1
|
Yang NJ, Isensee J, Neel DV, Quadros AU, Zhang HXB, Lauzadis J, Liu SM, Shiers S, Belu A, Palan S, Marlin S, Maignel J, Kennedy-Curran A, Tong VS, Moayeri M, Röderer P, Nitzsche A, Lu M, Pentelute BL, Brüstle O, Tripathi V, Foster KA, Price TJ, Collier RJ, Leppla SH, Puopolo M, Bean BP, Cunha TM, Hucho T, Chiu IM. Anthrax toxins regulate pain signaling and can deliver molecular cargoes into ANTXR2 + DRG sensory neurons. Nat Neurosci 2021; 25:168-179. [PMID: 34931070 DOI: 10.1038/s41593-021-00973-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Accepted: 11/01/2021] [Indexed: 11/09/2022]
Abstract
Bacterial products can act on neurons to alter signaling and function. In the present study, we found that dorsal root ganglion (DRG) sensory neurons are enriched for ANTXR2, the high-affinity receptor for anthrax toxins. Anthrax toxins are composed of protective antigen (PA), which binds to ANTXR2, and the protein cargoes edema factor (EF) and lethal factor (LF). Intrathecal administration of edema toxin (ET (PA + EF)) targeted DRG neurons and induced analgesia in mice. ET inhibited mechanical and thermal sensation, and pain caused by formalin, carrageenan or nerve injury. Analgesia depended on ANTXR2 expressed by Nav1.8+ or Advillin+ neurons. ET modulated protein kinase A signaling in mouse sensory and human induced pluripotent stem cell-derived sensory neurons, and attenuated spinal cord neurotransmission. We further engineered anthrax toxins to introduce exogenous protein cargoes, including botulinum toxin, into DRG neurons to silence pain. Our study highlights interactions between a bacterial toxin and nociceptors, which may lead to the development of new pain therapeutics.
Collapse
Affiliation(s)
- Nicole J Yang
- Department of Immunology, Harvard Medical School, Boston, MA, USA
| | - Jörg Isensee
- Translational Pain Research, Department of Anesthesiology and Intensive Care Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Dylan V Neel
- Department of Immunology, Harvard Medical School, Boston, MA, USA
| | - Andreza U Quadros
- Center for Research in Inflammatory Diseases, Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | | | - Justas Lauzadis
- Department of Anesthesiology, Stony Brook Medicine, Stony Brook, NY, USA
| | | | - Stephanie Shiers
- Department of Neuroscience, Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, USA
| | - Andreea Belu
- Translational Pain Research, Department of Anesthesiology and Intensive Care Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | | | | | | | | | - Victoria S Tong
- Department of Immunology, Harvard Medical School, Boston, MA, USA
| | - Mahtab Moayeri
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Pascal Röderer
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty and University Hospital Bonn, Bonn, Germany.,Cellomics Unit, LIFE & BRAIN GmbH, Bonn, Germany
| | - Anja Nitzsche
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty and University Hospital Bonn, Bonn, Germany.,Cellomics Unit, LIFE & BRAIN GmbH, Bonn, Germany
| | - Mike Lu
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Bradley L Pentelute
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA.,The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.,Broad Institute of MIT and Harvard, Cambridge, MA, USA.,Center for Environmental Health Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Oliver Brüstle
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty and University Hospital Bonn, Bonn, Germany
| | | | | | - Theodore J Price
- Department of Neuroscience, Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, USA
| | - R John Collier
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
| | - Stephen H Leppla
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Michelino Puopolo
- Department of Anesthesiology, Stony Brook Medicine, Stony Brook, NY, USA
| | - Bruce P Bean
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Thiago M Cunha
- Center for Research in Inflammatory Diseases, Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Tim Hucho
- Translational Pain Research, Department of Anesthesiology and Intensive Care Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Isaac M Chiu
- Department of Immunology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
2
|
Abstract
Biologicals, e.g. TNF inhibitors, have improved dramatically the efficacy of medical interventions in autoimmune diseases, such as in rheumatoid arthritis (RA). However, although progressive inflammation can be halted in this way, no drug-free remissions or lasting cures are reached. For this to become real, therapies based on induction antigen-specific immune tolerance are sought. This review describes mechanisms of tolerance and the current possibilities for induction of therapeutic tolerance through antigen-specific vaccination approaches. And despite the fact that for various diseases the search for appropriate autoantigens is ongoing, pioneering studies are now already developed that use more broadly inflammation associated antigens. Through their capacity to preferentially induce regulatory T cells, heat shock proteins are an attractive source of such broadly inflammation associated antigens.
