1
|
Kastner AL, Marx AF, Dimitrova M, Abreu-Mota T, Ertuna YI, Bonilla WV, Stauffer K, Künzli M, Wagner I, Kreutzfeldt M, Merkler D, Pinschewer DD. Durable lymphocyte subset elimination upon a single dose of AAV-delivered depletion antibody dissects immune control of chronic viral infection. Immunity 2025; 58:481-498.e10. [PMID: 39719711 DOI: 10.1016/j.immuni.2024.11.021] [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: 06/19/2024] [Revised: 10/02/2024] [Accepted: 11/26/2024] [Indexed: 12/26/2024]
Abstract
To interrogate the role of specific immune cells in infection, cancer, and autoimmunity, immunologists commonly use monoclonal depletion antibodies (depletion-mAbs) or genetically engineered mouse models (GEMMs). To generate a tool that combines specific advantages and avoids select drawbacks of the two methods, we engineered adeno-associated viral vectors expressing depletion mAbs (depletion-AAVs). Single-dose depletion-AAV administration durably eliminated lymphocyte subsets in mice and avoided accessory deficiencies of GEMMs, such as marginal zone defects in B cell-deficient animals. Depletion-AAVs can be used in animals of different genetic backgrounds, and multiple depletion-AAVs can readily be combined. Exploiting depletion-AAV technology, we showed that B cells were required for unimpaired CD4+ and CD8+ T cell responses to chronic lymphocytic choriomeningitis virus (LCMV) infection. Upon B cell depletion, CD8+ T cells failed to suppress viremia, and they only helped resolve chronic infection when antibodies dampened viral loads. Our study positions depletion-AAVs as a versatile tool for immunological research.
Collapse
Affiliation(s)
- Anna Lena Kastner
- Department of Biomedicine, University of Basel, 4009 Basel, Switzerland
| | | | - Mirela Dimitrova
- Department of Biomedicine, University of Basel, 4009 Basel, Switzerland
| | - Tiago Abreu-Mota
- Department of Biomedicine, University of Basel, 4009 Basel, Switzerland
| | - Yusuf I Ertuna
- Department of Biomedicine, University of Basel, 4009 Basel, Switzerland
| | - Weldy V Bonilla
- Department of Biomedicine, University of Basel, 4009 Basel, Switzerland
| | - Karsten Stauffer
- Department of Biomedicine, University of Basel, 4009 Basel, Switzerland
| | - Marco Künzli
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., 4070 Basel, Switzerland
| | - Ingrid Wagner
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva, Switzerland
| | - Mario Kreutzfeldt
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva, Switzerland; Division of Clinical Pathology, Geneva University Hospital, 1206 Geneva, Switzerland
| | - Doron Merkler
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva, Switzerland; Division of Clinical Pathology, Geneva University Hospital, 1206 Geneva, Switzerland
| | | |
Collapse
|
2
|
Malacopol AT, Holst PJ. Cancer Vaccines: Recent Insights and Future Directions. Int J Mol Sci 2024; 25:11256. [PMID: 39457036 PMCID: PMC11508577 DOI: 10.3390/ijms252011256] [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: 09/11/2024] [Revised: 10/13/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024] Open
Abstract
The field of cancer immunotherapy has seen incredible advancements in the past decades. mRNA-based cancer vaccines generating de novo T cell responses, particularly against tumor-specific antigens (TSAs), have demonstrated promising clinical outcomes and overcome diverse challenges. Despite the high potential of neoantigens to provide personalized immunotherapies through their tumor specificity and immunogenicity, challenges related to the scarcity of immunogenic neoepitopes have prompted continuous research towards finding new tumor-associated antigens (TAAs) and broader therapeutic frameworks, which may now learn from the genuine successes obtained with neoantigens. As an example, human endogenous retroviruses (HERVs) have emerged as potential alternatives to tumor neoantigens due to their high tumoral expression and ability to elicit both T cell reactivity and B cell responses associated with the efficacy of existing immunotherapies. This review aims to assess the status and limitations of TSA-directed mRNA cancer vaccines and the lessons that can be derived from these and checkpoint inhibitor studies to guide TAA vaccine development. We expect that shared B cell, CD4 and CD8 T cell antigen presentation will be key to stimulate continuous T cell expansion and efficacy for tumors that do not contain pre-existing tertiary lymphoid structures. When these structures are present in highly mutated tumors, the current checkpoint-based immunotherapies show efficacy even in immune privileged sites, and vaccines may hold the key to broaden efficacy to more tumor types and stages.
Collapse
Affiliation(s)
- Aretia-Teodora Malacopol
- HERVOLUTION Therapeutics, Copenhagen Bio Science (COBIS), 215 Nordre Fasanvej, DK2200 Copenhagen, Denmark;
| | - Peter Johannes Holst
- HERVOLUTION Therapeutics, Copenhagen Bio Science (COBIS), 215 Nordre Fasanvej, DK2200 Copenhagen, Denmark;
- Department of Biomedical Sciences, The Panum Institute, University of Copenhagen, DK2200 Copenhagen, Denmark
| |
Collapse
|
3
|
Zhong S, Li Q, Wen C, Li Y, Zhou Y, Jin Z, Ye G, Zhao Y, Hou J, Li Y, Tang L. Interferon α facilitates anti-HBV cellular immune response in a B cell-dependent manner. Antiviral Res 2022; 207:105420. [PMID: 36165866 DOI: 10.1016/j.antiviral.2022.105420] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 09/05/2022] [Accepted: 09/13/2022] [Indexed: 11/27/2022]
Abstract
OBJECTIVES Dissecting the underlying mechanism of T cells remodeling mediated by interferon α (IFN-α) is indispensable for achieving an optimum therapeutic response in chronic hepatitis B (CHB) patients. However, little is known about B cells in this process. This study aims to elucidate the roles of B cells in IFN-α-mediated anti-hepatitis B virus (HBV) cellular immunity. METHOD The effects of B cells on IFN-α-mediated T cell response were investigated in B cell-deficient mouse model with HBV and IFN-α plasmid hydrodynamic injection. Single-cell RNA sequencing was performed to dissect the crosstalk among B cell and T cell subsets and the underlying molecule and pathway signatures on longitudinal blood samples from IFN-α-treated CHB patients. RESULTS B cell depletion impaired the functional T cell subsets, including HBV-specific CD8+ T cells, and engendered a delayed HBV clearance. IFN-α treatment boosted the response of HBV-specific CD8+ T cells, whereas such effects disappeared in B cell-deficient mice. The underlying mechanisms were associated with IFN-α-reinforced connections of B cells toward T cells as mediated by the antigen presentation and costimulatory functions in B cells. CONCLUSION IFN-α orchestrates protective HBV-specific cellular immunity in a B cell-dependent manner.
Collapse
Affiliation(s)
- Shihong Zhong
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qiong Li
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chunhua Wen
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yifan Li
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yang Zhou
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zihan Jin
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Guofu Ye
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yanda Zhao
- School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jinlin Hou
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Yongyin Li
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Libo Tang
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
4
|
Becker PD, Ratnasothy K, Sen M, Peng Q, Romano M, Bazoer J, Suvitra E, Stout A, Hylton SG, Dorling A, Lechler RI, Smyth LA, Lombardi G. B lymphocytes contribute to indirect pathway T cell sensitization via acquisition of extracellular vesicles. Am J Transplant 2021; 21:1415-1426. [PMID: 32483894 DOI: 10.1111/ajt.16088] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 05/15/2020] [Accepted: 05/21/2020] [Indexed: 01/25/2023]
Abstract
B cells have been implicated in transplant rejection via antibody-mediated mechanisms and more recently by presenting donor antigens to T cells. We have shown in patients with chronic antibody-mediated rejection that B cells control the indirect T cell alloresponses. To understand more about the role of B cells as antigen-presenting cells for CD4+ T cell with indirect allospecificity, B cells were depleted in C57BL/6 mice, using an anti-CD20 antibody, prior to receiving MHC class I-mismatched (Kd ) skin. The absence of B cells at the time of transplantation prolonged skin graft survival. To study the mechanisms behind this observation, T cells with indirect allospecificity were transferred in mice receiving a Kd skin transplant. T cell proliferation was markedly inhibited in the absence of recipient B cells, suggesting that B cells contribute to indirect pathway sensitization. Furthermore, we have shown that a possible way in which B cells present alloantigens is via acquisition of MHC-peptide complexes. Finally, we demonstrate that the addition of B cell depletion to the transfer of regulatory T cells (Tregs) with indirect alloresponse further prolonged skin graft survival. This study supports an important role for B cells in indirect T cell priming and further emphasizes the advantage of combination therapies in prolonging transplant survival.
