Renaissance School of Medicine at Stony Brook University, 101 Nicolls Road, Health Sciences Center 15060-I, Stony Brook, NY, 11794, USA
| | - David Carlson
- Renaissance School of Medicine at Stony Brook University, 101 Nicolls Road, Health Sciences Center 15060-I, Stony Brook, NY, 11794, USA
| | - Nai-Kong V Cheung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Hong Xu
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - James Bean
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Katharine C Hsu
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Jean-Benoit Le Luduec
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Charles Kyriakos Vorkas
- Renaissance School of Medicine at Stony Brook University, 101 Nicolls Road, Health Sciences Center 15060-I, Stony Brook, NY, 11794, USA.
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, NY, 11794, USA.
- Center for Infectious Diseases, Stony Brook University, Stony Brook, NY, 11794, USA.
- Division of Infectious Diseases, Department of Medicine, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, 11794, USA.
| |
Collapse
|
2
|
Dolton G, Thomas H, Tan LR, Rius Rafael C, Doetsch S, Ionescu GA, Cardo LF, Crowther MD, Behiry E, Morin T, Caillaud ME, Srai D, Parolini L, Hasan MS, Fuller A, Topley K, Wall A, Hopkins JR, Omidvar N, Alvares C, Zabkiewicz J, Frater J, Szomolay B, Sewell AK. MHC-related protein 1-restricted recognition of cancer via a semi-invariant TCR-α chain. J Clin Invest 2025; 135:e181895. [PMID: 39744940 PMCID: PMC11684821 DOI: 10.1172/jci181895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 10/29/2024] [Indexed: 01/16/2025] Open
Abstract
The T cell antigen presentation platform MR1 consists of 6 allomorphs in humans that differ by no more than 5 amino acids. The principal function of this highly conserved molecule involves presenting microbial metabolites to the abundant mucosal-associated invariant T (MAIT) cell subset. Recent developments suggest that the role of MR1 extends to presenting antigens from cancer cells, a function dependent on the K43 residue in the MR1 antigen binding cleft. Here, we successfully cultured cancer-activated, MR1-restricted T cells from multiple donors and confirmed that they recognized a wide range of cancer types expressing the most common MR1*01 and/or MR1*02 allomorphs (over 95% of the population), while remaining inert to healthy cells including healthy B cells and monocytes. Curiously, in all but one donor these T cells were found to incorporate a conserved TCR-α chain motif, CAXYGGSQGNLIF (where X represents 3-5 amino acids), because of pairing between 10 different TRAV genes and the TRAJ42 gene segment. This semi-invariance in the TCR-α chain is reminiscent of MAIT cells and suggests recognition of a conserved antigen bound to K43.
Collapse
MESH Headings
- Humans
- Minor Histocompatibility Antigens/genetics
- Minor Histocompatibility Antigens/immunology
- Minor Histocompatibility Antigens/metabolism
- Histocompatibility Antigens Class I/immunology
- Histocompatibility Antigens Class I/genetics
- Histocompatibility Antigens Class I/metabolism
- Neoplasms/immunology
- Neoplasms/genetics
- Neoplasms/metabolism
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- Receptors, Antigen, T-Cell, alpha-beta/metabolism
- Mucosal-Associated Invariant T Cells/immunology
- Mucosal-Associated Invariant T Cells/metabolism
- Antigen Presentation
- Antigens, Neoplasm/immunology
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/metabolism
- Amino Acid Motifs
- Cell Line, Tumor
Collapse
Affiliation(s)
- Garry Dolton
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Hannah Thomas
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Li Rong Tan
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Cristina Rius Rafael
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Stephanie Doetsch
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Giulia-Andreea Ionescu
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Lucia F. Cardo
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Michael D. Crowther
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Enas Behiry
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Théo Morin
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Marine E. Caillaud
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Devinder Srai
- Nuffield Department of Medicine and Department of Chemistry, University of Oxford, Oxford, United Kingdom
| | - Lucia Parolini
- Nuffield Department of Medicine and Department of Chemistry, University of Oxford, Oxford, United Kingdom
| | - Md Samiul Hasan
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Anna Fuller
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Katie Topley
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Aaron Wall
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Jade R. Hopkins
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Nader Omidvar
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Caroline Alvares
- Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Joanna Zabkiewicz
- Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - John Frater
- Nuffield Department of Medicine and NIHR Biomedical Research Centre University of Oxford, Oxford, United Kingdom
| | - Barbara Szomolay
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
- Systems Immunology Research Institute, Cardiff University Cardiff, United Kingdom
| | - Andrew K. Sewell
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
- Systems Immunology Research Institute, Cardiff University Cardiff, United Kingdom
- Division of Infection and Immunity, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
3
|
Ringl F, Stadler M, van Dongen KA, Adib Razavi M, Saalmüller A, Mair KH. Solving technical issues in flow cytometry to characterize porcine CD8α/β expressing lymphocytes. Vet Immunol Immunopathol 2024; 278:110853. [PMID: 39500097 DOI: 10.1016/j.vetimm.2024.110853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 10/13/2024] [Accepted: 10/21/2024] [Indexed: 11/26/2024]
Abstract
The CD8 molecule is a cell surface receptor and well described as co-receptor on T cells, binding directly to the major histocompatibility complex class I on antigen presenting cells. CD8 antigens are comprised of two distinct polypeptide chains, the α and the β chain. In the pig, the CD8 receptor is expressed by several lymphocyte subsets, including Natural Killer cells, γδ T cells and antigen experienced CD4+ αβ T cells. On these cell populations CD8 is expressed as αα homodimers. Porcine cytolytic T cells on the other hand exclusively express CD8 αβ heterodimers. Several monoclonal antibodies (mAbs) for either of the two chains are available and are frequently used in flow cytometry. We observed that distinct combinations of mAb clones for CD8α and CD8β chains can cause troubles in multi-color staining panels. Therefore, we aimed for an in-depth study of the usage of different CD8-specific mAb clones and optimizing co-staining strategies for flow cytometry. We tested mAb clones 11/295/33 and 76-2-11 for the detection of CD8α and mAb clones PPT23 and PG164A for the detection of CD8β. The results indicate that the CD8α clone 11/295/33 should not be used together with either of the two CD8β clones in the same incubation step, as co-staining led to a highly reduced ability of CD8β mAb binding and loss in signal in flow cytometry. This can lead to potential false results in detecting CD8αβ cytolytic T cells. In case of the CD8α mAb clone 76-2-11, no inhibition in binding of either CD8β mAb clones was observed, making it the preferred choice in multi-color staining panels. The obtained data will help in future panel designs for flow cytometry in the pig and therefore improving studies of porcine immune cells.
Collapse
Affiliation(s)
- Florian Ringl
- Christian Doppler Laboratory for Optimized Prediction of Vaccination Success in Pigs, Immunology, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Maria Stadler
- Immunology, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Katinka A van Dongen
- Christian Doppler Laboratory for Optimized Prediction of Vaccination Success in Pigs, Immunology, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Mahsa Adib Razavi
- Christian Doppler Laboratory for Optimized Prediction of Vaccination Success in Pigs, Immunology, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Armin Saalmüller
- Immunology, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Kerstin H Mair
- Christian Doppler Laboratory for Optimized Prediction of Vaccination Success in Pigs, Immunology, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria; Immunology, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria.
| |
Collapse
|
4
|
Srinivasan S, Zhu C, McShan AC. Structure, function, and immunomodulation of the CD8 co-receptor. Front Immunol 2024; 15:1412513. [PMID: 39253084 PMCID: PMC11381289 DOI: 10.3389/fimmu.2024.1412513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 08/05/2024] [Indexed: 09/11/2024] Open
Abstract
Expressed on the surface of CD8+ T cells, the CD8 co-receptor is a key component of the T cells that contributes to antigen recognition, immune cell maturation, and immune cell signaling. While CD8 is widely recognized as a co-stimulatory molecule for conventional CD8+ αβ T cells, recent reports highlight its multifaceted role in both adaptive and innate immune responses. In this review, we discuss the utility of CD8 in relation to its immunomodulatory properties. We outline the unique structure and function of different CD8 domains (ectodomain, hinge, transmembrane, cytoplasmic tail) in the context of the distinct properties of CD8αα homodimers and CD8αβ heterodimers. We discuss CD8 features commonly used to construct chimeric antigen receptors for immunotherapy. We describe the molecular interactions of CD8 with classical MHC-I, non-classical MHCs, and Lck partners involved in T cell signaling. Engineered and naturally occurring CD8 mutations that alter immune responses are discussed. The applications of anti-CD8 monoclonal antibodies (mABs) that target CD8 are summarized. Finally, we examine the unique structure and function of several CD8/mAB complexes. Collectively, these findings reveal the promising immunomodulatory properties of CD8 and CD8 binding partners, not only to uncover basic immune system function, but to advance efforts towards translational research for targeted immunotherapy.
Collapse
Affiliation(s)
- Shreyaa Srinivasan
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA, United States
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States
| | - Cheng Zhu
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, United States
| | - Andrew C. McShan
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA, United States
| |
Collapse
|
5
|
Esgalhado AJ, Reste-Ferreira D, Weinhold S, Uhrberg M, Cardoso EM, Arosa FA. In vitro IL-15-activated human naïve CD8+ T cells down-modulate the CD8β chain and become CD8αα T cells. Front Immunol 2024; 15:1252439. [PMID: 38903513 PMCID: PMC11188365 DOI: 10.3389/fimmu.2024.1252439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 05/21/2024] [Indexed: 06/22/2024] Open
Abstract
Antigen-driven human effector-memory CD8+ T cells expressing low levels of the CD8β chain have been previously described. However, little is known on a possible antigen-independent trigger. We have examined the impact that IL-15 has on the expression of CD8β on purified human naïve CD8+ T cells after CFSE labeling and culture with IL-15. As expected, IL-15 induced naïve CD8+ T cells to proliferate and differentiate. Remarkably, the process was associated with a cell-cycle dependent down-modulation of CD8β from the cell surface, leading to the generation of CD8αβlow and CD8αβ- (i.e., CD8αα) T cells. In contrast, expression of the CD8α chain remained steady or even increased. Neither IL-2 nor IL-7 reproduced the effect of IL-15. Determination of mRNA levels for CD8α and CD8β isoforms by qPCR revealed that IL-15 promoted a significant decrease in mRNA levels of the CD8β M-4 isoform, while levels of the M-1/M-2 isoforms and of CD8α increased. Noteworthy, CD8+ T cell blasts obtained after culture of CD8+ T cells with IL-15 showed a cell-cycle dependent increase in the level of the tyrosine kinase Lck, when compared to CD8+ T cells at day 0. This study has shown for the first time that IL-15 generates CD8αα+αβlow and CD8αα+αβ- T cells containing high levels of Lck, suggesting that they may be endowed with unique functional features.
Collapse
Affiliation(s)
- André J. Esgalhado
- Health Sciences Research Centre, University of Beira Interior (CICS-UBI), Covilhã, Portugal
| | - Débora Reste-Ferreira
- Health Sciences Research Centre, University of Beira Interior (CICS-UBI), Covilhã, Portugal
| | - Sandra Weinhold
- Institute for Transplantation Diagnostics and Cell Therapeutics, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany
| | - Markus Uhrberg
- Institute for Transplantation Diagnostics and Cell Therapeutics, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany
| | - Elsa M. Cardoso
- Health Sciences Research Centre, University of Beira Interior (CICS-UBI), Covilhã, Portugal
- School of Health Sciences, Polytechnic of Guarda (ESS-IPG), Guarda, Portugal
| | - Fernando A. Arosa
- Health Sciences Research Centre, University of Beira Interior (CICS-UBI), Covilhã, Portugal
- Faculty of Health Sciences, University of Beira Interior (FCS-UBI), Covilhã, Portugal
| |
Collapse
|
6
|
Mehanna N, Pradhan A, Kaur R, Kontopoulos T, Rosati B, Carlson D, Cheung NK, Xu H, Bean J, Hsu K, Le Luduec JB, Vorkas CK. Loss of circulating CD8α + NK cells during human Mycobacterium tuberculosis infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.16.588542. [PMID: 38659858 PMCID: PMC11042275 DOI: 10.1101/2024.04.16.588542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Natural Killer (NK) cells can recognize and kill Mtb-infected cells in vitro, however their role after natural human exposure has not been well-studied. To identify Mtb-responsive NK cell populations, we analyzed the peripheral blood of healthy household contacts of active Tuberculosis (TB) cases and source community donors in an endemic region of Port-au-Prince, Haiti by flow cytometry. We observed higher CD8α expression on NK cells in putative resistors (IGRA- contacts) with a progressive loss of these circulating cells during household-associated latent infection and disease. In vitro assays and CITE-seq analysis of CD8α+ NK cells demonstrated enhanced maturity, cytotoxic gene expression, and response to cytokine stimulation relative to CD8α- NK cells. CD8α+ NK cells also displayed dynamic surface expression dependent on MHC I in contrast to conventional CD8+ T cells. Together, these results support a specialized role for CD8α+ NK cell populations during Mtb infection correlating with disease resistance.