Collapse
Affiliation(s)
- Willem van Eden
- Faculty of Veterinary Medicine, Utrecht University, The Netherlands
| |
Collapse
|
3
|
Andersen MH. Immune Regulation by Self-Recognition: Novel Possibilities for Anticancer Immunotherapy. J Natl Cancer Inst 2015; 107:djv154. [PMID: 26063792 DOI: 10.1093/jnci/djv154] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 05/11/2015] [Indexed: 02/06/2023] Open
Abstract
Circulating T cells that specifically target normal self-proteins expressed by regulatory immune cells were first described in patients with cancer, but can also be detected in healthy individuals. The adaptive immune system is distinguished for its ability to differentiate between self-antigens and foreign antigens. Thus, it was remarkable to discover T cells that apparently lacked tolerance to important self-proteins, eg, IDO, PD-L1, and FoxP3, expressed in regulatory immune cells. The ability of self-reactive T cells to react to and eliminate regulatory immune cells can influence general immune reactions. This suggests that they may be involved in immune homeostasis. It is here proposed that these T cells should be termed antiregulatory T cells (anti-Tregs). The role of anti-Tregs in immune-regulatory networks may be diverse. For example, pro-inflammatory self-reactive T cells that react to regulatory immune cells may enhance local inflammation and inhibit local immune suppression. Further exploration is warranted to investigate their potential role under different malignant conditions and the therapeutic possibilities they possess. Utilizing anti-Tregs for anticancer immunotherapy implies the direct targeting of cancer cells in addition to regulatory immune cells. Anti-Tregs provide the immune system with yet another level of immune regulation and contradict the notion that immune cells involved in the adjustment of immune responses only act as suppressor cells.
Collapse
Affiliation(s)
- Mads Hald Andersen
- Center for Cancer Immune Therapy (CCIT), Department of Hematology, Copenhagen University Hospital, Herlev, Denmark.
| |
Collapse
|
4
|
Hellings N, Raus J, Stinissen P. T-cell-based immunotherapy in multiple sclerosis: induction of regulatory immune networks by T-cell vaccination. Expert Rev Clin Immunol 2010; 2:705-16. [PMID: 20477626 DOI: 10.1586/1744666x.2.5.705] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the CNS with presumed autoimmune origin. Pathogenic autoimmune responses in MS are thought to be the result of a breakdown of self tolerance. Several mechanisms account for the natural state of immunological tolerance to self antigens, including clonal deletion of self-reactive T cells in the thymus. However, autoimmune T cells are also part of the normal T-cell repertoire, supporting the existence of peripheral regulatory mechanisms that keep these potentially pathogenic T cells under control. One such mechanism involves active suppression by regulatory T cells. It has been indicated that regulatory T cells do not function properly in autoimmune disease. Immunization with attenuated autoreactive T cells, T-cell vaccination, may enhance or restore the regulatory immune networks to specifically suppress autoreactive T cells, as shown in experimental autoimmune encephalomyelitis, an animal model for MS. In the past decade, T-cell vaccination has been tested for MS in several clinical trials. This review summarizes these clinical trials and updates our current knowledge on the induction of regulatory immune networks by T cell vaccination.
Collapse
Affiliation(s)
- Niels Hellings
- Hasselt University, Biomedical Research Institute, Diepenbeek, Belgium.
| | | | | |
Collapse
|
5
|
Abstract
Much emphasis has been placed on the so-called "biologics" in the treatment of immune disorders within the last few years. Here we discuss the expanding horizon of potential strategies for immunotherapies targeting T lymphocytes as key effectors and regulators of autoimmunity. We review emerging reagents in a variety of animal models and human disorders that may offer new therapeutic options in current or modified iterations.
Collapse
Affiliation(s)
- Erica Lee
- Department of Dermatology, Weill Medical College of Cornell University, New York, NY, USA
| | | |
Collapse
|
6
|
Prakken B, Albani S. Exploiting T cell crosstalk as a vaccination strategy for rheumatoid arthritis. ACTA ACUST UNITED AC 2007; 56:389-92. [PMID: 17265472 DOI: 10.1002/art.22317] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
7
|
Abstract
Since the discovery of human leukocyte antigen (HLA) as a genetic system, the search for the biological function of HLA molecules has been a very intense and attractive area of immunological research. It has been a major factor in the development of our understanding of the role of T cells in the initiation and regulation of autoimmune diseases. The currently increasing incidences of chronic inflammatory diseases, such as allergies but also several autoimmune diseases, possibly due to our Westernized modern life-style, are asking for novel intervention or prevention strategies. Heat shock proteins (or stress proteins) can be an example of relevant microbial antigens with an intrinsic capacity to trigger anti-inflammatory T cell regulation. Heat shock proteins may be part of an ancient system, which includes several stress-induced self-antigens, seen by a self-protective immune system. Peptide-based intervention, including clinical trials with HSP60 and HSP70 peptides, is now an area of intensive clinical research.
Collapse
Affiliation(s)
- Willem van Eden
- Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, The Netherlands.
| |
Collapse
|
8
|
Abstract
The complexity of a self-regulatory system demands a balance between effectors and regulators; that is, it is necessary for both cell types to exist. Regulation of self-reactive T cells can occur at several complementary but different levels: (1) at the level of priming itself: for example, inhibition of expansion of antigen-reactive T cells by regulatory CD4+ CD25+ T cells; (2) after the priming of self-reactive T cells, regulatory T-cell populations with reactivity to distinct self-determinants derived from the T-cell receptor (TCR) can be engaged via a negative feedback mechanism. Thus, these mechanisms ensure induction of effective and appropriately limited responses against foreign antigens while preventing autoreactivity from inflicting self-damage.