Collapse
Affiliation(s)
- Pablo D Becker
- MRC Centre for Transplantation, Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, King's College London, London, SE1 9RT, UK
| | - Kulachelvy Ratnasothy
- MRC Centre for Transplantation, Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, King's College London, London, SE1 9RT, UK
| | - Monica Sen
- MRC Centre for Transplantation, Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, King's College London, London, SE1 9RT, UK.,School of Health, Sports and Biosciences, University of East London, London, UK
| | - Qi Peng
- MRC Centre for Transplantation, Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, King's College London, London, SE1 9RT, UK
| | - Marco Romano
- MRC Centre for Transplantation, Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, King's College London, London, SE1 9RT, UK
| | - Jordan Bazoer
- School of Health, Sports and Biosciences, University of East London, London, UK
| | - Erik Suvitra
- School of Health, Sports and Biosciences, University of East London, London, UK
| | - Anas Stout
- School of Health, Sports and Biosciences, University of East London, London, UK
| | - Shannon G Hylton
- School of Health, Sports and Biosciences, University of East London, London, UK
| | - Anthony Dorling
- MRC Centre for Transplantation, Department of Inflammation Biology, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | - Robert I Lechler
- MRC Centre for Transplantation, Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, King's College London, London, SE1 9RT, UK
| | - Lesley A Smyth
- MRC Centre for Transplantation, Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, King's College London, London, SE1 9RT, UK.,School of Health, Sports and Biosciences, University of East London, London, UK
| | - Giovanna Lombardi
- MRC Centre for Transplantation, Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, King's College London, London, SE1 9RT, UK
| |
Collapse
|
5
|
Suprunenko T, Hofer MJ. Complexities of Type I Interferon Biology: Lessons from LCMV. Viruses 2019; 11:v11020172. [PMID: 30791575 PMCID: PMC6409748 DOI: 10.3390/v11020172] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 02/17/2019] [Accepted: 02/18/2019] [Indexed: 12/11/2022] Open
Abstract
Over the past decades, infection of mice with lymphocytic choriomeningitis virus (LCMV) has provided an invaluable insight into our understanding of immune responses to viruses. In particular, this model has clarified the central roles that type I interferons play in initiating and regulating host responses. The use of different strains of LCMV and routes of infection has allowed us to understand how type I interferons are critical in controlling virus replication and fostering effective antiviral immunity, but also how they promote virus persistence and functional exhaustion of the immune response. Accordingly, these discoveries have formed the foundation for the development of novel treatments for acute and chronic viral infections and even extend into the management of malignant tumors. Here we review the fundamental insights into type I interferon biology gained using LCMV as a model and how the diversity of LCMV strains, dose, and route of administration have been used to dissect the molecular mechanisms underpinning acute versus persistent infection. We also identify gaps in the knowledge regarding LCMV regulation of antiviral immunity. Due to its unique properties, LCMV will continue to remain a vital part of the immunologists' toolbox.
Collapse
Affiliation(s)
- Tamara Suprunenko
- School of Life and Environmental Sciences, the Marie Bashir Institute for Infectious Diseases and Biosecurity, Charles Perkins Centre, and the Bosch Institute, The University of Sydney, Sydney, NSW 2006, Australia.
| | - Markus J Hofer
- School of Life and Environmental Sciences, the Marie Bashir Institute for Infectious Diseases and Biosecurity, Charles Perkins Centre, and the Bosch Institute, The University of Sydney, Sydney, NSW 2006, Australia.
| |
Collapse
|
6
|
Xu H, Andersson AM, Ragonnaud E, Boilesen D, Tolver A, Jensen BAH, Blanchard JL, Nicosia A, Folgori A, Colloca S, Cortese R, Thomsen AR, Christensen JP, Veazey RS, Holst PJ. Mucosal Vaccination with Heterologous Viral Vectored Vaccine Targeting Subdominant SIV Accessory Antigens Strongly Inhibits Early Viral Replication. EBioMedicine 2017; 18:204-215. [PMID: 28302457 PMCID: PMC5405164 DOI: 10.1016/j.ebiom.2017.03.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 02/17/2017] [Accepted: 03/02/2017] [Indexed: 12/19/2022] Open
Abstract
Conventional HIV T cell vaccine strategies have not been successful in containing acute peak viremia, nor in providing long-term control. We immunized rhesus macaques intramuscularly and rectally using a heterologous adenovirus vectored SIV vaccine regimen encoding normally weakly immunogenic tat, vif, rev and vpr antigens fused to the MHC class II associated invariant chain. Immunizations induced broad T cell responses in all vaccinees. Following up to 10 repeated low-dose intrarectal challenges, vaccinees suppressed early viral replication (P = 0.01) and prevented the peak viremia in 5/6 animals. Despite consistently undetectable viremia in 2 out of 6 vaccinees, all animals showed evidence of infection induced immune responses indicating that infection had taken place. Vaccinees, with and without detectable viremia better preserved their rectal CD4 + T cell population and had reduced immune hyperactivation as measured by naïve T cell depletion, Ki-67 and PD-1 expression on T cells. These results indicate that vaccination towards SIV accessory antigens vaccine can provide a level of acute control of SIV replication with a suggestion of beneficial immunological consequences in infected animals of unknown long-term significance. In conclusion, our studies demonstrate that a vaccine encoding subdominant antigens not normally associated with virus control can exert a significant impact on acute peak viremia. Mucosal heterologousvirus-vectored vaccine used with MHC class II associated invariant chain linked SIV accessory antigens Mucosal vaccination targeting subdominant antigens delay SIV mac251 replication in rhesus macaques. Longterm reduction of immune hyperactivation following SIV infection of vaccinated macaques.
Mucosal immunization is used with heterologous viral vectors and a genetic adjuvant to raise responses against poorly immunogenic SIV antigens. Following repeated low-dose challenge we observed delayed establishment of chronic phase viremia and reduced immune hyperactivation 6 months after established infection. Vaccination was found to strongly reduce viremia at early, but not late time points, after detected infection and in 2 out of 6 animals infection could only observed as virus induced T cell responses. Subdominant antigen vaccines may thus be used to delay SIV mac251 infection and can enable control of chronic viremia in a minority of cases.
Collapse
Affiliation(s)
- Huanbin Xu
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, LA 70433, USA
| | - Anne-Marie Andersson
- Center for Medical Parasitology, Department of Immunology and Microbiology, University of Copenhagen, 1014, Denmark
| | - Emeline Ragonnaud
- Center for Medical Parasitology, Department of Immunology and Microbiology, University of Copenhagen, 1014, Denmark
| | - Ditte Boilesen
- Center for Medical Parasitology, Department of Immunology and Microbiology, University of Copenhagen, 1014, Denmark
| | - Anders Tolver
- Department of Mathematical Sciences, University of Copenhagen, 2100, Denmark
| | | | - James L Blanchard
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, LA 70433, USA
| | - Alfredo Nicosia
- ReiThera, viale Città d'Europa 679, 00144 Rome, Italy; CEINGE, via Gaetano Salvatore 486, 80145 Naples, Italy; Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, via S. Pansini 5, 80131 Naples, Italy
| | | | | | | | | | | | - Ronald S Veazey
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, LA 70433, USA
| | - Peter Johannes Holst
- Center for Medical Parasitology, Department of Immunology and Microbiology, University of Copenhagen, 1014, Denmark.
| |
Collapse
|
7
|
Hilpert C, Sitte S, Matthies A, Voehringer D. Dendritic Cells Are Dispensable for T Cell Priming and Control of Acute Lymphocytic Choriomeningitis Virus Infection. THE JOURNAL OF IMMUNOLOGY 2016; 197:2780-6. [PMID: 27549169 DOI: 10.4049/jimmunol.1502582] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 07/22/2016] [Indexed: 11/19/2022]
Abstract
Dendritic cells (DCs) are considered to be the major APCs with potent activity for priming of naive CD4 and CD8 T cells. However, T cell priming can also be achieved by other APCs including macrophages, B cells, or even nonhematopoietic cell types. Systemic low-dose infection of mice with lymphocytic choriomeningitis virus (LCMV) results in massive expansion of virus-specific CD4 and CD8 T cells. To determine the role of DCs as APCs and source of type I IFNs in this infection model, we used ΔDC mice in which DCs are constitutively ablated because of expression of the diphtheria toxin α subunit within developing DCs. ΔDC mice showed lower serum concentrations of IFN-β and IL-12p40, but normal IFN-α levels during the first days postinfection. No differences were found for proliferation of transferred TCR-transgenic cells during the early phase of infection, suggesting that T cell priming occurred with the same efficiency in wild-type and ΔDC mice. Expansion and cytokine expression of endogenous LCMV-specific T cells was comparable between wild-type and ΔDC mice during primary infection and upon rechallenge of memory mice. In both strains of infected mice the viral load was reduced below the limit of detection with the same kinetic. Further, germinal center formation and LCMV-specific Ab responses were not impaired in ΔDC mice. This indicates that DCs are dispensable as APCs for protective immunity against LCMV infection.