Collapse
Affiliation(s)
- Nezar Mehanna
- Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, 11794
| | - Atul Pradhan
- Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, 11794
| | - Rimanpreet Kaur
- Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, 11794
| | - Theodota Kontopoulos
- Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Barbara Rosati
- Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, 11794
| | - David Carlson
- Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, 11794
| | - Nai-Kong Cheung
- Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Hong Xu
- Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - James Bean
- Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Katherine Hsu
- Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Jean-Benoit Le Luduec
- Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Charles Kyriakos Vorkas
- Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, 11794
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, NY 11794
- Center for Infectious Diseases, Stony Brook University, Stony Brook, NY, 11794
| |
Collapse
|
7
|
Sutton MS, Bucsan AN, Lehman CC, Kamath M, Pokkali S, Magnani DM, Seder R, Darrah PA, Roederer M. Antibody-mediated depletion of select leukocyte subsets in blood and tissue of nonhuman primates. Front Immunol 2024; 15:1359679. [PMID: 38529287 PMCID: PMC10961357 DOI: 10.3389/fimmu.2024.1359679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 02/20/2024] [Indexed: 03/27/2024] Open
Abstract
Understanding the immunological control of pathogens requires a detailed evaluation of the mechanistic contributions of individual cell types within the immune system. While knockout mouse models that lack certain cell types have been used to help define the role of those cells, the biological and physiological characteristics of mice do not necessarily recapitulate that of a human. To overcome some of these differences, studies often look towards nonhuman primates (NHPs) due to their close phylogenetic relationship to humans. To evaluate the immunological role of select cell types, the NHP model provides distinct advantages since NHP more closely mirror the disease manifestations and immunological characteristics of humans. However, many of the experimental manipulations routinely used in mice (e.g., gene knock-out) cannot be used with the NHP model. As an alternative, the in vivo infusion of monoclonal antibodies that target surface proteins on specific cells to either functionally inhibit or deplete cells can be a useful tool. Such depleting antibodies have been used in NHP studies to address immunological mechanisms of action. In these studies, the extent of depletion has generally been reported for blood, but not thoroughly assessed in tissues. Here, we evaluated four depleting regimens that primarily target T cells in NHP: anti-CD4, anti-CD8α, anti-CD8β, and immunotoxin-conjugated anti-CD3. We evaluated these treatments in healthy unvaccinated and IV BCG-vaccinated NHP to measure the extent that vaccine-elicited T cells - which may be activated, increased in number, or resident in specific tissues - are depleted compared to resting populations in unvaccinated NHPs. We report quantitative measurements of in vivo depletion at multiple tissue sites providing insight into the range of cell types depleted by a given mAb. While we found substantial depletion of target cell types in blood and tissue of many animals, residual cells remained, often residing within tissue. Notably, we find that animal-to-animal variation is substantial and consequently studies that use these reagents should be powered accordingly.
Collapse
Affiliation(s)
- Matthew S. Sutton
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Allison N. Bucsan
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Chelsea C. Lehman
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Megha Kamath
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Supriya Pokkali
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Diogo M. Magnani
- Nonhuman Primate Reagent Resource, University of Massachusetts Chan Medical School, Worcester, MA, United States
| | - Robert Seder
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Patricia A. Darrah
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Mario Roederer
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States
| |
Collapse
|
8
|
Sutton MS, Bucsan AN, Lehman CC, Kamath M, Pokkali S, Magnani DM, Seder R, Darrah PA, Roederer M. Antibody-mediated depletion of select T cell subsets in blood and tissue of nonhuman primates. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.22.572898. [PMID: 38187627 PMCID: PMC10769432 DOI: 10.1101/2023.12.22.572898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Understanding the immunological control of pathogens requires a detailed evaluation of the mechanistic contributions of individual cell types within the immune system. While knockout mouse models that lack certain cell types have been used to help define the role of those cells, the biological and physiological characteristics of mice do not necessarily recapitulate that of a human. To overcome some of these differences, studies often look towards nonhuman primates (NHPs) due to their close phylogenetic relationship to humans. To evaluate the immunological role of select cell types, the NHP model provides distinct advantages since NHP more closely mirror the disease manifestations and immunological characteristics of humans. However, many of the experimental manipulations routinely used in mice (e.g., gene knock-out) cannot be used with the NHP model. As an alternative, the in vivo infusion of monoclonal antibodies that target surface proteins on specific cells to either functionally inhibit or deplete cells can be a useful tool. Such depleting antibodies have been used in NHP studies to address immunological mechanisms of action. In these studies, the extent of depletion has generally been reported for blood, but not thoroughly assessed in tissues. Here, we evaluated four depleting regimens that primarily target T cells in NHP: anti-CD4, anti-CD8α, anti-CD8β, and immunotoxin-conjugated anti-CD3. We evaluated these treatments in healthy unvaccinated and IV BCG-vaccinated NHP to measure the extent that vaccine-elicited T cells - which may be activated, increased in number, or resident in specific tissues - are depleted compared to resting populations in unvaccinated NHPs. We report quantitative measurements of in vivo depletion at multiple tissue sites providing insight into the range of cell types depleted by a given mAb. While we found substantial depletion of target cell types in blood and tissue of many animals, residual cells remained, often residing within tissue. Notably, we find that animal-to-animal variation is substantial and consequently studies that use these reagents should be powered accordingly.
Collapse
|
9
|
Thaller AL, Jönsson F, Fiquet O, Marie S, Doisne JM, Girelli-Zubani G, Eri T, Fernandes P, Tatirovsky E, Langa-Vives F, Bruhns P, Strick-Marchand H, Di Santo JP. A human immune system (HIS) mouse model that dissociates roles for mouse and human FcR + cells during antibody-mediated immune responses. Eur J Immunol 2023; 53:e2350454. [PMID: 37621208 DOI: 10.1002/eji.202350454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 07/21/2023] [Accepted: 08/21/2023] [Indexed: 08/26/2023]
Abstract
Human immune system (HIS) mice provide a model to study human immune responses in vivo. Currently available HIS mouse models may harbor mouse Fc Receptor (FcR)-expressing cells that exert potent effector functions following administration of human Ig. Previous studies showed that the ablation of the murine FcR gamma chain (FcR-γ) results in loss of antibody-dependent cellular cytotoxicity and antibody-dependent cellular phagocytosis in vivo. We created a new FcR-γ-deficient HIS mouse model to compare host (mouse) versus graft (human) effects underlying antibody-mediated immune responses in vivo. FcR-γ-deficient HIS recipients lack expression and function of mouse activating FcRs and can be stably and robustly reconstituted with human immune cells. By screening blood B-cell depletion by rituximab Ig variants, we found that human FcγRs-mediated IgG1 effects, whereas mouse activating FcγRs were dominant in IgG4 effects. Complement played a role as an IgG1 variant (IgG1 K322A) lacking complement binding activity was largely ineffective. Finally, we provide evidence that FcγRIIIA on human NK cells could mediate complement-independent B-cell depletion by IgG1 K322A. We anticipate that our FcR-γ-deficient HIS model will help clarify mechanisms of action of exogenous administered human antibodies in vivo.
Collapse
Affiliation(s)
- Anna Louisa Thaller
- Institut Pasteur, Innate Immunity Unit, Université Paris Cité, Inserm U1223, Paris, France
| | - Friederike Jönsson
- Institut Pasteur, Antibodies in Therapy and Pathology Unit, Université Paris Cité, Inserm U1222, Paris, France
| | - Oriane Fiquet
- Institut Pasteur, Innate Immunity Unit, Université Paris Cité, Inserm U1223, Paris, France
| | - Solenne Marie
- Institut Pasteur, Innate Immunity Unit, Université Paris Cité, Inserm U1223, Paris, France
| | - Jean-Marc Doisne
- Institut Pasteur, Innate Immunity Unit, Université Paris Cité, Inserm U1223, Paris, France
| | - Giulia Girelli-Zubani
- Institut Pasteur, Innate Immunity Unit, Université Paris Cité, Inserm U1223, Paris, France
| | - Toshiki Eri
- Institut Pasteur, Innate Immunity Unit, Université Paris Cité, Inserm U1223, Paris, France
| | - Priyanka Fernandes
- Institut Pasteur, Innate Immunity Unit, Université Paris Cité, Inserm U1223, Paris, France
| | - Evgeny Tatirovsky
- Institut Pasteur, Innate Immunity Unit, Université Paris Cité, Inserm U1223, Paris, France
| | - Francina Langa-Vives
- Institut Pasteur, Mouse Genetics Engineering Platform, Université Paris Cité, Paris, France
| | - Pierre Bruhns
- Institut Pasteur, Antibodies in Therapy and Pathology Unit, Université Paris Cité, Inserm U1222, Paris, France
| | - Hélène Strick-Marchand
- Institut Pasteur, Innate Immunity Unit, Université Paris Cité, Inserm U1223, Paris, France
| | - James P Di Santo
- Institut Pasteur, Innate Immunity Unit, Université Paris Cité, Inserm U1223, Paris, France
| |
Collapse
|
10
|
Gätjen D, Wieczorek M, Listek M, Tomszak F, Nölle V, Hanack K, Droste M. A switchable secrete-and-capture system enables efficient selection of Pichia pastoris clones producing high yields of Fab fragments. J Immunol Methods 2022; 511:113383. [PMID: 36356896 DOI: 10.1016/j.jim.2022.113383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 11/03/2022] [Accepted: 11/03/2022] [Indexed: 11/09/2022]
Abstract
Pichia pastoris (syn. Komagataella phaffii) represents a commonly used expression system in the biotech industry. High clonal variation of transformants, however, typically results in a broad range of specific productivities for secreted proteins. To isolate rare clones with exceedingly high product titers, an extensive number of clones need to be screened. In contrast to high-throughput screenings of P. pastoris clones in microtiter plates, secrete-and-capture methodologies have the potential to efficiently isolate high-producer clones among millions of cells through fluorescence-activated cell sorting (FACS). Here, we describe a novel approach for the non-covalent binding of fragment antigen-binding (Fab) proteins to the cell surface for the isolation of high-producing clones. Eight different single-chain variable fragment (scFv)-based capture matrices specific for the constant part of the Fabs were fused to the Saccharomyces cerevisiae alpha-agglutinin (SAG1) anchor protein for surface display in P. pastoris. By encoding the capture matrix on an episomal plasmid harboring inherently unstable autonomously replicating sequences (ARS), this secrete-and-capture system offers a switchable scFv display. Efficient plasmid clearance upon removal of selective pressure enabled the direct use of isolated clones for subsequent Fab production. Flow-sorted clones (n = 276) displaying high amounts of Fabs showed a significant increase in median Fab titers detected in the cell-free supernatant (CFS) compared to unsorted clones (n = 276) when cells were cultivated in microtiter plates (factor in the range of ∼21-49). Fab titers of clones exhibiting the highest product titer observed for each of the two approaches were increased by up to 8-fold for the sorted clone. Improved Fab yields of sorted cells vs. unsorted cells were confirmed in an upscaled shake flask cultivation of selected candidates (factor in the range of ∼2-3). Hence, the developed display-based selection method proved to be a valuable tool for efficient clone screening in the early stages of our bioprocess development.
Collapse
Affiliation(s)
- Dominic Gätjen
- Miltenyi Biotec B.V. & Co. KG, Friedrich-Ebert-Straße 68, 51429 Bergisch Gladbach, Germany; Institute of Biochemistry and Biology, University of Potsdam, Karl-Liebknecht-Straße 24-25, 14476 Potsdam, Germany
| | - Marek Wieczorek
- Miltenyi Biotec B.V. & Co. KG, Friedrich-Ebert-Straße 68, 51429 Bergisch Gladbach, Germany
| | - Martin Listek
- Institute of Biochemistry and Biology, University of Potsdam, Karl-Liebknecht-Straße 24-25, 14476 Potsdam, Germany
| | - Florian Tomszak
- Miltenyi Biotec B.V. & Co. KG, Friedrich-Ebert-Straße 68, 51429 Bergisch Gladbach, Germany
| | - Volker Nölle
- Miltenyi Biotec B.V. & Co. KG, Friedrich-Ebert-Straße 68, 51429 Bergisch Gladbach, Germany
| | - Katja Hanack
- Institute of Biochemistry and Biology, University of Potsdam, Karl-Liebknecht-Straße 24-25, 14476 Potsdam, Germany
| | - Miriam Droste
- Miltenyi Biotec B.V. & Co. KG, Friedrich-Ebert-Straße 68, 51429 Bergisch Gladbach, Germany.
| |
Collapse
|
11
|
Esgalhado AJ, Reste-Ferreira D, Albino SE, Sousa A, Amaral AP, Martinho A, Oliveira IT, Verde I, Lourenço O, Fonseca AM, Cardoso EM, Arosa FA. CD45RA, CD8β, and IFNγ Are Potential Immune Biomarkers of Human Cognitive Function. Front Immunol 2020; 11:592656. [PMID: 33324408 PMCID: PMC7723833 DOI: 10.3389/fimmu.2020.592656] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 10/12/2020] [Indexed: 11/23/2022] Open
Abstract
There is increasing evidence that in humans the adaptive immunological system can influence cognitive functions of the brain. We have undertaken a comprehensive immunological analysis of lymphocyte and monocyte populations as well as of HLA molecules expression in a cohort of elderly volunteers (age range, 64–101) differing in their cognitive status. Hereby, we report on the identification of a novel signature in cognitively impaired elderly characterized by: (1) elevated percentages of CD8+ T effector-memory cells expressing high levels of the CD45RA phosphate receptor (Temrahi); (2) high percentages of CD8+ T cells expressing high levels of the CD8β chain (CD8βhi); (3) augmented production of IFNγ by in vitro activated CD4+ T cells. Noteworthy, CD3+CD8+ Temrahi and CD3+CD8βhi cells were associated with impaired cognition. Cytomegalovirus seroprevalence showed that all volunteers studied but one were CMV positive. Finally, we show that some of these phenotypic and functional features are associated with an increased frequency of the HLA-B8 serotype, which belongs to the ancestral haplotype HLA-A1, Cw7, B8, DR3, DQ2, among cognitively impaired volunteers. To our knowledge, this is the first proof in humans linking the amount of cell surface CD45RA and CD8β chain expressed by CD8+ Temra cells, and the amount of IFNγ produced by in vitro activated CD4+ T cells, with impaired cognitive function in the elderly.