Collapse
Affiliation(s)
- Vipin Kumar
- Torrey Pines Institute for Molecular Studies, San Diego, California 92121, USA.
| | | |
Collapse
|
9
|
Lal G, Shaila MS, Nayak R. Activated mouse T-cells synthesize MHC class II, process, and present morbillivirus nucleocapsid protein to primed T-cells. Cell Immunol 2005; 234:133-45. [PMID: 16083868 DOI: 10.1016/j.cellimm.2005.06.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2004] [Revised: 06/02/2005] [Accepted: 06/06/2005] [Indexed: 11/25/2022]
Abstract
A pivotal step in the initiation of T-cell immunity is the presentation of antigenic peptides by major histocompatibility complex (MHC) expressed on antigen presenting cells. The expression of MHC class II molecules by mouse T-cells has not been shown unequivocally. In the present work, we demonstrate that activated mouse T-cells synthesize MHC class II molecules de novo and express them on their surface. Further, we have demonstrated that in vitro activated T-cells take up extra-cellular soluble nucleocapsid protein of a morbillivirus. The internalized antigen goes to antigen processing compartment as shown by co-localization of antigen and LAMP-1 using confocal microscopy. We show that activated T-cells express H2M, a chaperone molecule known to have a role in antigen presentation. Further, we demonstrate that activated T-cells process and present internalized extra-cellular antigen to primed T-cells as shown by IL-2 secretion and in vitro proliferation. The presentation of antigen by T-cells may have implications in immuno-regulation, control of infection by lymphotropic viruses and maintenance of immunological memory.
Collapse
Affiliation(s)
- Girdhari Lal
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore 560012, India
| | | | | |
Collapse
|
10
|
Hellings N, Raus J, Stinissen P. T-cell vaccination in multiple sclerosis: update on clinical application and mode of action. Autoimmun Rev 2004; 3:267-75. [PMID: 15246022 DOI: 10.1016/j.autrev.2003.10.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2003] [Accepted: 10/12/2003] [Indexed: 11/19/2022]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system (CNS). Autoreactive T cells specific for myelin antigens are considered to play a prominent role in the initiation of the local inflammatory response, ultimately leading to myelin damage. Several studies indicate that autoreactive T cells are not completely deleted in the thymus, but are part of the normal T cell repertoire. Accidentally activated autoreactive T cells, however, may not automatically lead to autoimmune disease. Several reports support the existence of peripheral regulatory networks that prevent the activation and expansion of pathogenic T cells. Anti-idiotypic and anti-ergotypic T cells are part of this regulatory network and are thought to control autoreactive T cells by recognition of certain clonotypic and ergotypic determinants. These clonotypic networks may not function properly in patients with MS. Immunization with attenuated autoreactive T cells, termed T cell vaccination (TCV), may enhance or restore the regulatory networks to specifically suppress the autoreactive T cells as shown in experimental autoimmune encephalomyelitis (EAE), a commonly used animal model for MS. In the past decade, TCV has been tested for MS in several clinical trails. This review summarizes these clinical trails and updates our current knowledge on the mode of action of T cell vaccination.
Collapse
Affiliation(s)
- Niels Hellings
- Biomedisch Onderzoeksinstituut, Limburgs Universitair Centrum, University Campus Building A, Diepenbeek B-3590, Belgium
| | | | | |
Collapse
|
11
|
Van Eden W, Waksman BH. Immune regulation in adjuvant-induced arthritis: possible implications for innovative therapeutic strategies in arthritis. ARTHRITIS AND RHEUMATISM 2003; 48:1788-96. [PMID: 12847671 DOI: 10.1002/art.11077] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
12
|
Zhang XY, Liu XG, Wang W, Wang WC, Gao XM. Anti-T-cell humoral and cellular responses in healthy BALB/c mice following immunization with ovalbumin or ovalbumin-specific T cells. Immunology 2003; 108:465-73. [PMID: 12667208 PMCID: PMC1782931 DOI: 10.1046/j.1365-2567.2003.01604.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
It is known that T-cell vaccination (TCV) elicits cellular immune responses against the immunizing T cells in vivo. However, it is still unclear whether similar anti-T-cell responses are also induced in individuals responding to foreign antigen (Ag) challenge. This study was undertaken to characterize and compare anti-T-cell cellular and humoral responses of BALB/c mice after ovalbumin (OVA) immunization or TCV. Splenocytes from OVA-primed BALB/c mice proliferated in response to stimulation with a syngeneic OVA-specific T-cell clone, OVA-T3, and secreted interferon-gamma (IFN-gamma) but not interleukin-4 (IL-4). Cytotoxic T-lymphocyte (CTL) activity against the T-cell clone was also observed. Serum samples from these animals discriminated between activated and resting murine T cells, as determined by indirect immunofluorescence staining. Vaccination of BALB/c mice with OVA-T3 cells induced similar, but stronger, cellular and humoral responses. TCV-induced antibodies were not only able to distinguish between activated and resting syngeneic T cells but also positively stained allogeneic T cells from BXSB mice. Furthermore, TCV resulted in hyporesponsiveness of BALB/c mice to subsequent Ag challenge, and antisera from the immunized animals inhibited T-cell proliferation in vitro. Our data suggest that anti-T-cell antibodies, and CD4+ and CD8+ regulatory T lymphocytes may form a complex and co-ordinated regulatory network that plays an important role in regulating the adaptive immune responses in vivo. Implications of this observation for our understanding of the immunoregulation and peripheral tolerance are discussed.