Collapse
Affiliation(s)
- Cornelia Hilpert
- Department of Infection Biology, University Hospital Erlangen and Friedrich Alexander University Erlangen-Nuremberg, 91054 Erlangen, Germany
| | - Selina Sitte
- Department of Infection Biology, University Hospital Erlangen and Friedrich Alexander University Erlangen-Nuremberg, 91054 Erlangen, Germany
| | - Alexander Matthies
- Department of Infection Biology, University Hospital Erlangen and Friedrich Alexander University Erlangen-Nuremberg, 91054 Erlangen, Germany
| | - David Voehringer
- Department of Infection Biology, University Hospital Erlangen and Friedrich Alexander University Erlangen-Nuremberg, 91054 Erlangen, Germany
| |
Collapse
|
8
|
Marino J, Paster JT, Trowell A, Maxwell L, Briggs KH, Crosby Bertorini P, Benichou G. B Cell Depletion With an Anti-CD20 Antibody Enhances Alloreactive Memory T Cell Responses After Transplantation. Am J Transplant 2016; 16:672-8. [PMID: 26552037 PMCID: PMC4733428 DOI: 10.1111/ajt.13483] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Revised: 07/29/2015] [Accepted: 07/31/2015] [Indexed: 01/25/2023]
Abstract
Alloreactive memory T cells mediate accelerated allograft rejection and transplant tolerance resistance. Recent studies have shown that B cell deficient-μMT mice fail to mount donor-specific memory T cell responses after transplantation. At the same time, other studies showed that pretransplant B cell depletion using rituximab (IgG1 anti-CD20 mAb) combined with cyclosporine A promoted the survival of islet allografts in monkeys. In this study, we investigated the effect of anti-CD20 antibody-mediated B cell depletion on the memory T cell alloresponse in mice. Wild-type and anti-OVA TCR transgenic mice were treated with an IgG2a anti-CD20 monoclonal antibody, which depleted nearly all B cells in the peripheral blood and secondary lymphoid organs but spared some B cells in the bone marrow. B cell depletion did not affect the direct alloresponse but resulted in a marked increase of indirect alloresponse after skin transplantation of naïve mice. Furthermore, in allosensitized mice, anti-CD20 mAb treatment enhanced the reactivation of allospecific memory T cells and accelerated second set rejection of skin allografts. This suggests that the effect of anti-CD20 antibodies on alloimmunity and allograft rejection might vary upon the nature of the antibodies as well as the circumstances under which they are delivered.
Collapse
|
9
|
Immunogenicity of HLA Class I and II Double Restricted Influenza A-Derived Peptides. PLoS One 2016; 11:e0145629. [PMID: 26731261 PMCID: PMC4701504 DOI: 10.1371/journal.pone.0145629] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 12/06/2015] [Indexed: 01/10/2023] Open
Abstract
The aim of the present study was to identify influenza A-derived peptides which bind to both HLA class I and -II molecules and by immunization lead to both HLA class I and class II restricted immune responses. Eight influenza A-derived 9-11mer peptides with simultaneous binding to both HLA-A*02:01 and HLA-DRB1*01:01 molecules were identified by bioinformatics and biochemical technology. Immunization of transgenic HLA-A*02:01/HLA-DRB1*01:01 mice with four of these double binding peptides gave rise to both HLA class I and class II restricted responses by CD8 and CD4 T cells, respectively, whereas four of the double binding peptides did result in HLA-A*02:01 restricted responses only. According to their cytokine profile, the CD4 T cell responses were of the Th2 type. In influenza infected mice, we were unable to detect natural processing in vivo of the double restricted peptides and in line with this, peptide vaccination did not decrease virus titres in the lungs of intranasally influenza challenged mice. Our data show that HLA class I and class II double binding peptides can be identified by bioinformatics and biochemical technology. By immunization, double binding peptides can give rise to both HLA class I and class I restricted responses, a quality which might be of potential interest for peptide-based vaccine development.
Collapse
|
10
|
Lapke N, Tartz S, Lee KH, Jacobs T. The application of anti-Toso antibody enhances CD8(+) T cell responses in experimental malaria vaccination and disease. Vaccine 2015; 33:6763-70. [PMID: 26597034 DOI: 10.1016/j.vaccine.2015.10.065] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Revised: 09/28/2015] [Accepted: 10/12/2015] [Indexed: 10/22/2022]
Abstract
Toso is a molecule highly expressed on B cells. It influences their survival and was identified as an IgM binding molecule. B cells and natural antibodies play a role in vaccination-induced CD8(+) T cell responses. We investigated the impact of an anti-Toso antibody on vaccination efficiency in a malaria vaccination model. In this model, CD8(+) T cells exert antiparasitic functions on infected hepatocytes in the liver stage of the disease. In vaccinated anti-Toso treated mice, more antigen-specific CD8(+) T cells were induced than in control mice and after infection with Plasmodium berghei ANKA (PbA) sporozoites, the liver parasite burden was lower. In B cell deficient mice, the anti-Toso antibody did not stimulate the CD8(+) T cell response, indicating that B cells were mediating this effect. Furthermore, we analyzed the influence of anti-Toso treatment on non-vaccinated mice in the PbA infection model, in which CD8(+) T cells cause brain pathology. Anti-Toso treatment increased cerebral pathology and the accumulation of CD8(+) T cells in the brain. Thus, anti-Toso treatment enhanced the CD8(+) T cell response against PbA in a vaccination and in an infection model. Our findings indicate that Toso may be a novel target to boost vaccine-induced CD8(+) T cell responses.
Collapse
Affiliation(s)
- Nina Lapke
- Bernhard Nocht Institute for Tropical Medicine, Bernhard-Nocht-Straße 74, 20359 Hamburg, Germany
| | - Susanne Tartz
- Bernhard Nocht Institute for Tropical Medicine, Bernhard-Nocht-Straße 74, 20359 Hamburg, Germany
| | - Kyeong-Hee Lee
- Institute for Clinical Chemistry and Inflammation Research, Hannover Medical School, Carl-Neuberg-Straße 1, 30625 Hannover, Germany
| | - Thomas Jacobs
- Bernhard Nocht Institute for Tropical Medicine, Bernhard-Nocht-Straße 74, 20359 Hamburg, Germany.
| |
Collapse
|
11
|
Guan H, Lan Y, Wan Y, Wang Q, Wang C, Xu L, Chen Y, Liu W, Zhang X, Li Y, Gu Y, Wang Z, Xie F. PD-L1 mediated the differentiation of tumor-infiltrating CD19 + B lymphocytes and T cells in Invasive breast cancer. Oncoimmunology 2015; 5:e1075112. [PMID: 27057444 DOI: 10.1080/2162402x.2015.1075112] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Revised: 07/14/2015] [Accepted: 07/15/2015] [Indexed: 10/23/2022] Open
Abstract
Accumulating evidence suggests that B cells play important roles in inhibiting the immune response in autoimmune disorders and human tumors as well as murine tumor models. In an effort to explore the role of B cells in human breast cancer etiology, we examined the presence of CD19+ B lymphocytes in 134 cases of invasive breast carcinoma (IBCa) and 31 breast fibroadenoma, and assessed its relationship with PD-L1 (programmed death-ligand 1) expression in breast cancer. We found that the density of CD19+ B lymphocytes was higher in IBCa compared with fibroadenoma, and significantly associated with increasing tumor grade, negative estrogen status. Similar findings were observed for the expression of IL-10 in IBCa. Meanwhile, CD19+ B lymphocytes were shown to be highly coincident with PD-L1 and IL-10 in IBCa. We further demonstrated that CD19+ B cells can differentiate into CD19+CD24+CD38+ B cells when co-cultured with PD-L1hi MDA-MB231 cells. In addition, the percentage of CD19+CD24+CD38+ B cells was higher in breast tissue and peripheral blood cells of IBCa patients than that of benign tumor and health individuals. And CD19+CD24+CD38+ B cells were found to be IL-10 secreting B cells. Finally, we showed that CD19+ B cells from IBCa patients but not healthy individuals induced formation of CD4+CD25+Foxp3+ T cells when co-cultured with T cells from IBCa patients and healthy subjects (80.4% and 30.8% respectively). The induction of CD4+CD25+Foxp3+ T cells by CD19+ B cells was further shown to be mediated by PD-L1. Together, these results are suggestive of a role for CD19+ B lymphocytes in immune suppression and tumor evasion via PD-L1 in breast cancer.