Collapse
Affiliation(s)
- André J Esgalhado
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Débora Reste-Ferreira
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Stephanie E Albino
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Adriana Sousa
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Ana Paula Amaral
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - António Martinho
- Molecular Genetics Laboratory, Coimbra Blood and Transplantation Center, Coimbra, Portugal
| | - Isabel T Oliveira
- C4-UBI, Cloud Computing Competence Centre, University of Beira Interior, Covilhã, Portugal
| | - Ignacio Verde
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal.,Faculty of Health Sciences, University of Beira Interior, Covilhã, Portugal
| | - Olga Lourenço
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal.,Faculty of Health Sciences, University of Beira Interior, Covilhã, Portugal
| | - Ana M Fonseca
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal.,Faculty of Health Sciences, University of Beira Interior, Covilhã, Portugal
| | - Elsa M Cardoso
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal.,Faculty of Health Sciences, University of Beira Interior, Covilhã, Portugal.,IPG, Guarda Polytechnic Institute, Guarda, Portugal
| | - Fernando A Arosa
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal.,Faculty of Health Sciences, University of Beira Interior, Covilhã, Portugal
| |
Collapse
|
12
|
Goodall KJ, Nguyen A, McKenzie C, Eckle SBG, Sullivan LC, Andrews DM. The murine CD94/NKG2 ligand, Qa-1 b, is a high-affinity, functional ligand for the CD8αα homodimer. J Biol Chem 2020; 295:3239-3246. [PMID: 31992596 PMCID: PMC7062157 DOI: 10.1074/jbc.ra119.010509] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 01/22/2020] [Indexed: 11/06/2022] Open
Abstract
The immune co-receptor CD8 molecule (CD8) has two subunits, CD8α and CD8β, which can assemble into homo or heterodimers. Nonclassical (class-Ib) major histocompatibility complex (MHC) molecules (MHC-Ibs) have recently been identified as ligands for the CD8αα homodimer. This was demonstrated by the observation that histocompatibility 2, Q region locus 10 (H2-Q10) is a high-affinity ligand for CD8αα which also binds the MHC-Ib molecule H2-TL. This suggests that MHC-Ib proteins may be an extended source of CD8αα ligands. Expression of H2-T3/TL and H2-Q10 is restricted to the small intestine and liver, respectively, yet CD8αα-containing lymphocytes are present more broadly. Therefore, here we sought to determine whether murine CD8αα binds only to tissue-specific MHC-Ib molecules or also to ubiquitously expressed MHC-Ib molecules. Using recombinant proteins and surface plasmon resonance-based binding assays, we show that the MHC-Ib family furnishes multiple binding partners for murine CD8αα, including H2-T22 and the CD94/NKG2-A/B-activating NK receptor (NKG2) ligand Qa-1b We also demonstrate a hierarchy among MHC-Ib proteins with respect to CD8αα binding, in which Qa-1b > H2-Q10 > TL. Finally, we provide evidence that Qa-1b is a functional ligand for CD8αα, distinguishing it from its human homologue MHC class I antigen E (HLA-E). These findings provide additional clues as to how CD8αα-expressing cells are controlled in different tissues. They also highlight an unexpected immunological divergence of Qa-1b/HLA-E function, indicating the need for more robust studies of murine MHC-Ib proteins as models for human disease.
Collapse
Affiliation(s)
- Katharine Jennifer Goodall
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Victoria, 3004, Australia.
| | - Angela Nguyen
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Victoria, 3004, Australia
| | - Craig McKenzie
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Victoria, 3004, Australia
| | - Sidonia Barbara Guiomar Eckle
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, 3000, Australia
| | - Lucy Catherine Sullivan
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, 3000, Australia
| | - Daniel Mark Andrews
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Victoria, 3004, Australia
| |
Collapse
|
13
|
Abstract
Cluster of differentiation 8 (CD8) is a cell surface glycoprotein, which is expressed as 2 forms, αα homodimer or αβ heterodimer. Peptide-loaded major histocompatibility complex class I (pMHC-I) molecules are major ligands for both forms of CD8. CD8αβ is a coreceptor for the T cell receptor (TCR) and binds to the same cognate pMHC-I as the TCR, thus enabling or augmenting T cell responses. The function of CD8αα homodimers is largely unknown. While CD8αβ heterodimer is expressed exclusively on CD8+ T cells, the CD8αα homodimer is present in subsets of T cells and human natural killer (NK) cells. Here, we report that the CD8αα homodimer functions as a coreceptor for KIR3DL1, an inhibitory receptor of NK cells that is specific for certain MHC-I allotypes. CD8αα enhances binding of pMHC-I to KIR3DL1, increases KIR3DL1 clustering at the immunological synapse, and augments KIR3DL1-mediated inhibition of NK cell activation. Additionally, interactions between pMHC-I and CD8αα homodimers regulate KIR3DL1+ NK cell education. Together, these findings reveal another dimension to the modulation of NK cell activity.
Collapse
|
14
|
Sutton MS, Ellis-Connell A, Balgeman AJ, Barry G, Weiler AM, Hetzel SJ, Zhou Y, Lau-Kilby AW, Mason RD, Biris KK, Mascola JR, Sullivan NJ, Roederer M, Friedrich TC, O'Connor SL. CD8β Depletion Does Not Prevent Control of Viral Replication or Protection from Challenge in Macaques Chronically Infected with a Live Attenuated Simian Immunodeficiency Virus. J Virol 2019; 93:e00537-19. [PMID: 31092584 PMCID: PMC6639280 DOI: 10.1128/jvi.00537-19] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 05/11/2019] [Indexed: 11/20/2022] Open
Abstract
We evaluated the contribution of CD8αβ+ T cells to control of live-attenuated simian immunodeficiency virus (LASIV) replication during chronic infection and subsequent protection from pathogenic SIV challenge. Unlike previous reports with a CD8α-specific depleting monoclonal antibody (mAb), the CD8β-specific mAb CD8β255R1 selectively depleted CD8αβ+ T cells without also depleting non-CD8+ T cell populations that express CD8α, such as natural killer (NK) cells and γδ T cells. Following infusion with CD8β255R1, plasma viremia transiently increased coincident with declining peripheral CD8αβ+ T cells. Interestingly, plasma viremia returned to predepletion levels even when peripheral CD8αβ+ T cells did not. Although depletion of CD8αβ+ T cells in the lymph node (LN) was incomplete, frequencies of these cells were 3-fold lower (P = 0.006) in animals that received CD8β255R1 than in those that received control IgG. It is possible that these residual SIV-specific CD8αβ+ T cells may have contributed to suppression of viremia during chronic infection. We also determined whether infusion of CD8β255R1 in the LASIV-vaccinated animals increased their susceptibility to infection following intravenous challenge with pathogenic SIVmac239. We found that 7/8 animals infused with CD8β255R1, and 3/4 animals infused with the control IgG, were resistant to SIVmac239 infection. These results suggest that infusion with CD8β255R1 did not eliminate the protection afforded to LASIV vaccination. This provides a comprehensive description of the impact of CD8β255R1 infusion on the immunological composition in cynomolgus macaques, compared to an isotype-matched control IgG, while showing that the control of LASIV viremia and protection from challenge can occur even after CD8β255R1 administration.IMPORTANCE Studies of SIV-infected macaques that deplete CD8+ T cells in vivo with monoclonal antibodies have provided compelling evidence for their direct antiviral role. These studies utilized CD8α-specific mAbs that target both the major (CD8αβ+) and minor (CD8αα+) populations of CD8+ T cells but additionally deplete non-CD8+ T cell populations that express CD8α, such as NK cells and γδ T cells. In the current study, we administered the CD8β-specific depleting mAb CD8β255R1 to cynomolgus macaques chronically infected with a LASIV to selectively deplete CD8αβ+ T cells without removing CD8αα+ lymphocytes. We evaluated the impact on control of virus replication and protection from pathogenic SIVmac239 challenge. These results underscore the utility of CD8β255R1 for studying the direct contribution of CD8αβ+ T cells in various disease states.
Collapse
Affiliation(s)
- Matthew S Sutton
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Amy Ellis-Connell
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Alexis J Balgeman
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Gabrielle Barry
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Andrea M Weiler
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Scott J Hetzel
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Yan Zhou
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Annie W Lau-Kilby
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Rosemarie D Mason
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Kristin K Biris
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - John R Mascola
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Nancy J Sullivan
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Mario Roederer
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Thomas C Friedrich
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Shelby L O'Connor
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
15
|
Piranlioglu R, Lee E, Ouzounova M, Bollag RJ, Vinyard AH, Arbab AS, Marasco D, Guzel M, Cowell JK, Thangaraju M, Chadli A, Hassan KA, Wicha MS, Celis E, Korkaya H. Primary tumor-induced immunity eradicates disseminated tumor cells in syngeneic mouse model. Nat Commun 2019; 10:1430. [PMID: 30926774 PMCID: PMC6441000 DOI: 10.1038/s41467-019-09015-1] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 02/14/2019] [Indexed: 02/07/2023] Open
Abstract
Although clinically apparent metastasis is associated with late stages of cancer development, micro-metastatic dissemination may be an early event. However, the fate of these early disseminated tumor cells (DTC) remains elusive. We show that despite their capacity to disseminate into secondary organs, 4T1 tumor models develop overt metastasis while EMT6-tumor bearing mice clear DTCs shed from primary tumors as well as those introduced by intravenous (IV) injection. Following the surgical resection of primary EMT6 tumors, mice do not develop detectable metastasis and reject IV-injected tumor cells. In contrast, these cells readily grow and metastasize in immuno-deficient athymic or Rag2−/− mice, an effect mimicked by CD8+ T-cell depletion in immunocompetent mice. Furthermore, recombinant G-CSF or adoptive transfer of granulocytic-MDSCs isolated from 4T1 tumor-bearing mice, induce metastasis by suppressing CD8+ T-cells in EMT6-primed mice. Our studies support the concept of immune surveillance providing molecular insights into the immune mechanisms during tumor progression. Dissemination of tumor cells from the primary site is an early event. Here, the authors show that the early disseminated tumor cells are actively cleared by the host cytotoxic T lymphocytes induced by the primary tumor and that infiltration of granulocytic myeloid-derived suppressor cells counteracts such immune protection and allow metastasis development.
Collapse
Affiliation(s)
- Raziye Piranlioglu
- Georgia Cancer Center, Department of Biochemistry and Molecular Biology, Augusta University, 1410 Laney Walker Blvd. CN2136, Augusta, GA, 30912, USA
| | - EunMi Lee
- Georgia Cancer Center, Department of Biochemistry and Molecular Biology, Augusta University, 1410 Laney Walker Blvd. CN2136, Augusta, GA, 30912, USA
| | - Maria Ouzounova
- Cancer Research Center of Lyon, 28 Rue Laennec, 69008, Lyon, France
| | - Roni J Bollag
- Georgia Cancer Center, Department of Biochemistry and Molecular Biology, Augusta University, 1410 Laney Walker Blvd. CN2136, Augusta, GA, 30912, USA
| | - Alicia H Vinyard
- Georgia Cancer Center, Department of Biochemistry and Molecular Biology, Augusta University, 1410 Laney Walker Blvd. CN2136, Augusta, GA, 30912, USA
| | - Ali S Arbab
- Georgia Cancer Center, Department of Biochemistry and Molecular Biology, Augusta University, 1410 Laney Walker Blvd. CN2136, Augusta, GA, 30912, USA
| | - Daniela Marasco
- Department of Pharmacy, University of Naples "Federico II", 80134, Naples, Italy
| | - Mustafa Guzel
- Regenerative and Restorative Research Center (REMER), Medipol University, Kavacık Mah. Ekinciler Cad. No.19 Kavacık Kavşağı - Beykoz, 34810, İstanbul Istanbul, Turkey
| | - John K Cowell
- Georgia Cancer Center, Department of Biochemistry and Molecular Biology, Augusta University, 1410 Laney Walker Blvd. CN2136, Augusta, GA, 30912, USA
| | - Muthushamy Thangaraju
- Georgia Cancer Center, Department of Biochemistry and Molecular Biology, Augusta University, 1410 Laney Walker Blvd. CN2136, Augusta, GA, 30912, USA
| | - Ahmed Chadli
- Georgia Cancer Center, Department of Biochemistry and Molecular Biology, Augusta University, 1410 Laney Walker Blvd. CN2136, Augusta, GA, 30912, USA
| | - Khaled A Hassan
- Comprehensive Cancer Center, University of Michigan, 1500 E. Medical Center Dr, Ann Arbor, MI, 48109, USA
| | - Max S Wicha
- Comprehensive Cancer Center, University of Michigan, 1500 E. Medical Center Dr, Ann Arbor, MI, 48109, USA
| | - Esteban Celis
- Georgia Cancer Center, Department of Biochemistry and Molecular Biology, Augusta University, 1410 Laney Walker Blvd. CN2136, Augusta, GA, 30912, USA
| | - Hasan Korkaya
- Georgia Cancer Center, Department of Biochemistry and Molecular Biology, Augusta University, 1410 Laney Walker Blvd. CN2136, Augusta, GA, 30912, USA.