Collapse
Affiliation(s)
- Xiao-Yu Zhang
- Department of Immunology, Peking University Health Science Center, Peking University, Beijing, China
| | | | | | | | | |
Collapse
|
13
|
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system (CNS). Significant progress has been made in our understanding of the etiology of MS. MS is widely believed to be an autoimmune disease that results from aberrant immune responses to CNS antigens. T cells are considered to be crucial in orchestrating an immunopathological cascade that results in damage to the myelin sheath. This review summarizes the currently available data supporting the idea that myelin reactive T cells are actively involved in the immunopathogenesis of MS. Some of the therapeutic strategies for MS are discussed with a focus on immunotherapies that aim to specifically target the myelin reactive T cells.
Collapse
Affiliation(s)
- Niels Hellings
- Biomedical Research Institute, Limburg University Center, School for Life Sciences, Transnational University Diepenbeek, Belgium
| | | | | |
Collapse
|
14
|
Vandenbark AA, Morgan E, Bartholomew R, Bourdette D, Whitham R, Carlo D, Gold D, Hashim G, Offner H. TCR peptide therapy in human autoimmune diseases. Neurochem Res 2001; 26:713-30. [PMID: 11519731 DOI: 10.1023/a:1010951706830] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Inflammatory Th1 cells reacting to tissue/myelin derived antigens likely contribute to the pathogenesis of diseases such as multiple sclerosis (MS), rheumatoid arthritis (RA), and psoriasis. One regulatory mechanism that may be useful for treating autoimmune diseases involves an innate second set of Th2 cells specific for portions of the T cell receptor of clonally expanded pathogenic Th1 cells. These Th2 cells are programmed to respond to internally modified V region peptides from the T cell receptor (TCR) that are expressed on the Th1 cell surface in association with major histocompatibility molecules. Once the regulatory Th2 cells are specifically activated, they may inhibit inflammatory Th1 cells through a non-specific bystander mechanism. A variety of strategies have been used by us to identify candidate disease-associated TCR V genes present on pathogenic Th1 cells, including BV5S2, BV6S5, and BV13SI in MS, BV3, BV14, and BV17 in RA, and BV3 and BV13S1 in psoriasis. TCR peptides corresponding to the mid region of these BV genes were found to be consistently immunogenic in vivo when administered either i.d. in saline or i.m. in incomplete Freund's adjuvant (IFA). In MS patients, repeated injection of low doses of peptides (100-300 microg) significantly boosted the number of TCR-reactive Th2 cells. These activated cells secreted cytokines, including IL-10, that are known to inhibit inflammatory Th1 cells. Cytokine release could also be induced in TCR-reactive Th2 cells by direct cell-cell contact with Th1 cells expressing the target V gene. These findings indicate the potential of regulatory Th2 cells to inhibit not only the target Th1 cells, but also bystander Th1 cells expressing different V genes specific for other autoantigens. TCR peptide vaccines have been used in our studies to treat a total of 171 MS patients (6 trials), 484 RA patients (7 trials), and 177 psoriasis patients (2 trials). Based on this experience in 824 patients with autoimmune diseases, TCR peptide vaccination is safe and well tolerated, and can produce significant clinical improvement in a subset of patients that respond to immunization. TCR peptide vaccination represents a promising approach that is well-suited for treating complex autoimmune diseases.
Collapse
Affiliation(s)
- A A Vandenbark
- Neuroimmunology Research, Veterans Affairs Medical Center, Portland, OR 97201, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
15
|
van Tienhoven EA, Broeren CP, Noordzij A, Wagenaar JP, van Eden W, Wauben MH. Nasal application of a naturally processed and presented T cell epitope derived from TCR AV11 protects against adjuvant arthritis. Int Immunol 2000; 12:1715-21. [PMID: 11099311 DOI: 10.1093/intimm/12.12.1715] [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: 11/13/2022] Open
Abstract
Reactivity towards TCR peptides plays an important role in the regulation of several experimental autoimmune diseases. In a previous paper, we showed the TCRAV11 usage by an arthritogenic T cell clone isolated from a rat with adjuvant arthritis (AA). Moreover, we identified three immunogenic peptides in AV11: AV11 24-40, 41-55 and 66-80. In the present study, we show that T cells directed towards all three epitopes are part of the immune repertoire. The strongest delayed-type hypersensitivity (DTH) reaction was observed against the peptide derived from the third framework region, peptide AV11 66-80. DTH reactions to this peptide were detectable in naive rats and increased significantly after AA induction. Interestingly, modulation of the AV11 66-80 T cell response by nasal AV11 66-80 administration resulted in reduced DTH responses and in a strong inhibition of AA. These findings suggest that during the natural course of AA, T cells directed towards the third framework region of AV11 do not have a disease regulatory function, but instead play a role in the deterioration of AA.