Collapse
Affiliation(s)
- Honggeng Guan
- Department of General Surgery, The First Affiliated Hospital of Soochow University , Suzhou, P.R. China
| | - Yang Lan
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, P.R. China; Department of General Surgery, Wuxi Third People's Hospital, Wuxi, P. R. China
| | - Yuqiu Wan
- Department of General Surgery, The First Affiliated Hospital of Soochow University , Suzhou, P.R. China
| | - Qin Wang
- Department Immunology, Medical College of Soochow University , Suzhou, P. R. China
| | - Cheng Wang
- The Ultrasonagraphy Center of the Second Affiliated Hospital of Soochow University , Suzhou, P. R. China
| | - Longjiang Xu
- The Second Affiliated Hospital of Soochow University , Suzhou, P. R. China
| | - Yongjing Chen
- Department Immunology, Medical College of Soochow University , Suzhou, P. R. China
| | - Wenting Liu
- Department of Pathology, Medical College of Soochow University , Suzhou, P. R. China
| | - Xueguang Zhang
- Jiangsu stem cell lab center , Jiangsu, Suzhou, P. R. China
| | - Yecheng Li
- The Second Affiliated Hospital of Soochow University , Suzhou, P. R. China
| | - Yongping Gu
- Department of Pathology, Medical College of Soochow University , Suzhou, P. R. China
| | - Zemin Wang
- Department of Environmental Health, School of Public Health, Indiana University , Bloomington, IN, USA
| | - Fang Xie
- Department of Pathology, Medical College of Soochow University , Suzhou, P. R. China
| |
Collapse
|
12
|
Holst PJ, Jensen BAH, Ragonnaud E, Thomsen AR, Christensen JP. Targeting of non-dominant antigens as a vaccine strategy to broaden T-cell responses during chronic viral infection. PLoS One 2015; 10:e0117242. [PMID: 25679375 PMCID: PMC4334508 DOI: 10.1371/journal.pone.0117242] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Accepted: 12/22/2014] [Indexed: 12/26/2022] Open
Abstract
In this study, we compared adenoviral vaccine vectors with the capacity to induce equally potent immune responses against non-dominant and immunodominant epitopes of murine lymphocytic choriomeningitis virus (LCMV). Our results demonstrate that vaccination targeting non-dominant epitopes facilitates potent virus-induced T-cell responses against immunodominant epitopes during subsequent challenge with highly invasive virus. In contrast, when an immunodominant epitope was included in the vaccine, the T-cell response associated with viral challenge remained focussed on that epitope. Early after challenge with live virus, the CD8+ T cells specific for vaccine-encoded epitopes, displayed a phenotype typically associated with prolonged/persistent antigenic stimulation marked by high levels of KLRG-1, as compared to T cells reacting to epitopes not included in the vaccine. Notably, this association was lost over time in T cells specific for the dominant T cell epitopes, and these cells were fully capable of expanding in response to a new viral challenge. Overall, our data suggests a potential for broadening of the antiviral CD8+ T-cell response by selecting non-dominant antigens to be targeted by vaccination. In addition, our findings suggest that prior adenoviral vaccination is not likely to negatively impact the long-term and protective immune response induced and maintained by a vaccine-attenuated chronic viral infection.
Collapse
Affiliation(s)
- Peter J. Holst
- Department of International Health, Immunology and Microbiology, University of Copenhagen, The Panum Institute, Copenhagen, Denmark
- Centre for Medical Parasitology, Department of International Health, Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- * E-mail:
| | - Benjamin A. H. Jensen
- Department of International Health, Immunology and Microbiology, University of Copenhagen, The Panum Institute, Copenhagen, Denmark
| | - Emeline Ragonnaud
- Centre for Medical Parasitology, Department of International Health, Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Allan R. Thomsen
- Department of International Health, Immunology and Microbiology, University of Copenhagen, The Panum Institute, Copenhagen, Denmark
| | - Jan P. Christensen
- Department of International Health, Immunology and Microbiology, University of Copenhagen, The Panum Institute, Copenhagen, Denmark
| |
Collapse
|
13
|
Bassi MR, Kongsgaard M, Steffensen MA, Fenger C, Rasmussen M, Skjødt K, Finsen B, Stryhn A, Buus S, Christensen JP, Thomsen AR. CD8+ T cells complement antibodies in protecting against yellow fever virus. THE JOURNAL OF IMMUNOLOGY 2014; 194:1141-53. [PMID: 25539816 DOI: 10.4049/jimmunol.1402605] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The attenuated yellow fever (YF) vaccine (YF-17D) was developed in the 1930s, yet little is known about the protective mechanisms underlying its efficiency. In this study, we analyzed the relative contribution of cell-mediated and humoral immunity to the vaccine-induced protection in a murine model of YF-17D infection. Using different strains of knockout mice, we found that CD4(+) T cells, B cells, and Abs are required for full clinical protection of vaccinated mice, whereas CD8(+) T cells are dispensable for long-term survival after intracerebral challenge. However, by analyzing the immune response inside the infected CNS, we observed an accelerated T cell influx into the brain after intracerebral challenge of vaccinated mice, and this T cell recruitment correlated with improved virus control in the brain. Using mice deficient in B cells we found that, in the absence of Abs, YF vaccination can still induce some antiviral protection, and in vivo depletion of CD8(+) T cells from these animals revealed a pivotal role for CD8(+) T cells in controlling virus replication in the absence of a humoral response. Finally, we demonstrated that effector CD8(+) T cells also contribute to viral control in the presence of circulating YF-specific Abs. To our knowledge, this is the first time that YF-specific CD8(+) T cells have been demonstrated to possess antiviral activity in vivo.
Collapse
Affiliation(s)
- Maria R Bassi
- Department of International Health, Immunology and Microbiology, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Michael Kongsgaard
- Department of International Health, Immunology and Microbiology, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Maria A Steffensen
- Department of International Health, Immunology and Microbiology, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Christina Fenger
- Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, DK-5000 Odense, Denmark; and
| | - Michael Rasmussen
- Department of International Health, Immunology and Microbiology, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Karsten Skjødt
- Department of Cancer and Inflammation, Institute for Molecular Medicine, University of Southern Denmark, DK-5000 Odense, Denmark
| | - Bente Finsen
- Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, DK-5000 Odense, Denmark; and
| | - Anette Stryhn
- Department of International Health, Immunology and Microbiology, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Søren Buus
- Department of International Health, Immunology and Microbiology, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Jan P Christensen
- Department of International Health, Immunology and Microbiology, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Allan R Thomsen
- Department of International Health, Immunology and Microbiology, University of Copenhagen, DK-2200 Copenhagen N, Denmark;
| |
Collapse
|
14
|
León B, Ballesteros-Tato A, Randall TD, Lund FE. Prolonged antigen presentation by immune complex-binding dendritic cells programs the proliferative capacity of memory CD8 T cells. ACTA ACUST UNITED AC 2014; 211:1637-55. [PMID: 25002751 PMCID: PMC4113940 DOI: 10.1084/jem.20131692] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
Antibodies can regulate the quality and functionality of a subset of antiviral CD8+ T cell memory responses to influenza by promoting sustained DC antigen presentation during the contraction phase of primary responses. The commitment of naive CD8 T cells to effector or memory cell fates can occur after a single day of antigenic stimulation even though virus-derived antigens (Ags) are still presented by DCs long after acute infection is resolved. However, the effects of extended Ag presentation on CD8 T cells are undefined and the mechanisms that regulate prolonged Ag presentation are unknown. We showed that the sustained presentation of two different epitopes from influenza virus by DCs prevented the premature contraction of the primary virus-specific CD8 T cell response. Although prolonged Ag presentation did not alter the number of memory CD8 T cells that developed, it was essential for programming the capacity of these cells to proliferate, produce cytokines, and protect the host after secondary challenge. Importantly, prolonged Ag presentation by DCs was dependent on virus-specific, isotype-switched antibodies (Abs) that facilitated the capture and cross-presentation of viral Ags by FcγR-expressing DCs. Collectively, our results demonstrate that B cells and Abs can regulate the quality and functionality of a subset of antiviral CD8 T cell memory responses and do so by promoting sustained Ag presentation by DCs during the contraction phase of the primary T cell response.
Collapse
Affiliation(s)
- Beatriz León
- Division of Allergy, Immunology, and Rheumatology, Department of Medicine, University of Rochester Medical Center, Rochester, NY 14642
| | - André Ballesteros-Tato
- Division of Allergy, Immunology, and Rheumatology, Department of Medicine, University of Rochester Medical Center, Rochester, NY 14642
| | - Troy D Randall
- Division of Allergy, Immunology, and Rheumatology, Department of Medicine, University of Rochester Medical Center, Rochester, NY 14642
| | - Frances E Lund
- Division of Allergy, Immunology, and Rheumatology, Department of Medicine, University of Rochester Medical Center, Rochester, NY 14642
| |
Collapse
|
15
|
Misumi I, Whitmire JK. B cell depletion curtails CD4+ T cell memory and reduces protection against disseminating virus infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2014; 192:1597-608. [PMID: 24453250 PMCID: PMC3925510 DOI: 10.4049/jimmunol.1302661] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Dynamic interactions between CD4(+) T cells and B cells are needed for humoral immunity and CD4(+) T cell memory. It is not known whether B cells are needed early on to induce the formation of memory precursor cells or are needed later to sustain memory cells. In this study, primary and memory CD4(+) T cells responses were followed in wild-type mice that were depleted of mature B cells by anti-CD20 before or different times after acute lymphocytic choriomeningitis virus infection. The Ab treatment led to a 1000-fold reduction in B cell number that lasted 6 wk. Primary virus-specific CD4(+) Th1 cells were generated in B cell-depleted mice; however, there was a decrease in the CD4(+)Ly6C(lo)Tbet(+) memory precursor population and a corresponding 4-fold reduction in CD4(+) memory cell number. Memory T cells showed impaired cytokine production when they formed without B cells. B cell depletion had no effect on established memory populations. During disseminating virus infection, B cell depletion led to sustained weight loss and functional exhaustion of CD4(+) and CD8(+) T cells, and prevented mice from resolving the infection. Thus, B cells contribute to the establishment and survival of memory CD4(+) T cells post-acute infection and play an essential role in immune protection against disseminating virus infection.