| |
Collapse
|
16
|
Transcriptome analysis of immune genes in peripheral blood mononuclear cells of young foals and adult horses. PLoS One 2018; 13:e0202646. [PMID: 30183726 PMCID: PMC6124769 DOI: 10.1371/journal.pone.0202646] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 08/07/2018] [Indexed: 12/14/2022] Open
Abstract
During the neonatal period, the ability to generate immune effector and memory responses to vaccines or pathogens is often questioned. This study was undertaken to obtain a global view of the natural differences in the expression of immune genes early in life. Our hypothesis was that transcriptome analyses of peripheral blood mononuclear cells (PBMCs) of foals (on day 1 and day 42 after birth) and adult horses would show differential gene expression profiles that characterize natural immune processes. Gene ontology enrichment analysis provided assessment of biological processes affected by age, and a list of 897 genes with ≥2 fold higher (p<0.01) expression in day 42 when compared to day 1 foal samples. Up-regulated genes included B cell and T cell receptor diversity genes; DNA replication enzymes; natural killer cell receptors; granzyme B and perforin; complement receptors; immunomodulatory receptors; cell adhesion molecules; and cytokines/chemokines and their receptors. The list of 1,383 genes that had higher (p<0.01) expression on day 1 when compared to day 42 foal samples was populated by genes with roles in innate immunity such as antimicrobial proteins; pathogen recognition receptors; cytokines/chemokines and their receptors; cell adhesion molecules; co-stimulatory molecules; and T cell receptor delta chain. Within the 742 genes with increased expression between day 42 foal and adult samples, B cell immunity was the main biological process (p = 2.4E-04). Novel data on markedly low (p<0.0001) TLR3 gene expression, and high (p≤0.01) expression of IL27, IL13RA1, IREM-1, SIRL-1, and SIRPα on day 1 compared to day 42 foal samples point out potential mechanisms of increased susceptibility to pathogens in early life. The results portray a progression from innate immune gene expression predominance early in life to adaptive immune gene expression increasing with age with a putative overlay of immune suppressing genes in the neonatal phase. These results provide insight to the unique attributes of the equine neonatal and young immune system, and offer many avenues of future investigation.
Collapse
|
17
|
The composition of T cell subtypes in duodenal biopsies are altered in coeliac disease patients. PLoS One 2017; 12:e0170270. [PMID: 28166225 PMCID: PMC5293270 DOI: 10.1371/journal.pone.0170270] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 01/02/2017] [Indexed: 12/11/2022] Open
Abstract
One of the hallmarks of Celiac disease (CD) is intraepithelial lymphocytosis in the small intestine. Until now, investigations to characterize the T cell subpopulations within the epithelial layer have not discriminated between the heterodimeric co-receptor molecule, CD8αβ, and the possibly immunoregulatory CD8αα homodimer molecule. Besides TCRαβ+ CD4+ cells, no other phenotypes have been shown to be gluten-reactive. Using flow cytometry on lymphocytes from duodenal biopsies, we determined that the number of B cells (CD3- CD19+) and the number of CD3+ CD4- CD8- double-negative (DN) T cells were elevated 6–7 fold in children with CD. We next isolated and quantified intraepithelial lymphocytes (IELs) from biopsies obtained from patients (both children and adults) with CD, potential CD and non-CD controls. Flow cytometric analysis of the duodenal T cell subpopulations was performed including the markers TCRαβ, TCRγδ, CD4, CD8α and CD8β. Proportions of γδ T cells and CD8αβ+ cells among IELs were increased in CD patients, whereas proportions of CD4+ CD8αα+ and CD4+ single-positive T cells were decreased. Additionally, two gluten-reactive T cell lines (TCLs) derived from CD biopsies were analyzed for changes in proportions of T cell subsets before and after gluten stimulation. In a proliferation assay, dividing cells were tracked with carboxyfluorescein succinimidyl ester (CFSE), and both αβ and γδ T cells proliferated in response to gluten. Changes in duodenal T cell subpopulations in potential CD patients followed the same pattern as for CD patients, but with less pronounced effect.
Collapse
|
18
|
Mostafa H, Pala A, Högel J, Hlavac M, Dietrich E, Westhoff MA, Nonnenmacher L, Burster T, Georgieff M, Wirtz CR, Schneider EM. Immune phenotypes predict survival in patients with glioblastoma multiforme. J Hematol Oncol 2016; 9:77. [PMID: 27585656 PMCID: PMC5009501 DOI: 10.1186/s13045-016-0272-3] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 04/13/2016] [Indexed: 11/24/2022] Open
Abstract
Background Glioblastoma multiforme (GBM), a common primary malignant brain tumor, rarely disseminates beyond the central nervous system and has a very bad prognosis. The current study aimed at the analysis of immunological control in individual patients with GBM. Methods Immune phenotypes and plasma biomarkers of GBM patients were determined at the time of diagnosis using flow cytometry and ELISA, respectively. Results Using descriptive statistics, we found that immune anomalies were distinct in individual patients. Defined marker profiles proved highly relevant for survival. A remarkable relation between activated NK cells and improved survival in GBM patients was in contrast to increased CD39 and IL-10 in patients with a detrimental course and very short survival. Recursive partitioning analysis (RPA) and Cox proportional hazards models substantiated the relevance of absolute numbers of CD8 cells and low numbers of CD39 cells for better survival. Conclusions Defined alterations of the immune system may guide the course of disease in patients with GBM and may be prognostically valuable for longitudinal studies or can be applied for immune intervention. Electronic supplementary material The online version of this article (doi:10.1186/s13045-016-0272-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Haouraa Mostafa
- Sektion Experimentelle Anaesthesiologie, University Hospital Ulm, Albert Einstein Allee 23, 89081, Ulm, Germany.,Klinik für Anaesthesiologie, University Hospital Ulm, Albert Einstein Allee 23, 89081, Ulm, Germany
| | - Andrej Pala
- Department of Neurosurgery, University Hospital Ulm Albert Einstein Allee 23, 89081 Ulm and Bezirkskrankenhaus Günzburg, Ludwig-Heilmeyer-Str. 2, 89312, Günzburg, Germany
| | - Josef Högel
- Institute for Human Genetics, Albert Einstein Allee 11, 89081, Ulm, Germany
| | - Michal Hlavac
- Department of Neurosurgery, University Hospital Ulm Albert Einstein Allee 23, 89081 Ulm and Bezirkskrankenhaus Günzburg, Ludwig-Heilmeyer-Str. 2, 89312, Günzburg, Germany
| | - Elvira Dietrich
- Sektion Experimentelle Anaesthesiologie, University Hospital Ulm, Albert Einstein Allee 23, 89081, Ulm, Germany.,Klinik für Anaesthesiologie, University Hospital Ulm, Albert Einstein Allee 23, 89081, Ulm, Germany
| | - M Andrew Westhoff
- Department of Pediatric Hematology and Oncology, University Hospital Ulm, Prittwitzstr. 43, 89075, Ulm, Germany
| | - Lisa Nonnenmacher
- Department of Neurosurgery, University Hospital Ulm Albert Einstein Allee 23, 89081 Ulm and Bezirkskrankenhaus Günzburg, Ludwig-Heilmeyer-Str. 2, 89312, Günzburg, Germany
| | - Timo Burster
- Department of Neurosurgery, University Hospital Ulm Albert Einstein Allee 23, 89081 Ulm and Bezirkskrankenhaus Günzburg, Ludwig-Heilmeyer-Str. 2, 89312, Günzburg, Germany
| | - Michael Georgieff
- Klinik für Anaesthesiologie, University Hospital Ulm, Albert Einstein Allee 23, 89081, Ulm, Germany
| | - C Rainer Wirtz
- Department of Neurosurgery, University Hospital Ulm Albert Einstein Allee 23, 89081 Ulm and Bezirkskrankenhaus Günzburg, Ludwig-Heilmeyer-Str. 2, 89312, Günzburg, Germany
| | - E Marion Schneider
- Sektion Experimentelle Anaesthesiologie, University Hospital Ulm, Albert Einstein Allee 23, 89081, Ulm, Germany. .,Klinik für Anaesthesiologie, University Hospital Ulm, Albert Einstein Allee 23, 89081, Ulm, Germany.
| |
Collapse
|
19
|
Mikalsen AB, Haugland O, Rode M, Solbakk IT, Evensen O. Atlantic salmon reovirus infection causes a CD8 T cell myocarditis in Atlantic salmon (Salmo salar L.). PLoS One 2012; 7:e37269. [PMID: 22693625 PMCID: PMC3367920 DOI: 10.1371/journal.pone.0037269] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2012] [Accepted: 04/17/2012] [Indexed: 12/22/2022] Open
Abstract
Heart and skeletal inflammation (HSMI) of farmed Atlantic salmon (Salmo salar L.) is a disease characterized by a chronic myocarditis involving the epicardium and the compact and spongious part of the heart ventricle. Chronic myositis of the red skeletal muscle is also a typical finding of HSMI. Piscine reovirus (PRV) has been detected by real-time PCR from farmed and wild salmon with and without typical changes of HSMI and thus the causal relationship between presence of virus and the disease has not been fully determined. In this study we show that the Atlantic salmon reovirus (ASRV), identical to PRV, can be passaged in GF-1 cells and experimental challenge of naïve Atlantic salmon with cell culture passaged reovirus results in cardiac and skeletal muscle pathology typical of HSMI with onset of pathology from 6 weeks, peaking by 9 weeks post challenge. ASRV replicates in heart tissue and the peak level of virus replication coincides with peak of heart lesions. We further demonstrate mRNA transcript assessment and in situ characterization that challenged fish develop a CD8+ T cell myocarditis.
Collapse
Affiliation(s)
- Aase B. Mikalsen
- Department of Basic Sciences and Aquatic Medicine, Norwegian School of Veterinary Science, Oslo, Norway
| | - Oyvind Haugland
- Department of Basic Sciences and Aquatic Medicine, Norwegian School of Veterinary Science, Oslo, Norway
| | | | | | - Oystein Evensen
- Department of Basic Sciences and Aquatic Medicine, Norwegian School of Veterinary Science, Oslo, Norway
- * E-mail:
| |
Collapse
|
20
|
Laugel B, Cole DK, Clement M, Wooldridge L, Price DA, Sewell AK. The multiple roles of the CD8 coreceptor in T cell biology: opportunities for the selective modulation of self-reactive cytotoxic T cells. J Leukoc Biol 2011; 90:1089-99. [PMID: 21954283 DOI: 10.1189/jlb.0611316] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Short peptide fragments generated by intracellular protein cleavage are presented on the surface of most nucleated cells bound to highly polymorphic MHCI molecules. These pMHCI complexes constitute an interface that allows the immune system to identify and eradicate anomalous cells, such as those that harbor infectious agents, through the activation of CTLs. Molecular recognition of pMHCI complexes is mediated primarily by clonally distributed TCRs expressed on the surface of CTLs. The coreceptor CD8 contributes to this antigen-recognition process by binding to a largely invariant region of the MHCI molecule and by promoting intracellular signaling, the effects of which serve to enhance TCR stimuli triggered by cognate ligands. Recent investigations have shed light on the role of CD8 in the activation of MHCI-restricted, antigen-experienced T cells and in the processes of T cell selection and lineage commitment in the thymus. Here, we review these data and discuss their implications for the development of potential therapeutic strategies that selectively target pathogenic CTL responses erroneously directed against self-derived antigens.
Collapse
Affiliation(s)
- Bruno Laugel
- School of Medicine, Cardiff University, Heath Park, Cardiff, CF14 4XN Wales, UK.
| | | | | | | | | | | |
Collapse
|
21
|
Wang R, Natarajan K, Margulies DH. Structural basis of the CD8 alpha beta/MHC class I interaction: focused recognition orients CD8 beta to a T cell proximal position. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2009; 183:2554-64. [PMID: 19625641 PMCID: PMC2782705 DOI: 10.4049/jimmunol.0901276] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
In the immune system, B cells, dendritic cells, NK cells, and T lymphocytes all respond to signals received via ligand binding to receptors and coreceptors. Although the specificity of T cell recognition is determined by the interaction of T cell receptors with MHC/peptide complexes, the development of T cells in the thymus and their sensitivity to Ag are also dependent on coreceptor molecules CD8 (for MHC class I (MHCI)) and CD4 (for MHCII). The CD8alphabeta heterodimer is a potent coreceptor for T cell activation, but efforts to understand its function fully have been hampered by ignorance of the structural details of its interactions with MHCI. In this study we describe the structure of CD8alphabeta in complex with the murine MHCI molecule H-2D(d) at 2.6 A resolution. The focus of the CD8alphabeta interaction is the acidic loop (residues 222-228) of the alpha3 domain of H-2D(d). The beta subunit occupies a T cell membrane proximal position, defining the relative positions of the CD8alpha and CD8beta subunits. Unlike the CD8alphaalpha homodimer, CD8alphabeta does not contact the MHCI alpha(2)- or beta(2)-microglobulin domains. Movements of the CD8alpha CDR2 and CD8beta CDR1 and CDR2 loops as well as the flexibility of the H-2D(d) CD loop facilitate the monovalent interaction. The structure resolves inconclusive data on the topology of the CD8alphabeta/MHCI interaction, indicates that CD8beta is crucial in orienting the CD8alphabeta heterodimer, provides a framework for understanding the mechanistic role of CD8alphabeta in lymphoid cell signaling, and offers a tangible context for design of structurally altered coreceptors for tumor and viral immunotherapy.