Collapse
Affiliation(s)
- E A van Tienhoven
- Institute of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, PO Box 80165, Yalelaan 1, 3508 TD Utrecht, The Netherlands
| | | | | | | | | | | |
Collapse
|
16
|
Lombardi G, Dunne PJ, Scheel-Toellner D, Sanyal T, Pilling D, Taams LS, Life P, Lord JM, Salmon M, Akbar AN. Type 1 IFN maintains the survival of anergic CD4+ T cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 165:3782-9. [PMID: 11034383 DOI: 10.4049/jimmunol.165.7.3782] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Anergic T cells have immunoregulatory activity and can survive for extended periods in vivo. It is unclear how anergic T cells escape from deletion, because both anergy and apoptosis can occur after TCR ligation. Stimulation of human CD4+ T cell clones reactive to influenza hemagglutinin peptides can occur in the absence of APCs when MHC class II-expressing, activated T cells present peptide to each other. This T:T peptide presentation can induce CD95-mediated apoptosis, while the cells that do not die are anergic. We found that the death after peptide or anti-CD3 treatment of a panel of CD4+ T cell clones is blocked by IFN-beta secreted by fibroblasts and also by IFN-alpha. This increases cell recovery after stimulation, which is not due to T cell proliferation. This mechanism for apoptosis inhibition rapidly stops protein kinase C-delta translocation from the cytoplasm to the nucleus, which is an early event in the death process. A central observation was that CD4+ T cells that are rescued from apoptosis after T:T presentation of peptide by IFN-alphabeta remain profoundly anergic to rechallenge with Ag-pulsed APCs. However, anergized cells retain the ability to respond to IL-2, showing that they are nonresponsive but functional. The prevention of peptide-induced apoptosis in activated T cells by IFN-alphabeta is a novel mechanism that may enable the survival and maintenance of anergic T cell populations after TCR engagement. This has important implications for the persistence of anergic T cells with the potential for immunoregulatory function in vivo.
Collapse
Affiliation(s)
- G Lombardi
- Department of Clinical Immunology, Royal Free and University College Medical School, London, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Abstract
T cell anergy is one of the mechanisms leading to the establishment and maintenance of peripheral tolerance. Recent data from our and other laboratories indicate that anergic T cells are not functionally inert but in fact are capable of regulating the immune response in an active manner. In this review, we describe our viewpoint on how anergic self-reactive T cells could contribute to regulation of the immune response.
Collapse
Affiliation(s)
- L S Taams
- Institute of Infectious Diseases and Immunology, Department of Immunology, Utrecht University, The Netherlands
| | | |
Collapse
|
18
|
Zang YC, Hong J, Rivera VM, Killian J, Zhang JZ. Preferential recognition of TCR hypervariable regions by human anti-idiotypic T cells induced by T cell vaccination. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 164:4011-7. [PMID: 10754292 DOI: 10.4049/jimmunol.164.8.4011] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
T cell responses to myelin basic protein (MBP) are potentially involved in the pathogenesis of multiple sclerosis (MS). Immunization with irradiated MBP-reactive T cells (T cell vaccination) induces anti-idiotypic T cell responses that suppress circulating MBP-reactive T cells. This T cell-T cell interaction is thought to involve the recognition of TCR expressed on target T cells. The study was undertaken to define the idiotypic determinants responsible for triggering CD8+ cytotoxic anti-idiotypic T cell responses by T cell vaccination in patients with MS. A panel of 9-mer synthetic TCR peptides corresponding to complementarity-determining region 2 (CDR2) and CDR3 of the immunizing MBP-reactive T cell clones were used to isolate anti-idiotypic T cell lines from immunized MS patients. The resulting TCR-specific T cell lines expressed exclusively the CD8 phenotype and recognized preferentially the CDR3 peptides. CDR3-specific T cell lines were found to lyze specifically autologous immunizing MBP-reactive T cell clones. The findings suggest that CDR3-specific T cells represented anti-idiotypic T cell population induced by T cell vaccination. In contrast, the CDR2 peptides were less immunogenic and contained cryptic determinants as the CDR2-specific T cell lines did not recognize autologous immunizing T cell clones from which the peptide sequence was derived. The study has important implications in our understanding of in vivo idiotypic regulation of autoimmune T cells and the regulatory mechanism underlying T cell vaccination.