Collapse
Affiliation(s)
- Ichiro Misumi
- Department of Genetics, UNC-Chapel Hill School of Medicine, Chapel Hill, NC 27599
| | - Jason K. Whitmire
- Department of Genetics, UNC-Chapel Hill School of Medicine, Chapel Hill, NC 27599
- Department of Microbiology & Immunology, UNC-Chapel Hill School of Medicine, Chapel Hill, NC 27599
| |
Collapse
|
16
|
Pedersen SR, Sørensen MR, Buus S, Christensen JP, Thomsen AR. Comparison of vaccine-induced effector CD8 T cell responses directed against self- and non-self-tumor antigens: implications for cancer immunotherapy. THE JOURNAL OF IMMUNOLOGY 2013; 191:3955-67. [PMID: 24018273 DOI: 10.4049/jimmunol.1300555] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
It is generally accepted that CD8 T cells play a major role in tumor control, yet vaccination aimed at eliciting potent CD8 T cell responses are rarely efficient in clinical trials. To try and understand why this is so, we have generated potent adenoviral vectors encoding the endogenous tumor Ags (TA) tyrosinase-related protein-2 (TRP-2) and glycoprotein 100 (GP100) tethered to the invariant chain (Ii). Using these vectors, we sought to characterize the self-TA-specific CD8 T cell response and compare it to that induced against non-self-Ags expressed from a similar vector platform. Prophylactic vaccination with adenoviral vectors expressing either TRP-2 (Ad-Ii-TRP-2) or GP100 (Ad-Ii-GP100) had little or no effect on the growth of s.c. B16 melanomas, and only Ad-Ii-TRP-2 was able to induce a marginal reduction of B16 lung metastasis. In contrast, vaccination with a similar vector construct expressing a foreign (viral) TA induced efficient tumor control. Analyzing the self-TA-specific CD8 T cells, we observed that these could be activated to produce IFN-γ and TNF-α. In addition, surface expression of phenotypic markers and inhibitory receptors, as well as in vivo cytotoxicity and degranulation capacity matched that of non-self-Ag-specific CD8 T cells. However, the CD8 T cells specific for self-TAs had a lower functional avidity, and this impacted on their in vivo performance. On the basis of these results and a low expression of the targeted TA epitopes on the tumor cells, we suggest that low avidity of the self-TA-specific CD8 T cells may represent a major obstacle for efficient immunotherapy of cancer.
Collapse
Affiliation(s)
- Sara R Pedersen
- Department of International Health, Immunology and Microbiology, University of Copenhagen, DK-2200 Copenhagen North, Denmark
| | | | | | | | | |
Collapse
|
17
|
Straub T, Schweier O, Bruns M, Nimmerjahn F, Waisman A, Pircher H. Nucleoprotein-specific nonneutralizing antibodies speed up LCMV elimination independently of complement and FcγR. Eur J Immunol 2013; 43:2338-48. [PMID: 23749409 DOI: 10.1002/eji.201343565] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Revised: 05/21/2013] [Accepted: 06/04/2013] [Indexed: 12/12/2022]
Abstract
CD8(+) T cells have an essential role in controlling lymphocytic choriomeningitis virus (LCMV) infection in mice. Here, we examined the contribution of humoral immunity, including nonneutralizing antibodies (Abs), in this infection induced by low virus inoculation doses. Mice with impaired humoral immunity readily terminated infection with the slowly replicating LCMV strain Armstrong but showed delayed virus elimination after inoculation with the faster replicating LCMV strain WE and failed to clear the rapidly replicating LCMV strain Docile, which is in contrast to the results obtained with wild-type mice. Thus, the requirement for adaptive humoral immunity to control the infection was dependent on the replication speed of the LCMV strains used. Ab transfers further showed that LCMV-specific IgG Abs isolated from LCMV immune serum accelerated virus elimination. These Abs were mainly directed against the viral nucleoprotein (NP) and completely lacked virus neutralizing activity. Moreover, mAbs specific for the LCMV NP were also able to decrease viral titers after transfer into infected hosts. Intriguingly, neither C3 nor Fcγ receptors were required for the antiviral activity of the transferred Abs. In conclusion, our study suggests that rapidly generated nonneutralizing Abs specific for the viral NP speed up virus elimination and thereby may counteract T-cell exhaustion.
Collapse
Affiliation(s)
- Tobias Straub
- Department of Immunology, Institute of Medical Microbiology and Hygiene, University of Freiburg, Freiburg, Germany; Faculty of Biology, University of Freiburg, Freiburg, Germany
| | | | | | | | | | | |
Collapse
|
18
|
Pillai S, Mattoo H, Cariappa A. B cells and autoimmunity. Curr Opin Immunol 2011; 23:721-31. [PMID: 22119110 DOI: 10.1016/j.coi.2011.10.007] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2011] [Revised: 10/17/2011] [Accepted: 10/27/2011] [Indexed: 12/21/2022]
Abstract
There is a growing appreciation for the role for B cells in autoimmune disorders in which inflammation is driven by T cells, in addition to the well-established role for B cells in autoimmune disorders characterized by pathogenic auto-antibodies. Current information on tolerance checkpoints in B cells, B cell depletion, BAFF blockade, regulatory B cells and clonal ignorance mediated by the SIAE/Siglec pathway will be reviewed.
Collapse
Affiliation(s)
- Shiv Pillai
- Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, United States.
| | | | | |
Collapse
|
19
|
Holst PJ, Christensen JP, Thomsen AR. Vaccination against lymphocytic choriomeningitis virus infection in MHC class II-deficient mice. THE JOURNAL OF IMMUNOLOGY 2011; 186:3997-4007. [PMID: 21357263 DOI: 10.4049/jimmunol.1001251] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The impact of prophylactic vaccination against acute and chronic infection in a Th-deficient host has not been adequately addressed because of difficulties in generating protective immunity in the absence of CD4(+) T cell help. In this study, we demonstrated that a broad CD8(+) T cell immune response could be elicited in MHC class II-deficient mice by vaccination with adenovirus encoding lymphocytic choriomeningitis virus (LCMV) glycoprotein tethered to MHC class II-associated invariant chain. Moreover, the response induced conferred significant cytolytic CD8(+) T cell-mediated protection against challenge with a high dose of the invasive clone 13 strain of LCMV. In contrast, vaccination with adenovirus encoding unlinked LCMV glycoprotein induced weak virus control in the absence of CD4(+) T cells, and mice may die of increased immunopathology associated with incomplete protection. Acute mortality was not observed in any vaccinated mice following infection with the less-invasive Traub strain. However, LCMV Traub infection caused accelerated late mortality in unvaccinated MHC class II-deficient mice; in this case, we observed a strong trend toward delayed mortality in vaccinated mice, irrespective of the nature of the vaccine. These results indicated that optimized vaccination may lead to efficient protection against acute viral infection, even in Th-deficient individuals, but that the duration of such immunity is limited. Nevertheless, for select immunodeficiencies in which CD4(+) T cell deficiency is incomplete or transient, these results are very encouraging.
Collapse
Affiliation(s)
- Peter Johannes Holst
- Department of International Health, Immunology and Microbiology, University of Copenhagen, Copenhagen DK-2200, Denmark
| | | | | |
Collapse
|
20
|
Ng YH, Oberbarnscheidt MH, Chandramoorthy HCK, Hoffman R, Chalasani G. B cells help alloreactive T cells differentiate into memory T cells. Am J Transplant 2010; 10:1970-80. [PMID: 20883532 PMCID: PMC2956128 DOI: 10.1111/j.1600-6143.2010.03223.x] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
B cells are recognized as effector cells in allograft rejection that are dependent upon T cell help to produce alloantibodies causing graft injury. It is not known if B cells can also help T cells differentiate into memory cells in the alloimmune response. We found that in B-cell-deficient hosts, differentiation of alloreactive T cells into effectors was intact whereas their development into memory T cells was impaired. To test if B cell help for T cells was required for their continued differentiation into memory T cells, activated T cells were sorted from alloimmunized mice and transferred either with or without B cells into naïve adoptive hosts. Activated T cells cotransferred with B cells gave rise to more memory T cells than those transferred without B cells and upon recall, mediated accelerated rejection of skin allografts. Cotransfer of B cells led to increased memory T cells by enhancing activated CD4 T-cell proliferation and activated CD8 T-cell survival. These results indicate that B cells help alloreactive T-cell differentiation, proliferation and survival to generate optimal numbers of functional memory T cells.