Collapse
Affiliation(s)
- Rui Wang
- Molecular Biology Section, Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892-1892
| | - Kannan Natarajan
- Molecular Biology Section, Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892-1892,Address correspondence and reprint requests to Dr. Kannan Natarajan, or Dr. David H. Margulies, Molecular Biology Section, Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bldg. 10, Room 11N311; 10 Center Drive, Bethesda, MD 20892-1892. and
| | - David H. Margulies
- Molecular Biology Section, Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892-1892,Address correspondence and reprint requests to Dr. Kannan Natarajan, or Dr. David H. Margulies, Molecular Biology Section, Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bldg. 10, Room 11N311; 10 Center Drive, Bethesda, MD 20892-1892. and
| |
Collapse
|
22
|
Clarke RL, Thiemann S, Refaeli Y, Werlen G, Potter TA. A new function for LAT and CD8 during CD8-mediated apoptosis that is independent of TCR signal transduction. Eur J Immunol 2009; 39:1619-31. [PMID: 19449311 DOI: 10.1002/eji.200839062] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The majority (>95%) of thymocytes undergo apoptosis during selection in the thymus. Several mechanisms have been proposed to explain how apoptosis of thymocytes that are not positively selected occurs; however, it is unknown whether thymocytes die purely by "neglect" or whether signaling through a cell-surface receptor initiates an apoptotic pathway. We have previously demonstrated that on double positive thymocytes the ligation of CD8 in the absence of TCR engagement results in apoptosis and have postulated this is a mechanism to remove thymocytes that have failed positive selection. On mature single positive T cells CD8 acts as a co-receptor to augment signaling through the TCR that is dependent on the phosphorylation of the adaptor protein, linker for activation of T cells (LAT). Here, we show that during CD8-mediated apoptosis of double positive thymocytes there is an increase in the association of CD8 with LAT and an increase in LAT tyrosine phosphorylation. Decreasing LAT expression and mutation of tyrosine residues of LAT reduced apoptosis upon crosslinking of CD8. Our results identify novel functions for both CD8 and LAT that are independent of TCR signal transduction and suggest a mechanism for signal transduction leading to apoptosis upon CD8 crosslinking.
Collapse
Affiliation(s)
- Raedun L Clarke
- Integrated Department of Immunology, National Jewish Health, Denver, CO 80206, USA.
| | | | | | | | | |
Collapse
|
23
|
Yagihara H, Uematsu Y, Koike A, Tamura K, Isotani M, Yamaguchi T, Ono K, Washizu T, Bonkobara M. Immunophenotyping and gene rearrangement analysis in dogs with lymphoproliferative disorders characterized by small-cell lymphocytosis. J Vet Diagn Invest 2009; 21:197-202. [PMID: 19286497 DOI: 10.1177/104063870902100203] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Lymphocytosis caused by neoplastic proliferation of small lymphocytes is occasionally difficult to distinguish by morphological examination from nonneoplastic lymphocytosis. To examine the clinical utility of gene rearrangement analysis for demonstrating neoplastic proliferation of small lymphocytes, gene rearrangement analysis was performed in comparison with immunophenotyping using peripheral lymphocytes in dogs with small lymphocytosis. Thirty-one dogs with small-cell lymphocytosis (8,100-884,300/microl) were enrolled. By immunophenotyping, lymphocytosis of all dogs was suggested to be neoplastic in nature based on the detection of marked expansion of phenotypically homogeneous lymphocytes or the presence of an aberrant antigen-expressing population of lymphocytes. In contrast, gene rearrangement analysis represented clonality in 27 dogs (detection rate of 87%). From the present study, gene rearrangement analysis was considered to be worthwhile to strengthen the evidence of neoplastic proliferation of small lymphocytes when coupled with immunophenotyping and to be a suitable diagnostic substitute if immunophenotyping is not available in clinical practice.
Collapse
Affiliation(s)
- Hiroko Yagihara
- Department of Veterinary Clinical Pathology, Nippon Veterinary and Life Science University, Tokyo, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Werwitzke S, Tiede A, Jacobs R, Zielinska-Skowronek M, Buyny S, Schmidt RE, Witte T. CD8alpha+beta(low) effector T cells in systemic lupus erythematosus. Scand J Immunol 2008; 67:501-8. [PMID: 18405327 DOI: 10.1111/j.1365-3083.2008.02093.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Systemic lupus erythematosus (SLE) is a complex autoimmune disorder characterized by the loss of self-tolerance to nuclear antigens. Aberrant T-cell function plays a central role in lupus pathogenesis. We and others previously demonstrated that peripheral TCRalphabeta+CD3+ T cells express CD8beta either at a high (CD8beta(high)) or low density (CD8beta(low)), thereby defining two functionally distinct subsets. CD8beta(low) T cells express predominantly CD8alphaalpha and less CD8alphabeta as a coreceptor, display a differentiated phenotype and exert effector function. CD8beta(high) T cells appear to be the precursors expressing predominantly the heterodimeric efficient CD8alphabeta coreceptor, exhibiting a naïve phenotype and high proliferative capacity. In the present study, the distribution and functional properties of CD8beta(high) and CD8beta(low) T cells of SLE patients were compared (n = 20) with those of healthy subjects (n = 16). It was found that expansion of CD8beta(low) T-cell subset correlated with disease activity indicating chronic antigenic stimulation leading to a major lack of naïve CD8beta(high) precursor T cells in SLE. Functional characteristics of CD8beta(low) T cells including production of cytokines and cytotoxic granules were not significantly different between patients with SLE and healthy individuals. We speculate that unbalanced CD8beta(high)/CD8beta(low) T-cell relation reflects a skewed homeostasis within the CD8+ T-cell compartment towards fully differentiated effector T cells possibly due to persistent antigen stimulation in SLE.
Collapse
Affiliation(s)
- S Werwitzke
- Clinic for Immunology and Rheumatology, Hannover Medical School, Hannover, Germany
| | | | | | | | | | | | | |
Collapse
|
25
|
Abstract
"The beginning of wisdom is found in doubting; by doubting we come to question, and by seeking we may come upon the truth." -Pierre Abélard. CD8 is a glycoprotein expressed on hematopoietic cells. Two isoforms of CD8, CD8alphabeta and CD8alphaalpha, have been identified that are distinct in their expression and function. Whereas CD8alphabeta serves as a T cell receptor (TCR) coreceptor to enhance the functional avidity and is constitutively expressed on MHC class I-restricted T cells, CD8alphaalpha marks T cells that are distinct from the conventional thymus-selected and MHC-restricted CD4(+) or CD8alphabeta(+) T cells. Inconsistent with a coreceptor function, CD8alphaalpha decreases antigen sensitivity of the TCR, and it can be transiently or permanently expressed on T cells, regardless of the MHC restriction of the TCR or the presence of conventional coreceptors. Together, these observations indicate that CD8alphaalpha on T cells marks a differentiation stage and that it likely functions as a TCR corepressor to negatively regulate T cell activation.
Collapse
|
26
|
Boulassel MR, Mercier F, Gilmore N, Routy JP. Immunophenotypic patterns of CD8+ T cell subsets expressing CD8alphaalpha and IL-7Ralpha in viremic, aviremic and slow progressor HIV-1-infected subjects. Clin Immunol 2007; 124:149-57. [PMID: 17560832 DOI: 10.1016/j.clim.2007.05.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2006] [Revised: 05/07/2007] [Accepted: 05/08/2007] [Indexed: 10/23/2022]
Abstract
Evidence from animal models suggests that the expression of CD8alphaalpha homodimer on CD8(+) T cells plays a key role in the generation of long-lived memory cells. Here, we studied the quantitative alterations of CD8(+) T cell subsets expressing CD8alphaalpha, interleukin-7 receptor (IL-7Ralpha) and activation markers in HIV-1-infected individuals including aviremic, viremic and slow progressor subjects using eight-color flow cytometry. Compared to slow progressor subjects, expression of CD8alphaalpha was significantly reduced in aviremic and viremic patients and this reduction occurred mainly within central memory cell subsets and not in naive and effector memory compartments. Persistence of antigenemia leads to IL-7Ralpha loss mainly on central and pre-terminal memory CD8(+) T cell subsets in viremic patients but not in slow progressor subjects. Compared to aviremic and viremic patients, slow progressor subjects had lower levels of IL-7 and reduced activated cells. The expression of CD8alphaalpha was not significantly related to IL-7Ralpha although negative associations were evidenced within all CD8(+) T cell subsets. Collectively, these results further advance the characterization of immunophenotypic patterns of CD8(+) T cell subsets expressing CD8alphaalpha/IL-7Ralpha and provide new insights into the ability of HIV-1 infection to alter memory cell population.
Collapse
Affiliation(s)
- Mohamed-Rachid Boulassel
- Immunodeficiency Service, Montreal Chest Institute, McGill University Health Centre, Montreal, Quebec, Canada
| | | | | | | |
Collapse
|
27
|
Chang HC, Tan K, Hsu YM. CD8αβ Has Two Distinct Binding Modes of Interaction with Peptide-Major Histocompatibility Complex Class I. J Biol Chem 2006; 281:28090-6. [PMID: 16840780 DOI: 10.1074/jbc.m604931200] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Interaction of CD8 (CD8alphaalpha or CD8alphabeta) with the peptide-major histocompatibility complex (MHC) class I (pMHCI) is critical for the development and function of cytolytic T cells. Although the crystal structure of CD8alphaalpha.pMHCI complex revealed that two symmetric CD8alpha subunits interact with pMHCI asymmetrically, with one subunit engaged in more extensive interaction than the other, the details of the interaction between the CD8alphabeta heterodimer and pMHCI remained unknown. The Ig-like domains of mouse CD8alphabeta and CD8alphaalpha are similar in the size, shape, and surface electrostatic potential of their pMHCI-binding regions, suggesting that their interactions with pMHCI could be very similar. Indeed, we found that the CD8alpha variants CD8alpha(R8A) and CD8alpha(E27A), which were functionally inactive as homodimers, could form an active co-receptor with wild-type (WT) CD8beta as a CD8alpha(R8A)beta or CD8alpha(E27A)beta heterodimer. We also identified CD8beta variants that could form active receptors with WT CD8alpha but not with CD8alpha(R8A). This observation is consistent with the notion that the CD8beta subunit may replace either CD8alpha subunit in CD8alphaalpha.pMHCI complex. In addition, we showed that both anti-CD8alpha and anti-CD8beta antibodies were unable to completely block the co-receptor activity of WT CD8alphabeta. We propose that CD8alphabeta binds to pMHCI in at least two distinguishable orientations.
Collapse
Affiliation(s)
- Hsiu-Ching Chang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02115, USA.
| | | | | |
Collapse
|
28
|
Chang HC, Tan K, Ouyang J, Parisini E, Liu JH, Le Y, Wang X, Reinherz EL, Wang JH. Structural and Mutational Analyses of a CD8αβ Heterodimer and Comparison with the CD8αα Homodimer. Immunity 2005; 23:661-71. [PMID: 16356863 DOI: 10.1016/j.immuni.2005.11.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2005] [Revised: 10/20/2005] [Accepted: 11/16/2005] [Indexed: 12/31/2022]
Abstract
The crystal structure of a recombinant mouse single chain CD8alphabeta ectodomains at 2.4 A resolution reveals paired immunoglobulin variable region-like domains with a striking resemblance to CD8alphaalpha in size, shape, and surface electrostatic potential of complementarity-determining regions (CDR), despite <20% sequence identity between the CD8alpha and CD8beta subunits. Unlike the CD8alpha subunit(s) in the heterodimer or homodimer, the CDR1 loop of CD8beta tilts away from its corresponding CDR2 and CDR3 loops. Consistent with this observation, independent mutational studies reveal that alanine substitutions of residues in the CDR1 loop of CD8beta have no effect on CD8alphabeta coreceptor function, whereas mutations in CD8beta CDR2 and CDR3 loops abolish CD8alphabeta coreceptor activity. The implications of these findings and additional CD8alpha mutational studies for CD8alphabeta- versus CD8alphaalpha-MHCI binding are discussed.
Collapse
Affiliation(s)
- Hsiu-Ching Chang
- Laboratory of Immunobiology, Department of Medical Oncology, Dana-Farber Cancer Institute, Department of Medicine, Harvard Medical School, Boston, Massachusetts 02115, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Stove V, Van de Walle I, Naessens E, Coene E, Stove C, Plum J, Verhasselt B. Human immunodeficiency virus Nef induces rapid internalization of the T-cell coreceptor CD8alphabeta. J Virol 2005; 79:11422-33. [PMID: 16103193 PMCID: PMC1193625 DOI: 10.1128/jvi.79.17.11422-11433.2005] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2005] [Accepted: 06/13/2005] [Indexed: 11/20/2022] Open
Abstract
Human immunodeficiency virus (HIV) Nef is a membrane-associated protein decreasing surface expression of CD4, CD28, and major histocompatibility complex class I on infected cells. We report that Nef strongly down-modulates surface expression of the beta-chain of the CD8alphabeta receptor by accelerated endocytosis, while CD8 alpha-chain expression is less affected. By mutational analysis of the cytoplasmic tail of the CD8 beta-chain, an FMK amino acid motif was shown to be critical for Nef-induced endocytosis. Although independent of CD4, endocytosis of the CD8 beta-chain was abrogated by the same mutations in Nef that affect CD4 down-regulation, suggesting common molecular interactions. The ability to down-regulate the human CD8 beta-chain was conserved in HIV-1, HIV-2, and simian immunodeficiency virus SIVmac239 Nef and required an intact AP-2 complex. The Nef-mediated internalization of receptors, such as CD4, major histocompatibility complex class I, CD28, and CD8alphabeta, may contribute to the subversion of the host immune system and progression towards AIDS.