Collapse
Affiliation(s)
- Y C Zang
- Multiple Sclerosis Research Laboratory, Department of Neurology and Baylor-Methodist Multiple Sclerosis Center, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | | | | |
Collapse
|
19
|
Taams LS, van Eden W, Wauben MHM. Dose-Dependent Induction of Distinct Anergic Phenotypes: Multiple Levels of T Cell Anergy. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.162.4.1974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Abstract
T cell anergy has been proposed as one of the mechanisms underlying peripheral T cell tolerance. In recent years, the functional relevance of T cell anergy has been studied extensively in vitro and in vivo, using different species, cell systems, and ways to induce anergy. Although these studies concurred about the induction of unresponsiveness, conflicting findings were obtained with respect to the function of anergic T cells and to the persistence of T cell anergy. In the present study, T cell anergy was induced through T-T presentation of the specific Ag by rat MHC class II+ T cells in the absence of professional APC. We show that, depending on the Ag dose with which T cells were incubated, distinct anergic phenotypes were induced. Incubation of T cell clones with a low (suboptimal) Ag dose induced hyporesponsiveness. Incubation with a higher (optimal) Ag dose induced an anergic state capable of exerting immunoregulatory effects. Incubation with a high (supraoptimal) Ag dose led to an anergic suppressive phenotype that was persistent and was not reversed by APC, Ag, and rIL-2. These findings demonstrate that T cell anergy is not confined to a single state of functional inactivation. Instead, multiple levels of T cell anergy exist. Thus, anergic T cells can contribute to the regulation of the immune response either in a persistent and active manner or in a passive manner, depending on their level of T cell anergy.
Collapse
Affiliation(s)
- Leonie S. Taams
- Institute of Infectious Diseases and Immunology, Department of Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Willem van Eden
- Institute of Infectious Diseases and Immunology, Department of Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Marca H. M. Wauben
- Institute of Infectious Diseases and Immunology, Department of Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
20
|
Kumar V, Sercarz E. Induction or Protection from Experimental Autoimmune Encephalomyelitis Depends on the Cytokine Secretion Profile of TCR Peptide-Specific Regulatory CD4 T Cells. THE JOURNAL OF IMMUNOLOGY 1998. [DOI: 10.4049/jimmunol.161.12.6585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Abstract
Autoimmune diseases can result from the breakdown of regulation and subsequent activation of self-antigenic determinant-reactive T cells. During the evolution of the autoimmune response to myelin basic protein (MBP) in B10.PL mice, several distinct T cell populations expand: the effectors mediating experimental autoimmune encephalomyelitis (EAE) are MBP-reactive, CD4+, and predominantly TCR Vβ8.2+; in addition, at least two regulatory populations can be detected—one comprised of Vβ14+ CD4 T cells, reactive to a framework region 3 determinant on the Vβ8.2 chain, and a second that is CD8+ and reactive to another Vβ8.2 determinant. The combined action of these two regulatory cell types controls disease-causing effectors, resulting in spontaneous recovery from disease. In this report, we reveal that the cytokine secretion pattern of TCR peptide-specific regulatory CD4 T cells can profoundly influence whether a type 1 or type 2 population predominates among MBP-specific CD4 effectors. The priming of type 1 regulatory T cells results in deviation of the Ag-specific effector T cell population in a type 2 direction and protection from disease. In contrast, induction of type 2 regulatory T cells results in exacerbation of EAE, poor recovery, and an increased frequency of type 1 effectors. Thus, the encephalitogenic potential of the MBP-reactive effector population is crucially and dominantly influenced by the cytokine secretion phenotype of regulatory CD4 T cells. These findings have important implications in understanding peripheral tolerance to self-Ags as well as in the design of TCR-based therapeutic approaches.
Collapse
Affiliation(s)
- Vipin Kumar
- Department of Microbiology and Molecular Genetics, University of California, Los Angeles, CA 90095
| | - Eli Sercarz
- Department of Microbiology and Molecular Genetics, University of California, Los Angeles, CA 90095
| |
Collapse
|
21
|
Burrows G, Ariail K, Celnik B, Gambee J, Offner H, Vandenbark A. Multiple class I motifs revealed by sequencing naturally processed peptides eluted from rat T cell MHC molecules. J Neurosci Res 1997. [DOI: 10.1002/(sici)1097-4547(19970701)49:1<107::aid-jnr12>3.0.co;2-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
22
|
|
23
|
|
24
|
Vandenbark AA, Chou YK, Whitham R, Mass M, Buenafe A, Liefeld D, Kavanagh D, Cooper S, Hashim GA, Offner H. Treatment of multiple sclerosis with T-cell receptor peptides: results of a double-blind pilot trial. Nat Med 1996; 2:1109-15. [PMID: 8837609 DOI: 10.1038/nm1096-1109] [Citation(s) in RCA: 139] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
A T-cell receptor (TCR) peptide vaccine from the V beta 5.2 sequence expressed in multiple sclerosis (MS) plaques and on myelin basic protein (MBP)-specific T cells boosted peptide-reactive T cells in patients with progressive MS. Vaccine responders had a reduced MBP response and remained clinically stable without side effects during one year of therapy, whereas nonresponders had an increased MBP response and progressed clinically. Peptide-specific T helper 2 cells directly inhibited MBP-specific T helper 1 cells in vitro through the release of interleukin-10, implicating a bystander suppression mechanism that holds promise for treatment of MS and other autoimmune diseases.