Collapse
Affiliation(s)
- Yue-Harn Ng
- Departments of Medicine (Renal-Electrolyte) and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261,Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| | - Martin H. Oberbarnscheidt
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| | | | - Rosemary Hoffman
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| | - Geetha Chalasani
- Departments of Medicine (Renal-Electrolyte) and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261,Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261,Address correspondence and reprint requests to: Dr. Geetha Chalasani, University of Pittsburgh School of Medicine, BST W1554, 200 Lothrop Street, Pittsburgh, PA 15261. Phone: (412) 383-5924; Fax: (412) 383-9990;
| |
Collapse
|
21
|
Sorensen MR, Holst PJ, Steffensen MA, Christensen JP, Thomsen AR. Adenoviral vaccination combined with CD40 stimulation and CTLA-4 blockage can lead to complete tumor regression in a murine melanoma model. Vaccine 2010; 28:6757-64. [PMID: 20682365 DOI: 10.1016/j.vaccine.2010.07.066] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2010] [Revised: 07/06/2010] [Accepted: 07/21/2010] [Indexed: 10/24/2022]
Abstract
Therapeutic vaccination with replication deficient adenovirus expressing a viral antigen linked to invariant chain was recently found to markedly delay the growth of B16.F10 melanomas expressing the same antigen; however, complete regression of the tumors was never observed. Here we show that the delay in tumor growth can be converted to complete regression and long-term survival in 30-40% of the mice by a booster vaccination plus combinational treatment with agonistic anti-CD40 monoclonal antibodies (mAb) and anti-CTLA-4 mAb. Regarding the mechanism underlying the improved clinical effect, analysis of the tumor-specific response revealed a significantly prolonged tumor-specific CD8 T cell response in spleens of the mice receiving the combinational treatment compared with mice receiving either treatment individually. Matching this, CD8 T cell depletion completely prevented tumor control. These results indicate that even with a strong tumor vaccine candidate, combinatorial treatment may be required to obtain clinically relevant results.
Collapse
Affiliation(s)
- Maria R Sorensen
- Institute of International Health, Immunology, and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | | |
Collapse
|
22
|
Grujic M, Bartholdy C, Remy M, Pinschewer DD, Christensen JP, Thomsen AR. The Role of CD80/CD86 in Generation and Maintenance of Functional Virus-Specific CD8+ T Cells in Mice Infected with Lymphocytic Choriomeningitis Virus. THE JOURNAL OF IMMUNOLOGY 2010; 185:1730-43. [DOI: 10.4049/jimmunol.0903894] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
23
|
Lund FE, Randall TD. Effector and regulatory B cells: modulators of CD4+ T cell immunity. Nat Rev Immunol 2010; 10:236-47. [PMID: 20224569 DOI: 10.1038/nri2729] [Citation(s) in RCA: 488] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
B cells are essential for humoral immunity, but the role that they have in regulating CD4(+) T cell responses remains controversial. However, new data showing that the transient depletion of B cells potently influences the induction, maintenance and reactivation of CD4(+) T cells, with the recent identification of antibody-independent functions of B cells, have reinvigorated interest in the many roles of B cells in both infectious and autoimmune diseases. In this Review, we discuss recent data showing how effector and regulatory B cells modulate CD4(+) T cell responses to pathogens and autoantigens.
Collapse
Affiliation(s)
- Frances E Lund
- Department of Medicine, University of Rochester Medical Center, New York 14642, USA.
| | | |
Collapse
|
24
|
Zarkhin V, Chalasani G, Sarwal MM. The yin and yang of B cells in graft rejection and tolerance. Transplant Rev (Orlando) 2010; 24:67-78. [PMID: 20149626 DOI: 10.1016/j.trre.2010.01.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Various lineages of B cells are being increasingly recognized as important players in the etiology and prognosis of both acute and chronic graft rejection. The role of immature, chronically activated B cells, as efficient antigen-presenting cells, supporting recalcitrant cell-mediated graft rejection and late lineage B cells driving humoral rejections, is being increasingly recognized. This review captures the recent literature on this subject and discusses the various roles of the B cell in renal graft rejection and conversely, also in graft tolerance, both in animal and human studies. In addition, novel therapies targeting specific B-cell lineages in graft rejection are also discussed, with a view to developing more targeted therapies for graft rejection.
Collapse
Affiliation(s)
- Valeriya Zarkhin
- Department of Pediatrics, Stanford University, Stanford, CA, USA.
| | | | | |
Collapse
|
25
|
Hoegh-Petersen M, Thomsen AR, Christensen JP, Holst PJ. Mucosal immunization with recombinant adenoviral vectors expressing murine gammaherpesvirus-68 genes M2 and M3 can reduce latent viral load. Vaccine 2009; 27:6723-30. [DOI: 10.1016/j.vaccine.2009.08.104] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2008] [Revised: 07/13/2009] [Accepted: 08/26/2009] [Indexed: 12/22/2022]
|
26
|
Sorensen MR, Holst PJ, Pircher H, Christensen JP, Thomsen AR. Vaccination with an adenoviral vector encoding the tumor antigen directly linked to invariant chain induces potent CD4+T-cell-independent CD8+T-cell-mediated tumor control. Eur J Immunol 2009; 39:2725-36. [DOI: 10.1002/eji.200939543] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
27
|
Impaired antibody response causes persistence of prototypic T cell-contained virus. PLoS Biol 2009; 7:e1000080. [PMID: 19355789 PMCID: PMC2672599 DOI: 10.1371/journal.pbio.1000080] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2008] [Accepted: 02/24/2009] [Indexed: 11/19/2022] Open
Abstract
CD8 T cells are recognized key players in control of persistent virus infections, but increasing evidence suggests that assistance from other immune mediators is also needed. Here, we investigated whether specific antibody responses contribute to control of lymphocytic choriomeningitis virus (LCMV), a prototypic mouse model of systemic persistent infection. Mice expressing transgenic B cell receptors of LCMV-unrelated specificity, and mice unable to produce soluble immunoglobulin M (IgM) exhibited protracted viremia or failed to resolve LCMV. Virus control depended on immunoglobulin class switch, but neither on complement cascades nor on Fc receptor gamma chain or Fc gamma receptor IIB. Cessation of viremia concurred with the emergence of viral envelope-specific antibodies, rather than with neutralizing serum activity, and even early nonneutralizing IgM impeded viral persistence. This important role for virus-specific antibodies may be similarly underappreciated in other primarily T cell-controlled infections such as HIV and hepatitis C virus, and we suggest this contribution of antibodies be given consideration in future strategies for vaccination and immunotherapy.
Collapse
|
28
|
Grujic M, Holst PJ, Christensen JP, Thomsen AR. Fusion of a viral antigen to invariant chain leads to augmented T-cell immunity and improved protection in gene-gun DNA-vaccinated mice. J Gen Virol 2009; 90:414-422. [DOI: 10.1099/vir.0.002105-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
It has recently been demonstrated that a recombinant replication-deficient human adenovirus 5 (Ad5) vector expressing lymphocytic choriomeningitis virus (LCMV) glycoprotein (GP) fused to the p31 invariant (Ii) chain confers broad, long-lasting T-cell immunity that completely protects C57BL/6 mice against lethal peripheral challenge. The current study questioned whether the same strategy, i.e. linkage of GP to an Ii chain, could be applied to a naked DNA vaccine. Following gene-gun immunization with the linked construct (DNA–IiGP), GP-specific CD4+ T cells could not be detected by flow cytometry. However, inclusion of the Ii chain augmented the priming of GP-specific CD8+ T cells directed towards both immunodominant (GP33–41) and subdominant (GP276–286 and GP92–101) epitopes, and vaccination with DNA–IiGP conferred significantly improved protection against systemic LCMV infection compared with the unlinked construct. In contrast, substantial protection against peripheral challenge was not observed. Additional experiments with T-cell subset-depleted or perforin-deficient mice revealed that virus control in vaccinated mice depends critically on cytotoxic CD8+ T cells. Finally, priming with the naked DNA vaccine was shown to augment the immune response raised by subsequent immunization with the Ad5 vector. In conclusion, this study showed that the immunoenhancing effect of Ii chain linkage is not limited to the Ad5 vector, but is also relevant with a DNA platform. Furthermore, given the fact that the Ii chain enhances the presentation of more than one epitope, this suggests that Ii-chain-based DNA vaccines may be promising candidates for various heterologous prime–boost regimes.