Collapse
Affiliation(s)
- Veronique Stove
- Department of Clinical Chemistry, Microbiology and Immunology, Ghent University Hospita, Belgium
| | | | | | | | | | | | | |
Collapse
|
30
|
Giroux M, Denis F. Influence of calcium ions in the flow cytometric analysis of human CD8-positive cells. Cytometry A 2005; 62:61-4. [PMID: 15472901 DOI: 10.1002/cyto.a.20084] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
BACKGROUND The CD8 co-receptor is an important marker used to identify various lymphocyte subsets. A significant decrease in CD8alpha staining intensity was observed in the presence of divalent cation chelators. METHODS Peripheral blood mononuclear cells (PBMC) obtained from healthy volunteers were treated with calcium chelators, stained with different anti-human CD8 mAbs, and analyzed by flow cytometry. RESULTS Calcium chelators caused a dose-dependent decrease in fluorescence intensity, using specific anti-human CD8alpha mAbs. This phenomenon was not due to CD8 internalization and could be reversed by the addition of calcium ions. In contrast, calcium depletion increased staining intensity with one anti-CD8beta mAb. CONCLUSIONS Divalent cation chelators are used as cell anti-clumping agents in MACS or FACS applications. Researchers should be aware that such treatment could lead to the almost complete loss of fluorescence with selected anti-human CD8alpha mAbs. Since CD8 staining is used in conjunction with tetramer staining to identify antigen-specific cytotoxic human T cells, the effect of calcium depletion should be taken into account in experimental design.
Collapse
Affiliation(s)
- Martin Giroux
- INRS, Institut Armand-Frappier, 531 Boulevard des Prairies, Laval, Québec H7V 1B7, Canada
| | | |
Collapse
|
31
|
Blois SM, Joachim R, Kandil J, Margni R, Tometten M, Klapp BF, Arck PC. Depletion of CD8+ cells abolishes the pregnancy protective effect of progesterone substitution with dydrogesterone in mice by altering the Th1/Th2 cytokine profile. THE JOURNAL OF IMMUNOLOGY 2004; 172:5893-9. [PMID: 15128769 DOI: 10.4049/jimmunol.172.10.5893] [Citation(s) in RCA: 126] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
One of the most remarkable immunological regulations is the maternal immune tolerance toward the fetal semiallograft during pregnancy, which has been referred to as immunity's pregnant pause. Rejection of the semiallogeneic trophoblast cells must be selectively inhibited and pathways presumably include Th2 cytokines unopposed by Th1 cytokines. Steroid hormones, including progesterone, have similar effects. Low levels of progesterone and Th2 cytokines and high levels of Th1 cytokines are attributable for increased abortions in mammalians, which may be triggered by psychoemotional stress. Thus, the aim of the present study was to provide experimental evidence for the mechanism involved in the mediation of immune responses by endocrine signals during pregnancy and stress-triggered pregnancy failure. DBA/2J-mated CBA/J female mice were randomized in three groups: 1) control females, 2) mice exposed to stress on gestation day 5.5, and 3) mice exposed to stress and substituted with dydrogesterone, a progestogen with a binding profile highly selective for the progesterone receptor on gestation day 5.5. On gestation days 7.5, 9.5, and 10.5, mice of each group were sacrificed, and the frequency of CD8(+) cells and cytokine expression (IL-4, IL-12, TNF-alpha, IFN-gamma) in blood and uterus cells was evaluated by flow cytometry. Additionally, some mice were depleted of CD8 cells by injection of mAb. We observed that progesterone substitution abrogated the abortogenic effects of stress exposure by decreasing the frequency of abortogenic cytokines. This pathway was exceedingly CD8-dependent, because depletion of CD8 led to a termination of the pregnancy protective effect of progesterone substitution.
Collapse
Affiliation(s)
- Sandra M Blois
- Charité, Department of Internal Medicine, Biomedizinisches Forschungszentrum, Humboldt University of Berlin, Berlin, Germany
| | | | | | | | | | | | | |
Collapse
|
32
|
Gangadharan D, Cheroutre H. The CD8 isoform CD8αα is not a functional homologue of the TCR co-receptor CD8αβ. Curr Opin Immunol 2004; 16:264-70. [PMID: 15134773 DOI: 10.1016/j.coi.2004.03.015] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Although structurally similar, CD8alphabeta and CD8alphaalpha have notably diverted with regard to function. Whereas CD8alphabeta functions as a T-cell receptor (TCR) co-receptor on MHC-class-I-restricted thymocytes and mature T cells, CD8alphaalpha is unable to support conventional positive selection, and can be expressed on T cells independent of the MHC restriction of their TCR. CD8alphaalpha induction is consistent with antigenic stimulation through the TCR, and recent developments have now shown that CD8alphaalpha induced on agonist-triggered immature thymocytes, antigenic-stimulated conventional CD8alphabeta T cells and mucosal T cells mediates the specific modulation of TCR activation signals to facilitate their survival and differentiation into various specialized T-cell subsets.
Collapse
Affiliation(s)
- Denise Gangadharan
- Division of Developmental Immunology, La Jolla Institute for Allergy & Immunology, 10355 Science Center Drive, San Diego, California 92121, USA
| | | |
Collapse
|
33
|
Madakamutil LT, Christen U, Lena CJ, Wang-Zhu Y, Attinger A, Sundarrajan M, Ellmeier W, von Herrath MG, Jensen P, Littman DR, Cheroutre H. CD8alphaalpha-mediated survival and differentiation of CD8 memory T cell precursors. Science 2004; 304:590-3. [PMID: 15105501 DOI: 10.1126/science.1092316] [Citation(s) in RCA: 155] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Memory T cells are long-lived antigen-experienced T cells that are generally accepted to be direct descendants of proliferating primary effector cells. However, the factors that permit selective survival of these T cells are not well established. We show that homodimeric alpha chains of the CD8 molecule (CD8alphaalpha) are transiently induced on a selected subset of CD8alphabeta+ T cells upon antigenic stimulation. These CD8alphaalpha molecules promote the survival and differentiation of activated lymphocytes into memory CD8 T cells. Thus, memory precursors can be identified among primary effector cells and are selected for survival and differentiation by CD8alphaalpha.
Collapse
Affiliation(s)
- Loui T Madakamutil
- La Jolla Institute for Allergy and Immunology, 10355 Science Center Drive, San Diego, CA 92121, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Stievano L, Tosello V, Marcato N, Rosato A, Sebelin A, Chieco-Bianchi L, Amadori A. CD8+αβ+T Cells That Lack Surface CD5 Antigen Expression Are a Major Lymphotactin (XCL1) Source in Peripheral Blood Lymphocytes. THE JOURNAL OF IMMUNOLOGY 2003; 171:4528-38. [PMID: 14568926 DOI: 10.4049/jimmunol.171.9.4528] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
To better characterize the cellular source of lymphotactin (XCL1), we compared XCL1 expression in different lymphocyte subsets by real-time PCR. XCL1 was constitutively expressed in both PBMC and CD4(+) cells, but its expression was almost 2 log higher in CD8(+) cells. In vitro activation was associated with a substantial increase in XCL1 expression in both PBMC and CD8(+) cells, but not in CD4(+) lymphocytes. The preferential expression of XCL1 in CD8(+) cells was confirmed by measuring XCL1 production in culture supernatants, and a good correlation was found between figures obtained by real-time PCR and XCL1 contents. XCL1 expression was mostly confined to a CD3(+)CD8(+) subset not expressing CD5, where XCL1 expression equaled that shown by gammadelta(+) T cells. Compared with the CD5(+) counterpart, CD3(+)CD8(+)CD5(-) cells, which did not express CD5 following in vitro activation, showed preferential expression of the alphaalpha form of CD8 and a lower expression of molecules associated with a noncommitted/naive phenotype, such as CD62L. CD3(+)CD8(+)CD5(-) cells also expressed higher levels of the XCL1 receptor; in addition, although not differing from CD3(+)CD8(+)CD5(+) cells in terms of the expression of most alpha- and beta-chemokines, they showed higher expression of CCL3/macrophage inflammatory protein-1alpha. These data show that TCR alphabeta-expressing lymphocytes that lack CD5 expression are a major XCL1 source, and that the contribution to its synthesis by different TCR alphabeta-expressing T cell subsets, namely CD4(+) lymphocytes, is negligible. In addition, they point to the CD3(+)CD8(+)CD5(-) population as a particular T cell subset within the CD8(+) compartment, whose functional properties deserve further attention.
Collapse
Affiliation(s)
- Laura Stievano
- Department of Oncology and Surgical Sciences, University of Padova, Padova, Italy
| | | | | | | | | | | | | |
Collapse
|
35
|
Wong JS, Wang X, Witte T, Nie L, Carvou N, Kern P, Chang HC. Stalk region of beta-chain enhances the coreceptor function of CD8. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 171:867-74. [PMID: 12847256 DOI: 10.4049/jimmunol.171.2.867] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
CD8 glycoproteins are expressed as either alphaalpha homodimers or alphabeta heterodimers on the surface of T cells. CD8alphabeta is a more efficient coreceptor than the CD8alphaalpha for peptide Ag recognition by TCR. Each CD8 subunit is composed of four structural domains, namely, Ig-like domain, stalk region, transmembrane region, and cytoplasmic domain. In an attempt to understand why CD8alphabeta is a better coreceptor than CD8alphaalpha, we engineered, expressed, and functionally tested a chimeric CD8alpha protein whose stalk region is replaced with that of CD8beta. We found that the beta stalk region enhances the coreceptor function of chimeric CD8alphaalpha to a level similar to that of CD8alphabeta. Surprisingly, the beta stalk region also restored functional activity to an inactive CD8alpha variant, carrying an Ala mutation at Arg(8) (R8A), to a level similar to that of wild-type CD8alphabeta. Using the R8A variant of CD8alpha, a panel of anti-CD8alpha Abs, and three MHC class I (MHCI) variants differing in key residues known to be involved in CD8alpha interaction, we show that the introduction of the CD8beta stalk leads to a different topology of the CD8alpha-MHCI complex without altering the overall structure of the Ig-like domain of CD8alpha or causing the MHCI to employ different residues to interact with the CD8alpha Ig domain. Our results show that the stalk region of CD8beta is capable of fine-tuning the coreceptor function of CD8 proteins as a coreceptor, possibly due to its distinct protein structure, smaller physical size and the unique glycan adducts associated with this region.
Collapse
MESH Headings
- Adjuvants, Immunologic/genetics
- Adjuvants, Immunologic/metabolism
- Adjuvants, Immunologic/physiology
- Alanine/genetics
- Amino Acid Sequence
- Animals
- Antibodies, Blocking/pharmacology
- Antibodies, Monoclonal/pharmacology
- Antigen Presentation/genetics
- Arginine/genetics
- CD8 Antigens/genetics
- CD8 Antigens/immunology
- CD8 Antigens/metabolism
- CD8 Antigens/physiology
- Dimerization
- Glycosylation
- H-2 Antigens/genetics
- H-2 Antigens/physiology
- Immunoglobulins/metabolism
- Immunoglobulins/physiology
- Mice
- Molecular Sequence Data
- Mutagenesis, Site-Directed
- Protein Structure, Tertiary/genetics
- Protein Subunits/genetics
- Protein Subunits/immunology
- Protein Subunits/metabolism
- Protein Subunits/physiology
- Receptors, Immunologic/genetics
- Receptors, Immunologic/immunology
- Receptors, Immunologic/metabolism
- Receptors, Immunologic/physiology
- Recombinant Fusion Proteins/chemical synthesis
- Recombinant Fusion Proteins/metabolism
- Recombinant Fusion Proteins/physiology
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Jenny S Wong
- Dana-Farber Cancer Institute, Department of Cancer Immunology and AIDS, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | |
Collapse
|
36
|
Konno A, Okada K, Mizuno K, Nishida M, Nagaoki S, Toma T, Uehara T, Ohta K, Kasahara Y, Seki H, Yachie A, Koizumi S. CD8alpha alpha memory effector T cells descend directly from clonally expanded CD8alpha +beta high TCRalpha beta T cells in vivo. Blood 2002; 100:4090-7. [PMID: 12393564 DOI: 10.1182/blood-2002-04-1136] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Whereas most peripheral CD8(+) alphabeta T cells highly express CD8alphabeta heterodimer in healthy individuals, there is an increase of CD8alpha(+)beta(low) or CD8alphaalpha alphabeta T cells in HIV infection or Wiskott-Aldrich syndrome and after bone marrow transplantation. The significance of these uncommon cell populations is not well understood. There has been some question as to whether these subsets and CD8alpha(+)beta(high) cells belong to different ontogenic lineages or whether a fraction of CD8alpha(+)beta(high) cells have down-regulated CD8beta chain. Here we assessed clonality of CD8alphaalpha and CD8alpha(+)beta(low) alphabeta T cells as well as their phenotypic and functional characteristics. Deduced from surface antigens, cytotoxic granule constituents, and cytokine production, CD8alpha(+)beta(low) cells are exclusively composed of effector memory cells. CD8alphaalpha cells comprise effector memory cells and terminally differentiated CD45RO(-)CCR7(-) memory cells. T-cell receptor (TCR) Vbeta complementarity-determining region 3 (CDR3) spectratyping analysis and subsequent sequencing of CDR3 cDNA clones revealed polyclonality of CD8alpha(+)beta(high) cells and oligoclonality of CD8alpha(+)beta(low) and CD8alphaalpha cells. Importantly, some expanded clones within CD8alphaalpha cells were also identified within CD8alpha(+)beta(high) and CD8alpha(+)beta(low) subpopulations. Furthermore, signal-joint TCR rearrangement excision circles concentration was reduced with the loss of CD8beta expression. These results indicated that some specific CD8alpha(+)beta(high) alphabeta T cells expand clonally, differentiate, and simultaneously down-regulate CD8beta chain possibly by an antigen-driven mechanism. Provided that antigenic stimulation directly influences the emergence of CD8alphaalpha alphabeta T cells, these cells, which have been previously regarded as of extrathymic origin, may present new insights into the mechanisms of autoimmune diseases and immunodeficiencies, and also serve as a useful biomarker to evaluate the disease activities.