Collapse
Affiliation(s)
- A A Vandenbark
- Veterans Affairs Medical Center, Portland, Oregon 97201, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Haqqi TM, Qu XM, Anthony D, Ma J, Sy MS. Immunization with T cell receptor V beta chain peptides deletes pathogenic T cells and prevents the induction of collagen-induced arthritis in mice. J Clin Invest 1996; 97:2849-58. [PMID: 8675697 PMCID: PMC507379 DOI: 10.1172/jci118741] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Collagen-induced arthritis (CIA) in susceptible strains of mice is an animal model of T cell-mediated inflammatory polyarthritis. Analysis of T cell receptor (TCR) V beta gene usage in cells isolated from arthritic joints of BUB/BnJ (BUB) mice (H-2q, TCR V beta a) showed that TCR V beta chain gene usage was limited to TCR V beta 3 and V beta 10 gene families. All of the BUB mice immunized with a mixture of TCR V beta 3 and TCR V beta 10 peptides, but not with control TCR V beta 14 peptide, were refractory to the induction of CIA. Immunization with TCR V beta 3 and V beta 10 peptides completely blocked the development of clinical and subclinical inflammation, formation of pannus and synovial hyperplasia, and the erosion of cartilage and bone. Further studies revealed that preimmunization of BUB mice with V beta 10 peptide alone was sufficient to render the mice resistant to CIA. Analysis of TCR V beta chain gene expression in lymph node cells from arthritic and arthritis-protected mice showed the expression of TCR V beta 10 subfamily in all of the arthritic mice, but not in arthritis-protected mice. Immunization with TCR V beta peptides did not diminish the humoral responses to chicken type-II collagen and also elicited significant levels of anti-V beta 3 and anti-V beta 10 peptide antibodies. Antibodies cross-reactive with mouse chicken type-II collagen were detected in both the arthritic and arthritis-protected mice. Adoptive transfer of serum from arthritis-protected BUB mice significantly delayed the onset (P < 0.005) of arthritis in recipient BUB mice. In contrast, mice injected with serum from arthritic mice had early onset of arthritis. These results demonstrate that immunization of BUB mice with TCR V beta chain peptides elicited antibodies reactive with the self-TCR and prevented the induction of collagen-induced arthritis by eliminating or downregulating pathogenic T cells and consequently blocking the development of humoral immune response. These findings may have clinical applications in treating human autoimmune diseases characterized by common TCR gene usage.
Collapse
Affiliation(s)
- T M Haqqi
- Department of Medicine, Case Western Reserve University, Cleveland, OH 44106-4946, USA.
| | | | | | | | | |
Collapse
|
26
|
Abstract
The natural tendency in T cell-mediated autoimmune conditions to develop focused antigen-specific responses that over-utilize certain T cell receptor (TCR) V region segments prompts the induction of anti-TCR-specific T cells and antibodies that can inhibit the pathogenic T cells and promote recovery from disease. This natural regulatory network can be manipulated by injecting synthetic peptide vaccines that correspond to segments of the over-expressed V genes. In experimental autoimmune encephalomyelitis (EAE), an animal model for the human disease multiple sclerosis (MS), the pathogenic T cells are directed at myelin components, including basic protein (MBP). In some strains such as the Lewis rat and the PL/J mouse, the encephalitogenic BP-specific T cells overexpress a particular V region gene (V beta 8.2) in the TCR. In vivo administration of V beta 8.2 peptides in rats or mice can prevent and treat EAE by boosting regulatory anti-V beta 8.2-specific T cells that inhibit but do not delete the encephalitogenic specificities. This regulation is mediated by soluble factors, suggesting that the presence of regulatory TCR-specific T cells within the target organ (the central nervous system) may inhibit not only the stimulating V beta 8.2 + T cells, but also bystander T cells bearing different V genes. Parallel studies in MS patients have revealed striking V gene biases among BP-specific T cell clones from some patients that provided a rationale for TCR peptide therapy. Injection of V beta 5.2 and V beta 6.1 peptides boosted the frequency of TCR peptide-specific T cells and reduced responses to BP, in some cases with clinical benefit, indicating the presence of an anti-TCR regulatory network in humans that may also be manipulated with TCR peptide therapy.