Collapse
Affiliation(s)
- Mirjana Grujic
- Institute of International Health, Immunology and Microbiology, University of Copenhagen, The Panum Institute, 3C Blegdamsvej, DK-2200, Copenhagen, Denmark
| | - Peter J. Holst
- Institute of International Health, Immunology and Microbiology, University of Copenhagen, The Panum Institute, 3C Blegdamsvej, DK-2200, Copenhagen, Denmark
| | - Jan P. Christensen
- Institute of International Health, Immunology and Microbiology, University of Copenhagen, The Panum Institute, 3C Blegdamsvej, DK-2200, Copenhagen, Denmark
| | - Allan R. Thomsen
- Institute of International Health, Immunology and Microbiology, University of Copenhagen, The Panum Institute, 3C Blegdamsvej, DK-2200, Copenhagen, Denmark
| |
Collapse
|
29
|
Lindell DM, Berlin AA, Schaller MA, Lukacs NW. B cell antigen presentation promotes Th2 responses and immunopathology during chronic allergic lung disease. PLoS One 2008; 3:e3129. [PMID: 18769622 PMCID: PMC2518863 DOI: 10.1371/journal.pone.0003129] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2008] [Accepted: 08/14/2008] [Indexed: 12/30/2022] Open
Abstract
Background The role of B cells in allergic asthma remains undefined. One mechanism by which B cells clearly contribute to allergic disease is via the production of specific immunoglobulin, and especially IgE. Cognate interactions with specific T cells result in T cell help for B cells, resulting in differentiation and immunoglobulin secretion. Proximal to (and required for) T cell-dependent immunoglobulin production, however, is antigen presentation by B cells. While interaction with T cells clearly has implications for B cell function and differentiation, this study investigated the role that B cells have in shaping the T cell response during chronic allergic lung disease. Methodology/Principal Findings In these studies, we used a clinically relevant mouse model of chronic allergic lung disease to study the role of B cells and B cell antigen presentation in this disease. In these studies we present several novel findings: 1) Lung B cells from chronically allergen challenged mice up-regulated MHC II and costimulatory molecules CD40, CD80 and CD86. 2) Using in vitro studies, B cells from the lungs of allergen challenged mice could present antigen to T cells, as assessed by T cell proliferation and the preferential production of Th2 cytokines. 3) Following chronic allergen challenge, the levels of Th2 cytokines IL-4 and IL-5 in the lungs and airways were significantly attenuated in B cell −/− mice, relative to controls. 4) B cell driven Th2 responses and mucus hyper secretion in the lungs were dependent upon MHC II expression by B cells. Conclusions/Significance Collectively, these results provide evidence for antigen presentation as a novel mechanism by which B cells contribute to chronic allergic disease. These findings give new insight into the mechanisms by which B cells promote asthma and other chronic diseases.
Collapse
Affiliation(s)
- Dennis M Lindell
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, United States of America.
| | | | | | | |
Collapse
|
30
|
Holst PJ, Sorensen MR, Mandrup Jensen CM, Orskov C, Thomsen AR, Christensen JP. MHC class II-associated invariant chain linkage of antigen dramatically improves cell-mediated immunity induced by adenovirus vaccines. THE JOURNAL OF IMMUNOLOGY 2008; 180:3339-46. [PMID: 18292559 DOI: 10.4049/jimmunol.180.5.3339] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The ideal vaccine induces a potent protective immune response, which should be rapidly induced, long-standing, and of broad specificity. Recombinant adenoviral vectors induce potent Ab and CD8+ T cell responses against transgenic Ags within weeks of administration, and they are among the most potent and versatile Ag delivery vehicles available. However, the impact of chronic infections like HIV and hepatitis C virus underscore the need for further improvements. In this study, we show that the protective immune response to an adenovirus-encoded vaccine Ag can be accelerated, enhanced, broadened, and prolonged by tethering of the rAg to the MHC class II-associated invariant chain (Ii). Thus, adenovirus-vectored vaccines expressing lymphocytic choriomeningitis virus (LCMV)-derived glycoprotein linked to Ii increased the CD4+ and CD8+ T cell stimulatory capacity in vitro and in vivo. Furthermore, mice vaccinated with a single dose of adenovirus-expressing LCMV-derived glycoprotein linked to Ii were protected against lethal virus-induced choriomeningitis, lethal challenge with strains mutated in immunodominant T cell epitopes, and systemic infection with a highly invasive strain. In therapeutic tumor vaccination, the vaccine was as efficient as live LCMV. In comparison, animals vaccinated with a conventional adenovirus vaccine expressing unmodified glycoprotein were protected against systemic infection, but only temporarily against lethal choriomeningitis, and this vaccine was less efficient in tumor therapy.
Collapse
Affiliation(s)
- Peter Johannes Holst
- Institute of International Health, Immunology and Microbiology, The Panum Institute, Copenhagen, Denmark
| | | | | | | | | | | |
Collapse
|
31
|
Robson N, Donachie A, Mowat A. Simultaneous presentation and cross-presentation of immune-stimulating complex-associated cognate antigen by antigen-specific B cells. Eur J Immunol 2008; 38:1238-46. [DOI: 10.1002/eji.200737758] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
32
|
Ashour HM, Seif TM. The role of B cells in the induction of peripheral T cell tolerance. J Leukoc Biol 2007; 82:1033-9. [PMID: 17656652 DOI: 10.1189/jlb.0507310] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Affiliation(s)
- Hossam M Ashour
- Cairo University, Department of Microbiology and Immunology, Cairo, Egypt.
| | | |
Collapse
|
33
|
Mann MK, Maresz K, Shriver LP, Tan Y, Dittel BN. B cell regulation of CD4+CD25+ T regulatory cells and IL-10 via B7 is essential for recovery from experimental autoimmune encephalomyelitis. THE JOURNAL OF IMMUNOLOGY 2007; 178:3447-56. [PMID: 17339439 DOI: 10.4049/jimmunol.178.6.3447] [Citation(s) in RCA: 248] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
CD4(+)CD25(+) T regulatory (Treg) cells expressing the Foxp3 transcription factor have been shown to be present in the CNS during the autoimmune disease experimental autoimmune encephalomyelitis (EAE) and can inhibit EAE clinical disease by an IL-10-dependent mechanism. In addition, IL-10 expression in the CNS late in the EAE disease course has been attributed to recovery. However, it is not known how Treg cells and IL-10 expressions are regulated during EAE. We have previously shown a requirement for B cells in recovery from EAE and here investigated whether this was due to a deficiency in Treg cells and IL-10 in the CNS. We found that B cell deficiency resulted in a delay in the emergence of Foxp3-expressing Treg cells and IL-10 in the CNS during EAE, but not in the periphery. Reconstitution with wild-type B cells resulted in disease recovery and normalized IL-10 and Foxp3 expression. However, reconstitution with B7-deficient B cells did not. Furthermore, we show that IL-10 and Foxp3 expression is enhanced in CNS nonencephalitogenic T cells. These data suggest a novel mechanism whereby B cells regulate CD4(+)CD25(+) Treg cells via B7 and subsequently enter the CNS and suppress autoimmune inflammation, mediating recovery.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- B-Lymphocytes/immunology
- B-Lymphocytes/metabolism
- B-Lymphocytes/pathology
- B7-1 Antigen/biosynthesis
- B7-1 Antigen/genetics
- B7-1 Antigen/immunology
- Central Nervous System/immunology
- Central Nervous System/metabolism
- Central Nervous System/pathology
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/therapy
- Forkhead Transcription Factors/biosynthesis
- Forkhead Transcription Factors/immunology
- Gene Expression Regulation/immunology
- Interleukin-10/biosynthesis
- Interleukin-10/immunology
- Mice
- Mice, Knockout
- Recovery of Function/immunology
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- T-Lymphocytes, Regulatory/pathology
Collapse
Affiliation(s)
- Monica K Mann
- BloodCenter of Wisconsin, Blood Research Institute, Milwaukee, WI 53226, USA
| | | | | | | | | |
Collapse
|
34
|
Jones N, Eggena M, Baker C, Nghania F, Baliruno D, Mugyenyi P, Ssali F, Barugahare B, Cao H. Presence of distinct subsets of cytolytic CD8+ T cells in chronic HIV infection. AIDS Res Hum Retroviruses 2006; 22:1007-13. [PMID: 17067271 DOI: 10.1089/aid.2006.22.1007] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Cytolytic T lymphocytes (CTL) play an important role in the control of HIV infection. The eventual failure to contain HIV-1 infection may arise because of a functional impairment of HIV-specific CTL. We evaluated Gag-specific cytotoxicity in HIV-1-positive Ugandans. Expression of CD107, a marker for cytolytic activity, was present in CD45RA(bright) and CD45RA(dim) CD8(+) T cell populations in HIV-infected individuals. The frequency of Gag-specific CD107(+)CD45RA(bright)CD28(-)CCR7(-) CD8(+) T cells decreased with CD4 cell depletion and correlated with the presence of Gag-specific T helper response. In contrast, the frequency of Gag-specific CD107(+)CD45RA(dim)CD28(-)CCR7(-) CD8(+) T cells within the same individuals has no significant association with viral load or CD4 cell count. The ratio of CD45RA(bright) to CD45RA(dim) CTL correlates significantly with CD4 cell count. This positive association decreases with antiretroviral treatment (ARV), indicating that suppression of viral replication alters the balance of circulating Gag-specific CD8(+) effector T cells. Subsets of cytolytic T cells may have distinct antiviral functions and further characterization of these effector CD8(+) T cells may yield important information on T cell regulation and dysfunction in HIV infection.
Collapse
Affiliation(s)
- Norman Jones
- California Department of Health Services, Richmond, California 94804, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
McClellan KB, Gangappa S, Speck SH, Virgin HW. Antibody-independent control of gamma-herpesvirus latency via B cell induction of anti-viral T cell responses. PLoS Pathog 2006; 2:e58. [PMID: 16789842 PMCID: PMC1480598 DOI: 10.1371/journal.ppat.0020058] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2006] [Accepted: 04/28/2006] [Indexed: 11/19/2022] Open
Abstract
B cells can use antibody-dependent mechanisms to control latent viral infections. It is unknown whether this represents the sole function of B cells during chronic viral infection. We report here that hen egg lysozyme (HEL)-specific B cells can contribute to the control of murine gamma-herpesvirus 68 (gammaHV68) latency without producing anti-viral antibody. HEL-specific B cells normalized defects in T cell numbers and proliferation observed in B cell-/- mice during the early phase of gammaHV68 latency. HEL-specific B cells also reversed defects in CD8 and CD4 T cell cytokine production observed in B cell-/- mice, generating CD8 and CD4 T cells necessary for control of latency. Furthermore, HEL-specific B cells were able to present virally encoded antigen to CD8 T cells. Therefore, B cells have antibody independent functions, including antigen presentation, that are important for control of gamma-herpesvirus latency. Exploitation of this property of B cells may allow enhanced vaccine responses to chronic virus infection.