Collapse
Affiliation(s)
- Akihiro Konno
- Department of Pediatrics, Angiogenesis and Vascular Development, Graduate School of Medical Science and School of Medicine, Kanazawa University, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Bakhiet M, Hamadien M, Tjernlund A, Mousal A, Seiger A. African trypanosomes activate human fetal brain cells to proliferation and IFN-gamma production. Neuroreport 2002; 13:53-6. [PMID: 11924894 DOI: 10.1097/00001756-200201210-00015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
We addressed the host-parasite interplay and the immunopathogenetic events occurring in the central nervous system (CNS) during human African trypanosomiasis. Human first trimester forebrain cells were stimulated with a trypanosome lymphocyte-triggering factor (TLTF) and studied for their immune response as exemplified by cell proliferation and IFN-gamma production. TLTF induced proliferation of human first trimester forebrain cells and IFN-gamma production at the mRNA and protein levels. Astrocytes are the major producers of IFN-gamma in response toTLTE These data illustrated for the first time a direct effect of a parasite factor on human brain cells. TargetingTLTF during the course of the disease may be considered in preventing the deadly neurological complications of human African trypanosomiasis. NeuroReport
Collapse
Affiliation(s)
- Moiz Bakhiet
- Center for Infectious Medicine, Karolinska Institutet, Huddinge University Hospital, Sweden
| | | | | | | | | |
Collapse
|
38
|
de la Calle-Martin O, Hernandez M, Ordi J, Casamitjana N, Arostegui JI, Caragol I, Ferrando M, Labrador M, Rodriguez-Sanchez JL, Espanol T. Familial CD8 deficiency due to a mutation in the CD8 alpha gene. J Clin Invest 2001; 108:117-23. [PMID: 11435463 PMCID: PMC209336 DOI: 10.1172/jci10993] [Citation(s) in RCA: 70] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
CD8 glycoproteins play an important role in both the maturation and function of MHC class I-restricted T lymphocytes. A 25-year-old man, from a consanguineous family, with recurrent bacterial infections and total absence of CD8(+) cells, was studied. Ab deficiencies and ZAP-70 and TAP defects were ruled out. A missense mutation (gly90-->ser) in both alleles of the immunoglobulin domain of the CD8 alpha gene was shown to correlate with the absence of CD8 expression found in the patient and two sisters. Conversely, high percentages of CD4(-)CD8(-)TCR alpha beta(+) T cells were found in the three siblings. A novel autosomal recessive immunologic defect characterized by absence of CD8(+) cells is described. These findings may help to further understanding of the role of CD8 molecules in human immune response.
Collapse
Affiliation(s)
- O de la Calle-Martin
- Department of Immunology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
McDonough SP, Moore PF. Clinical, hematologic, and immunophenotypic characterization of canine large granular lymphocytosis. Vet Pathol 2000; 37:637-46. [PMID: 11105953 DOI: 10.1354/vp.37-6-637] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Clinical, hematologic, and immunophenotypic data were studied in 25 dogs with large granular lymphocyte (LGL) lymphocytosis. Primarily large-breed dogs were affected, with an average age at initial diagnosis of 10 years (range 5-14 years). All dogs had persistent (>4 months) LGL lymphocytosis except for three that were euthanized with aggressive disease. Splenomegaly was reported in 12 of 20 dogs in which splenic size was evaluated. The clinical course was heterogeneous and dogs were divided into four groups based on similar clinical and hematologic findings: acute leukemia (3/25), persistent lymphocytosis with anemia (12/25), persistent lymphocytosis without anemia (8/25), and reactive lymphocytosis (2/25). Immunophenotypes varied within groups but were homogeneous among cells from the same patient except in the two dogs classified as reactive LGL lymphocytosis. Analysis of T-cell receptor (TCR) usage identified three main LGL lineages. TCRalphabeta was expressed in 15/25 (60%) cases. TCRgammadelta was expressed in 8/25 (32%) cases, and 2/25 (8%) cases were CD3-, compatible with NK cells. beta2 integrin expression was distinctive. CD11a was consistently expressed, while CD11b was absent. CD11c was expressed only weakly in 16/25 (64%) cases. The leukointegrin alphadbeta2 was highly prevalent on all LGL lineages, being expressed in 23/25 (92%) cases. Prominent involvement of the spleen, relative sparing of bone marrow, an unexpectedly large proportion of gammadelta T-cell LGLs, and the distinctive beta2 integrin expression pattern on diverse lineages of LGLs suggest the disease arises from unique populations of lymphocytes that preferentially localize in the splenic red pulp.
Collapse
Affiliation(s)
- S P McDonough
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, Davis, USA.
| | | |
Collapse
|
40
|
Hibbard MK, Strehl S, Lalande M. Replication timing of CD4 and CD8 in single-positive peripheral blood lymphocytes. Cell Immunol 1999; 198:61-8. [PMID: 10612652 DOI: 10.1006/cimm.1999.1582] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The regulatory events leading to the mutually exclusive expression of CD4 and CD8 on peripheral lymphocytes are not fully understood. In particular, the association between DNA replication timing and transcriptional activity of these genes has not been previously investigated. Here, the replication kinetics of the CD4 and CD8 loci in mature single-positive T-cell populations have been examined using a novel approach to the separation of CD4(+) or CD8(+) lymphocytes into discrete cell cycle fractions and a competitive PCR replication timing assay. While the timing of replication of each of these loci is independent of their expression in mature CD4 or CD8 single positive T-cells, the replication of CD8, but not of CD4, shifts to a later time in S phase in transcriptionally silent HS68 fibroblast cells. These findings suggest that changes in DNA replication timing are associated with the developmentally regulated but not with the tissue-specific expression of CD4 and CD8.
Collapse
Affiliation(s)
- M K Hibbard
- Genetics Division, Children's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | |
Collapse
|
41
|
Fayen JD, Tykocinski ML. The expansion of human gammadelta T cells in response to Daudi cells requires the participation of CD4+ T cells. Immunology 1999; 97:272-9. [PMID: 10447742 PMCID: PMC2326839 DOI: 10.1046/j.1365-2567.1999.00761.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The Burkitt's lymphoma cell line Daudi is a potent inducer of human gammadelta T-cell expansion. Using an in vitro culture system comprised of irradiated Daudi cells as stimulators and normal human lymphocytes as responders, the cellular determinants of this response were investigated. Three of four monoclonal antibodies (mAbs 1-1C4, L243, and 9.3F10) directed against disparate epitopes of human major histocompatibility complex (MHC) class II, as well as a mAb with specificity for CD4 (OKT4), inhibited the expansion of gammadelta T cells in response to Daudi cell stimulators. mAbs with a specificity for CD74 and CD8 were non-inhibitory. Lymphocyte depletion experiments demonstrated a critical role for the CD4+ T-cell subset in the expansion of gammadelta T cells. Other data pointed towards requirements for direct cell contact in this system, and the addition of exogenous recombinant interleukin (IL)-2, IL-4, and IL-12 failed to reconstitute gammadelta T-cell expansion in CD4+ lymphocyte-depleted cultures. These results complement previous findings in murine infectious disease and mycobacterial systems, providing a direct demonstration that CD4+ T cells play a role in gammadelta T-cell expansion through an interaction with human leucocyte antigen (HLA) class II on Daudi cells. The data point towards important functional links between the acquired and natural immune systems.
Collapse
Affiliation(s)
- J D Fayen
- Institute of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | | |
Collapse
|
42
|
Kuroda MJ, Schmitz JE, Charini WA, Nickerson CE, Lord CI, Forman MA, Letvin NL. Comparative analysis of cytotoxic T lymphocytes in lymph nodes and peripheral blood of simian immunodeficiency virus-infected rhesus monkeys. J Virol 1999; 73:1573-9. [PMID: 9882363 PMCID: PMC103982 DOI: 10.1128/jvi.73.2.1573-1579.1999] [Citation(s) in RCA: 72] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/1998] [Accepted: 10/31/1998] [Indexed: 11/20/2022] Open
Abstract
Most studies of human immunodeficiency virus type 1 (HIV-1)-specific cytotoxic T lymphocytes (CTL) have been confined to the evaluation of these effector cells in the peripheral blood. What has not been clear is the extent to which CTL activity in the blood actually reflects this effector cell function in the lymph nodes, the major sites of HIV-1 replication. To determine the concordance between CTL activity in lymph nodes and peripheral blood lymphocytes (PBL), CTL specific for simian immunodeficiency virus of macaques (SIVmac) have been characterized in lymph nodes of infected, genetically selected rhesus monkeys by using both Gag peptide-specific functional CTL assays and tetrameric peptide-major histocompatibility complex (MHC) class I molecule complex staining techniques. In studies of six chronically SIVmac-infected rhesus monkeys, Gag epitope-specific functional lytic activity and specific tetrameric peptide-MHC class I staining were readily demonstrated in lymph node T lymphocytes. Although the numbers of tetramer-binding cells in some animals differed from those documented in their PBL, the numbers of tetramer-binding cells from these two different compartments were not statistically different. Phenotypic characterization of the tetramer-binding CD8(+) lymph node T lymphocytes of the infected monkeys demonstrated a high level of expression of the activation-associated adhesion molecules CD11a and CD49d, the Fas molecule CD95, and MHC class II-DR. These studies documented a low expression of the naive T-cell marker CD45RA and the adhesion molecule CD62L. This phenotypic profile of the tetramer-binding lymph node CD8(+) T cells was similar to that of tetramer-binding CD8(+) T cells from PBL. These observations suggest that characterization of AIDS virus-specific CTL activity by sampling of cells in the peripheral blood should provide a reasonable estimation of CTL in an individual's secondary lymphoid tissue.
Collapse
Affiliation(s)
- M J Kuroda
- Division of Viral Pathogenesis, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA.
| | | | | | | | | | | | | |
Collapse
|
43
|
Devine L, Sun J, Barr MR, Kavathas PB. Orientation of the Ig Domains of CD8αβ Relative to MHC Class I. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.162.2.846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Abstract
The cell surface glycoprotein CD8 functions as a coreceptor with the TCR for interaction with MHC class I. The cocrystal structure of the CD8αα-MHC complex showed that one CD8 Ig domain provided the majority of the contact with MHC class I and that residue R4 of that domain contacted the α2 domain of MHC class I. We previously showed by mutational analysis that this residue was critical for binding to MHC class I. To determine which of the Ig domains for the CD8αβ heterodimer would make the most contact with class I MHC, we expressed single-chain or dimeric forms of CD8 on COS-7 cells and measured the adhesion of MHC class I positive cells. We found that when one of the R4 residues was mutated in a CD8αα homodimer binding comparable to that of wild type was observed, whereas a double R4 mutant severely impaired binding. However, when mutant CD8α (R4K) was coexpressed with wild-type CD8β, binding was not observed. These results support the model in which it is CD8α, not CD8β, that is making the most of the contact with MHC class I, including the α2 domain. In addition, they demonstrate that a single-chain form of CD8αα can bind to MHC class I.
Collapse
Affiliation(s)
- Lesley Devine
- *Department of Laboratory Medicine and Section of Immunobiology, and
| | - Jiaren Sun
- *Department of Laboratory Medicine and Section of Immunobiology, and
| | - Mark R. Barr
- *Department of Laboratory Medicine and Section of Immunobiology, and
| | - Paula B. Kavathas
- *Department of Laboratory Medicine and Section of Immunobiology, and
- †Department of Genetics, Yale University School of Medicine, New Haven, CT 06520
| |
Collapse
|
44
|
Mulligan SP, Dao LP, Francis SE, Thomas ME, Gibson J, Cole-Sinclair MF, Wolf M. B-cell chronic lymphocytic leukaemia with CD8 expression: report of 10 cases and immunochemical analysis of the CD8 antigen. Br J Haematol 1998; 103:157-62. [PMID: 9792303 DOI: 10.1046/j.1365-2141.1998.00928.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
We report 10 cases of B-cell chronic lymphocytic leukaemia (B-CLL) with expression of the T-cell antigen CD8. The majority of patients had typical B-cell CLL with stable and non-progressive stage A(O) disease except for more common expression of lambda light chain and CD25. Two patients had progressive disease and required therapy, one with atypical morphological and phenotypic features. The incidence of CD8 expression was approximately 0.5% of B-CLL patients from our institutions. Immunoprecipitation of the CD8 antigen from four of these B-CLLs showed identity to the CD8 antigen expressed on T cells with precipitation of CD8alpha bands of molecular weight approximately 34 kD. In view of the known intracellular signalling mechanism of CD8 using the tyrosine kinase p56-lck, we studied p56-lck expression by Western blot and found lack of consistent expression of the CD8 surface antigen, with most lacking p56-lck. Our report indicates that CD8 expression in B-CLL is probably underrecognized but is not a marker of disease progression. The CD8 on the B-CLL surface is immunochemically identical to the antigen on T cells, but is not accompanied by its usual signalling mechanism of p56-lck tyrosine kinase and therefore is unlikely to be a functionally active receptor.