Collapse
Affiliation(s)
- A A Vandenbark
- Veterans Affairs Medical Center, New York, New York, USA
| | | | | |
Collapse
|
27
|
Van Eden W, Anderton SM, Van Der Zee R, Prakken BJ, Broeren CP, Wauben MH. (Altered) self peptides and the regulation of self reactivity in the peripheral T cell pool. Immunol Rev 1996; 149:55-73. [PMID: 9005220 DOI: 10.1111/j.1600-065x.1996.tb00899.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Tolerance for self has appeared incomplete for many self antigens. We have obtained experimental evidence that both for self heat shock proteins and T cell receptor V-gene products, reactive T cells are part of the normal immune repertoire. Furthermore, it has become apparent that stimulation of T cell responsiveness to these antigens, by using peptide immunisation or by transfer of activated T cells, raises resistance to experimentally induced autoimmune arthritis. In addition, available evidence has suggested that these reactivities may be functional during natural processes of disease remission. The observations with regard to heat-shock proteins have indicated that mechanism leading to disease resistance are most efficiently triggered by exposing the immune system to non-self antigens such as bacterial hsp's, which are similar to, but not identical to, self. Experimental evidence has been obtained, that conserved bacterial hsp peptides, may trigger self hsp reactive T cells, with disease suppressive regulatory potential. It is possible that such self hsp reactive T cells, being expanded by recognising bacterial peptides as full agonists, do, in fact, perceive the self epitopes as partial agonists, and therefore have the possibility of displaying downregulatory activity at the site of inflammation. Experiments with peptide analogs of self epitopes, being variants of disease critical T cell epitopes, have indeed suggested that also their activity in modulating disease may comply with the principles of dominant immunological tolerance.
Collapse
Affiliation(s)
- W Van Eden
- Department of Immunology, Utrecht University, Netherlands
| | | | | | | | | | | |
Collapse
|
28
|
Abstract
Studies of the immune response of mammals to infectious agents have revealed that members of the hsp60 and hsp 70 family are highly immunodominant. Given their high conservation during evolution this was surprising, because of the apparent risk of triggering of autoimmunity and autoimmune disease during the defense of a mammal against infection. However, detailed studies of the immune responses to HSP in models of autoimmune diseases in animals resulted in a change of the view that autoimmunity necessarily leads to autoimmune disease. It has been found that modulation of autoimmunity to HSP is one way to prevent autoimmune disease. At least in some cases even treatment of autoimmune diseases by immunization with heat shock protein appears feasible. This was shown in adjuvant arthritis in Lewis rats and insulin dependent diabetes in NOD mice. Hsp60 and hsp70 are ubiquitous proteins. Their involvement in regulatory loops of autoimmunity may serve as basis for the development of strategies, to prevent and/or treat autoimmune diseases even without knowledge of the causative (auto-)antigen.
Collapse
Affiliation(s)
- U Feige
- Department of Pharmacology, AMGEN Center, Thousand Oaks, CA, USA
| | | |
Collapse
|
29
|
Broeren CP, Wauben MH, Lucassen MA, Van Meurs M, Van Kooten PJ, Boog CJ, Claassen E, Van Eden W. Activated rat T cells synthesize and express functional major histocompatibility class II antigens. Immunology 1995; 84:193-201. [PMID: 7750994 PMCID: PMC1415087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
In the present report, we studied the presence and functional significance of major histocompatibility complex (MHC) class II antigen on rat T cells. Most rat T-cell lines cultured in vitro were found to be MHC class II+. Also, these T-cell lines were shown to synthesize MHC class II molecules. Immunohistochemical and flow cytometric double stainings for T-cell receptor (TCR) and MHC class II showed that in vivo as well a large proportion of T cells was MHC class II+. The immunohistochemical staining of spleen sections enabled us to characterize the MHC class II+ and MHC class II- T cells. It was shown that resting T cells in vivo were MHC class II-. In contrast, activated T cells, as determined by their localization in the marginal zone of the spleen, proved to be MHC class II+. Finally, T-cell clones were found to be able to present peptidic antigens, but could only poorly present more complex exogenous antigens, probably due to inefficient uptake of such antigens. These features would endow activated rat T cells with the capacity to present cell-specific self-proteins, such as TCR, to regulatory CD4+ MHC class II-restricted T cells, as was described by our group elsewhere.
Collapse
Affiliation(s)
- C P Broeren
- Institute of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, University of Utrecht, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Stangel M, Giegerich G, Torres-Nagel NE, Hünig T, Hartung HP. Structural analysis of the rat T-cell receptor Tcra V4 gene family. Immunogenetics 1995; 41:125-30. [PMID: 7806283 DOI: 10.1007/bf00182323] [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: 01/27/2023]
Abstract
The rat Tcra V gene locus is only poorly characterized, although rats are widely used in a variety of T-cell-mediated experimental animal models. Recently, we described the first monoclonal antibody, G99, directed against a rat Tcra V4 segment. We examined cDNA transcripts of G99-positively sorted T cells and show that the monoclonal antibody G99 most likely recognizes at least two members of the Tcra V4 family. Moreover, we analyzed the genomic repertoire of this VA family and report 15 novel Tcra V4 DNA sequences. Based on sequence and Southern blot analysis, the Tcra V4 family could be divided into four subgroups, which were also detected in mice. These findings corroborate previous findings of a similar genetic organization of the Tcra V loci in both species.
Collapse
Affiliation(s)
- M Stangel
- Department of Neurology, University of Würzburg, Germany
| | | | | | | | | |
Collapse
|