Collapse
Affiliation(s)
- Kelly B McClellan
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri, United States of America
| | - Shivaprakash Gangappa
- Emory Transplant Center, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Samuel H Speck
- Division of Microbiology and Immunology, Yerkes Regional Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Herbert W. Virgin
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri, United States of America
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
36
|
Lund FE, Hollifield M, Schuer K, Lines JL, Randall TD, Garvy BA. B cells are required for generation of protective effector and memory CD4 cells in response to Pneumocystis lung infection. THE JOURNAL OF IMMUNOLOGY 2006; 176:6147-54. [PMID: 16670323 DOI: 10.4049/jimmunol.176.10.6147] [Citation(s) in RCA: 144] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
B cell-deficient mice are susceptible to infection by Pneumocystis carinii f. sp. muris (PC). To determine whether this susceptibility is due to a requirement for B cells to prime T cells, we compared CD4 T cell responses to PC in bone marrow chimeric mice that express MHC class II (MHCII) on all APCs (wild-type (WT) chimeras) and in bone marrow chimeric mice that express MHCII on all APCs except B cells (MHCII(-/-) chimeras). Although PC was rapidly cleared by WT chimeric mice, PC levels remained high in chimeric mice that lacked MHCII on B cells. In addition, although T cells were primed in the draining lymph nodes of MHCII(-/-) chimeric mice, the number of activated CD4 T cells infiltrating the lungs of these mice was reduced relative to the number in the lungs of WT chimeras. We also adoptively transferred purified CD4 T cells from the draining lymph nodes of PC-infected normal or B cell-deficient mice into SCID mice. Mice that received CD4 cells from normal mice were able to mount a response to infection in the lungs and clear PC. However, mice that received CD4 cells from B cell-deficient mice had a delayed T cell response in the lungs and failed to control the infection. These data indicate that B cells play a vital role in generation of CD4(+) memory T cells in response to PC infection in the lungs.
Collapse
|
37
|
Ugleholdt R, Poulsen MLH, Holst PJ, Irminger JC, Orskov C, Pedersen J, Rosenkilde MM, Zhu X, Steiner DF, Holst JJ. Prohormone convertase 1/3 is essential for processing of the glucose-dependent insulinotropic polypeptide precursor. J Biol Chem 2006; 281:11050-7. [PMID: 16476726 DOI: 10.1074/jbc.m601203200] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The physiology of the incretin hormones, glucagon-like peptide 1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP), and their role in type 2 diabetes currently attract great interest. Recently we reported an essential role for prohormone convertase (PC) 1/3 in the cleavage of intestinal proglucagon, resulting in formation of GLP-1, as demonstrated in PC1/3-deficient mice. However, little is known about the endoproteolytic processing of the GIP precursor. This study investigates the processing of proGIP in PC1/3 and PC2 null mice and in cell lines using adenovirus-mediated overexpression. Supporting a role for PC1/3 in proGIP processing, we found co-localization of GIP and PC1/3 but not PC2 in intestinal sections by immunohistochemistry, and analysis of intestinal extracts from PC1/3-deficient animals demonstrated severely impaired processing to GIP, whereas processing to GIP was unaltered in PC2-deficient mice. Accordingly, overexpression of preproGIP in the neuroendocrine AtT-20 cell line that expresses high levels of endogenous PC1/3 and negligible levels of PC2 resulted in production of GIP. Similar results were obtained after co-expression of preproGIP and PC1/3 in GH4 cells that express no PC2 and only low levels of PC1/3. In addition, studies in GH4 cells and the alpha-TC1.9 cell line, expressing PC2 but not PC1/3, indicate that PC2 can mediate processing to GIP but also to other fragments not found in intestinal extracts. Taken together, our data indicate that PC1/3 is essential and sufficient for the production of the intestinal incretin hormone GIP, whereas PC2, although capable of cleaving proGIP, does not participate in intestinal proGIP processing and is not found in intestinal GIP-expressing cells.
Collapse
Affiliation(s)
- Randi Ugleholdt
- Department of Medical Physiology, the Panum Institute, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen N, Denmark
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Matter M, Mumprecht S, Pinschewer DD, Pavelic V, Yagita H, Krautwald S, Borst J, Ochsenbein AF. Virus-induced polyclonal B cell activation improves protective CTL memoryvia retained CD27 expression on memory CTL. Eur J Immunol 2005; 35:3229-39. [PMID: 16231287 DOI: 10.1002/eji.200535179] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Different viruses elicit distinct phenotypes of memory cytotoxic T lymphocytes (CTL). This is reflected in differential expression of homing receptors and costimulatory molecules like CD27. Memory CTL retained CD27 following lymphocytic choriomeningitis virus (LCMV) infection, but not after immunization with recombinant vaccinia virus or tumor cells expressing LCMV glycoprotein. Stable CD27 expression on memory CTL required ligation by CD70 expressed on polyclonally activated B cells during the contraction phase. The functional consequence of CD27 expressed on virus-specific CTL was analyzed in CD27-deficient mice. LCMV infection of CD27(-/-) mice revealed that primary CTL activation and expansion as well as elimination of the virus were independent of CD27 expression. In contrast, ligation of CD27 on memory CTL upon secondary antigen encounter increased clonal expansion and improved protection against re-infection. This points to novel B cell-CTL interactions during viral infection and to a beneficial role of polyclonal B cell activation that represents a characteristic of murine LCMV, human immunodeficiency virus and human hepatitis B and C virus infection.
Collapse
MESH Headings
- Animals
- Antigens, CD/biosynthesis
- Antigens, CD/genetics
- Antigens, CD/immunology
- B-Lymphocyte Subsets/immunology
- B-Lymphocyte Subsets/metabolism
- CD27 Ligand
- Cell Proliferation
- Clone Cells
- Humans
- Immunologic Memory
- Lymphocyte Activation/immunology
- Lymphocytic Choriomeningitis/genetics
- Lymphocytic Choriomeningitis/immunology
- Lymphocytic Choriomeningitis/metabolism
- Lymphocytic choriomeningitis virus/immunology
- Membrane Proteins/biosynthesis
- Membrane Proteins/genetics
- Membrane Proteins/immunology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Signal Transduction/immunology
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/metabolism
- T-Lymphocytes, Cytotoxic/virology
- Tumor Necrosis Factor Receptor Superfamily, Member 7/biosynthesis
- Tumor Necrosis Factor Receptor Superfamily, Member 7/genetics
- Tumor Necrosis Factor Receptor Superfamily, Member 7/metabolism
- Tumor Necrosis Factor Receptor Superfamily, Member 7/physiology
- Tumor Necrosis Factors/biosynthesis
- Tumor Necrosis Factors/genetics
- Tumor Necrosis Factors/immunology
Collapse
Affiliation(s)
- Matthias Matter
- Department of Clinical Research, University of Berne, Bern, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Seidel-Guyenot W, Alt R, Perschon S, Knop J, Steinbrink K. B cells are not required for T cell priming in low zone tolerance to contact allergens and contact hypersensitivity. Eur J Immunol 2004; 34:3082-90. [PMID: 15376190 DOI: 10.1002/eji.200425402] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Low zone tolerance (LZT) to contact allergens is induced by epicutaneous exposure to haptens in subsensitizing doses resulting in an inhibition of contact hypersensitivity (CHS), which, in contrast, occurs after sensitization with immunogenic doses of allergens. Performing the protocol of tolerance induction resulted in robust LZT to allergens in B cell-deficient mice in vivo, indicating that B cells are not required for the induction and effector phase of LZT. However, CHS reactions in vivo were restricted in B cell-deficient mice as compared to wild-type (WT) mice. In contrast, analysis of hapten-specific T cell activation in vitro revealed a strong proliferative response of T cells derived from both WT and B cell-deficient sensitized mice. Similar to WT animals, T cells obtained from tolerized B cell-deficient mice produced a Tc2 cytokine pattern of LZT with high levels of IL-4 and IL-10, whereas sensitization of B cell-deficient mice resulted in the typical Tc1 cytokine profile of CHS. Adoptive transfer of CD8+ effector T cells from tolerized or sensitized B cell-deficient mice induced significant LZT or CHS reactions, respectively, in WT recipients, demonstrating that the development of hapten-specific effector CD8+ T cells of LZT and CHS is independent of B cells.
Collapse
|
40
|
Affiliation(s)
- Arturo Casadevall
- Division of Infectious Diseases, Department of Medicine, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, NY 10461, USA.
| | | |
Collapse
|