Collapse
Affiliation(s)
- S P Mulligan
- Department of Haematology, Concord Hospital, Sydney, Australia
| | | | | | | | | | | | | |
Collapse
|
45
|
Moore LR, Zborowski M, Sun L, Chalmers JJ. Lymphocyte fractionation using immunomagnetic colloid and a dipole magnet flow cell sorter. JOURNAL OF BIOCHEMICAL AND BIOPHYSICAL METHODS 1998; 37:11-33. [PMID: 9825297 DOI: 10.1016/s0165-022x(98)00010-4] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The relationship between cell function and surface marker expression is a subject of active investigation in biology and medicine. These investigations require separating cells of a homogeneous subset into multiple fractions of varying marker expression. We have developed a novel cell sorter, the dipole magnet flow sorter (DMFS), which separates selected T lymphocyte subpopulations, targeted by immunomagnetic colloid, into multiple fractions according to cell surface marker expression, as determined by flow cytometry. A narrow stream of cells is introduced into a sheath of carrier fluid in a rectangular channel while subjected to a perpendicular magnetic force. The special design of the pole pieces ensures a constant magnetic force acting on the magnetically labeled cells in the separation area. Cells are spread across the flow in relation to their magnetophoretic mobility. Separation is achieved by control of the positions of the effluent stream boundaries, which separate fluid volumes with cells of different magnetophoretic mobility. CD4 and CD8 T lymphocytes labeled with primary antibody-fluorescein isothiocyanate (FITC) conjugate and anti-FITC-magnetic colloid are the chosen cell systems. Flow cytometry analysis shows that, for CD4 cells, a three-fold increase in total marker number per cell is observed when comparing the highest to the lowest fluorescence fractions. Similarly, a four-fold increase in total marker number is observed for CD8 cells. We also observed the separation of two dissimilar cell types that differed in expression of the CD4 marker, monocytes and T helper lymphocytes. We believe that this type of separation is applicable to any cells in suspension for which a suitable antibody exists and, due to the comparatively gentle nature of the process, is particularly suitable for the sorting of fragile cells.
Collapse
Affiliation(s)
- L R Moore
- Department of Biomedical Engineering, Cleveland Clinic Foundation, OH 44195-5254, USA.
| | | | | | | |
Collapse
|
46
|
Expression of the CD8αβ-Heterodimer on CD8+ T Lymphocytes in Peripheral Blood Lymphocytes of Human Immunodeficiency Virus− and Human Immunodeficiency Virus+Individuals. Blood 1998. [DOI: 10.1182/blood.v92.1.198.413k13_198_206] [Citation(s) in RCA: 45] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
CD8+ T lymphocytes play a pivotal role in controlling human immunodeficiency virus (HIV)-1 replication in vivo. We have performed four-color flow cytometric analysis of CD8+peripheral blood lymphocytes (PBL) from 21 HIV-1 seronegative and 103 seropositive individuals to explore the phenotypic heterogeneity of CD8β-chain expression on CD8+ T lymphocytes and to clarify how its expression on CD8+ T lymphocytes may relate to acquired immunodeficiency syndrome (AIDS) clinical progression. We showed that the single monoclonal antibody (MoAb) 2ST8-5H7, directed against the CD8αβ-heterodimer, identifies CD8+ T lymphocytes as effectively as the conventional combination of anti-CD3 and anti-CD8α antibodies. However, we detected a significantly lower mean fluorescence (MF) of anti-CD8αβ staining on PBL from HIV-1 seropositive donors as compared with seronegative donors. In fact, CD8+ T lymphocytes from HIV-1–infected individuals with the lowest CD4 counts showed the lowest levels of CD8αβ MF. To explore further this change in CD8αβ expression, we assessed the expression of 14 different cell surface molecules on CD8αβ+ T lymphocytes of PBL from 11 HIV-1 seronegative and 22 HIV-1 seropositive individuals. The MF of anti-CD8αβ staining was significantly reduced on CD8+T lymphocyte subsets that showed immunophenotypic evidence of activation. The subset of lymphocytes expressing low levels of CD8αβ expressed higher levels of activation, adhesion, and cytotoxic-associated molecules and was predominantly CD45RO+ and CD28−. Finally, we monitored the expression of the CD8αβ-heterodimer on PBL of eight HIV-1–infected individuals over a 16-week period after the initiation of highly active antiretroviral therapy (HAART), including zidovudine (ZDV), lamivudine (3TC), and indinavir (IDV), and found a significant increase in the expression of the CD8αβ-heterodimer. These results suggest that antibodies recognizing the CD8αβ-heterodimer are useful tools to specifically identify CD8+ T lymphocytes. Moreover, the quantitative monitoring of CD8αβ expression allows the detection of discrete CD8+ T lymphocyte subsets and may be useful for assessing the immune status of individuals infected with HIV-1.
Collapse
|
47
|
Kuroda MJ, Schmitz JE, Barouch DH, Craiu A, Allen TM, Sette A, Watkins DI, Forman MA, Letvin NL. Analysis of Gag-specific cytotoxic T lymphocytes in simian immunodeficiency virus-infected rhesus monkeys by cell staining with a tetrameric major histocompatibility complex class I-peptide complex. J Exp Med 1998; 187:1373-81. [PMID: 9565630 PMCID: PMC2212269 DOI: 10.1084/jem.187.9.1373] [Citation(s) in RCA: 246] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/1997] [Revised: 02/02/1997] [Indexed: 11/23/2022] Open
Abstract
A tetrameric recombinant major histocompatibility complex (MHC) class I-peptide complex was used as a staining reagent in flow cytometric analyses to quantitate and define the phenotype of Gag-specific cytotoxic T lymphocytes (CTLs) in the peripheral blood of simian immunodeficiency virus macaque (SIVmac)-infected rhesus monkeys. The heavy chain of the rhesus monkey MHC class I molecule Mamu-A*01 and beta2-microglobulin were refolded in the presence of an SIVmac Gag synthetic peptide (p11C, C-M) representing the optimal nine-amino acid peptide of Mamu-A*01-restricted predominant CTL epitope to create a tetrameric Mamu-A*01/p11C, C-M complex. Tetrameric Mamu-A*01/p11C, C-M complex bound to T cells of SIVmac-infected, Mamu-A*01(+), but not uninfected, Mamu-A*01(+), or infected, Mamu-A*01(-) rhesus monkeys. Specific staining of peripheral blood mononuclear cells (PBMC) from SIVmac-infected, Mamu-A*01(+) rhesus monkeys was only found in the cluster of differentiation (CD)8alpha/beta+ T lymphocyte subset and the percentage of CD8alpha/beta+ T cells in the peripheral blood of four SIVmac-infected, Mamu-A*01+ rhesus monkeys staining with this complex ranged from 0.7 to 10.3%. Importantly, functional SIVmac Gag p11C-specific CTL activity was seen in sorted and expanded tetrameric Mamu-A*01/p11C, C-M complex-binding, but not nonbinding, CD8alpha/beta+ T cells. Furthermore, the percentage of CD8alpha/beta+ T cells binding this tetrameric Mamu-A*01/p11C, C-M complex correlated well with p11C-specific cytotoxic activity as measured in both bulk and limiting dilution effector frequency assays. Finally, phenotypic characterization of the cells binding this tetrameric complex indicated that this lymphocyte population is heterogeneous. These studies indicate the power of this approach for examining virus-specific CTLs in in vivo settings.
Collapse
Affiliation(s)
- M J Kuroda
- Harvard Medical School, Division of Viral Pathogenesis, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02215, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Bacsó Z, Bene L, Bodnár A, Matkó J, Damjanovich S. A photobleaching energy transfer analysis of CD8/MHC-I and LFA-1/ICAM-1 interactions in CTL-target cell conjugates. Immunol Lett 1996; 54:151-6. [PMID: 9052870 DOI: 10.1016/s0165-2478(96)02665-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The photobleaching energy transfer (pbFRET) technique is a fluorescence method to measure proximity relationships between molecules, especially cell surface proteins, labeled with fluorophore-conjugated monoclonal antibodies, on a pixel-by-pixel base using digital imaging microscopy. This technique enables analysis of inter- and intramolecular proximities at cell surfaces at physiological conditions. We have developed a pbFRET approach to measure intercellular proximities in order to access spatial organization of interacting proteins in the contact region of two 'communicating' cells. Two examples, as possible application areas of this approach, are presented here: interaction between CD8 and MHC-I molecules in point contacts and interaction between LFA-1 and ICAM-1 molecules in focal contacts of CTL-target conjugates. The geometry of these protein contacts based on our resonance energy transfer (RET) data is consistent with the observed blocking effects of monoclonal antibodies (directed against the interacting proteins) on the cytolytic activity of CTLs and suggest a critical role for CD8beta-subunit in signal transmission in peripheral T-lymphocytes.
Collapse
Affiliation(s)
- Z Bacsó
- Department of Biophysics, Medical University School of Debrecen, Hungary
| | | | | | | | | |
Collapse
|
49
|
Bakhiet M, Olsson T, Ljungdahl A, Höjeberg B, Van Der Meide P, Kristensson K. Induction of interferon-gamma, transforming growth factor-beta, and interleukin-4 in mouse strains with different susceptibilities to Trypanosoma brucei brucei. J Interferon Cytokine Res 1996; 16:427-33. [PMID: 8807495 DOI: 10.1089/jir.1996.16.427] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
A Trypanosoma brucei brucei-derived lymphocyte triggering factor (TLTF) induced CD8+ T cells to produce IFN-gamma, which in turn stimulates parasite growth. This parasite-host interaction was studied in mouse strains that are either relatively susceptible (C3H/He) or resistant (C57Bl/6J) to infection, as well as in athymic nude mice. In all mouse strains, T. b. brucei infection caused a strong induction of IFN-gamma production by spleen mononuclear cells (MNC). In vivo blocking of IFN-gamma by intraperitoneal injection of mouse monoclonal anti-IFN-gamma antibody suppressed parasite growth and increased survival of both C3H/H3 and C57Bl/6J animals, suggesting that, irrespective of strain-related disease susceptibility, IFN-gamma is a growth-promoting stimulus for T. b. brucei. Spleen MNC from noninfected mice of all strains were in vitro like-wise strongly induced to IFN-gamma production when exposed to TLTF. This suggests that CD8+ expressing T cell receptor (TCR) alpha/beta, gamma/delta-bearing T cells and NK cells may all be triggered to IFN-gamma production by TLTF. In all mouse strains, TLTF also caused an increase in the number of cells expressing mRNA for TGF-beta in vitro. However, significant triggering to IL-4 mRNA expression only occurred in the relatively disease-resistant C57Bl/6J strain. As IL-4 is required for the synthesis and class switches of immunoglobulins, which are essential host immune defenses against T. b. brucei, the degree of resistance may be related to inherent strain ability to produce IL-4 in response to TLTF.
Collapse
Affiliation(s)
- M Bakhiet
- Department of Medicine, Karolinska Hospital, Stockholm, Sweden
| | | | | | | | | | | |
Collapse
|
50
|
Luhtala M, Koskinen R, Toivanen P, Vainio O. Characterization of chicken CD8-specific monoclonal antibodies recognizing novel epitopes. Scand J Immunol 1995; 42:171-4. [PMID: 7543213 DOI: 10.1111/j.1365-3083.1995.tb03641.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
CD8 is a heterodimeric cell surface glycoprotein expressed primarily on thymocytes and a subpopulation of mature T lymphocytes. It binds to the invariant part of the major histocompatibility complex class I molecule and participates in antigen recognition by the major histocompatibility complex class I restricted T cells. As in mammalian species, the majority of chicken thymocytes express both CD4 and CD8, whereas peripheral T cells are either CD4- or CD8-positive. We have created a panel of mouse monoclonal antibodies detecting different cell surface epitopes on chicken CD8. The antibodies precipitate a 32-34 kDa dimeric protein from surface labelled thymocytes under reducing conditions. The identical N-deglycosylation pattern confirms that these MoAb precipitate the same heterodimeric molecule from chicken thymocyte lysates. Binding of 11-38 and 11-39 MoAb to peripheral blood T cells is totally inhibited by 11-39 and previously characterized CT8 and EP72 MoAb, further confirming their CD8 specificity. CD8 alpha-chain specificity of MoAb 11-39, 11-38, 11-30 and 11-13 is conclusively proven by staining COS-cells transfected with a plasmid containing CD8 alpha cDNA. However, MoAb 11-13, 11-30 and 11-38 do not compete with MoAb 11-39 in binding to CD8. These results demonstrate recognition of different epitopes by these MoAb. Monoclonal antibodies detecting novel epitopes on chicken CD8 provide a valuable tool for further studies on T cell development.
Collapse
Affiliation(s)
- M Luhtala
- Department of Medical Microbiology, Turku University, Finland
| | | | | | | |
Collapse
|