1
|
Rau CN, Severin ME, Lee PW, Deffenbaugh JL, Liu Y, Murphy SP, Petersen-Cherubini CL, Lovett-Racke AE. MicroRNAs targeting TGF-β signaling exacerbate central nervous system autoimmunity by disrupting regulatory T cell development and function. Eur J Immunol 2024; 54:e2350548. [PMID: 38634287 PMCID: PMC11156541 DOI: 10.1002/eji.202350548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 03/04/2024] [Accepted: 03/07/2024] [Indexed: 04/19/2024]
Abstract
Transforming growth factor beta (TGF-β) signaling is essential for a balanced immune response by mediating the development and function of regulatory T cells (Tregs) and suppressing autoreactive T cells. Disruption of this balance can result in autoimmune diseases, including multiple sclerosis (MS). MicroRNAs (miRNAs) targeting TGF-β signaling have been shown to be upregulated in naïve CD4 T cells in MS patients, resulting in a limited in vitro generation of human Tregs. Utilizing the murine model experimental autoimmune encephalomyelitis, we show that perinatal administration of miRNAs, which target the TGF-β signaling pathway, enhanced susceptibility to central nervous system (CNS) autoimmunity. Neonatal mice administered with these miRNAs further exhibited reduced Treg frequencies with a loss in T cell receptor repertoire diversity following the induction of experimental autoimmune encephalomyelitis in adulthood. Exacerbated CNS autoimmunity as a result of miRNA overexpression in CD4 T cells was accompanied by enhanced Th1 and Th17 cell frequencies. These findings demonstrate that increased levels of TGF-β-associated miRNAs impede the development of a diverse Treg population, leading to enhanced effector cell activity, and contributing to an increased susceptibility to CNS autoimmunity. Thus, TGF-β-targeting miRNAs could be a risk factor for MS, and recovering optimal TGF-β signaling may restore immune homeostasis in MS patients.
Collapse
Affiliation(s)
- Christina N Rau
- Department of Microbial Infection and Immunity, Wexner Medical Center, The Ohio State University, Columbus, Ohio, USA
| | - Mary E Severin
- Department of Microbial Infection and Immunity, Wexner Medical Center, The Ohio State University, Columbus, Ohio, USA
- Biomedical Sciences Graduate Program, The Ohio State University, Columbus, Ohio, USA
| | - Priscilla W Lee
- Department of Microbial Infection and Immunity, Wexner Medical Center, The Ohio State University, Columbus, Ohio, USA
- Molecular, Cellular, and Developmental Biology Graduate Program, The Ohio State University, Columbus, Ohio, USA
| | - Joshua L Deffenbaugh
- Department of Microbial Infection and Immunity, Wexner Medical Center, The Ohio State University, Columbus, Ohio, USA
| | - Yue Liu
- Department of Microbial Infection and Immunity, Wexner Medical Center, The Ohio State University, Columbus, Ohio, USA
| | - Shawn P Murphy
- Department of Microbial Infection and Immunity, Wexner Medical Center, The Ohio State University, Columbus, Ohio, USA
| | - Cora L Petersen-Cherubini
- Department of Microbial Infection and Immunity, Wexner Medical Center, The Ohio State University, Columbus, Ohio, USA
- Neuroscience Graduate Program, The Ohio State University, Columbus, Ohio, USA
| | - Amy E Lovett-Racke
- Department of Microbial Infection and Immunity, Wexner Medical Center, The Ohio State University, Columbus, Ohio, USA
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
2
|
Koh CH, Lee S, Kwak M, Kim BS, Chung Y. CD8 T-cell subsets: heterogeneity, functions, and therapeutic potential. Exp Mol Med 2023; 55:2287-2299. [PMID: 37907738 PMCID: PMC10689838 DOI: 10.1038/s12276-023-01105-x] [Citation(s) in RCA: 68] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 08/11/2023] [Accepted: 08/12/2023] [Indexed: 11/02/2023] Open
Abstract
CD8 T cells play crucial roles in immune surveillance and defense against infections and cancer. After encountering antigenic stimulation, naïve CD8 T cells differentiate and acquire effector functions, enabling them to eliminate infected or malignant cells. Traditionally, cytotoxic T cells, characterized by their ability to produce effector cytokines and release cytotoxic granules to directly kill target cells, have been recognized as the constituents of the predominant effector T-cell subset. However, emerging evidence suggests distinct subsets of effector CD8 T cells that each exhibit unique effector functions and therapeutic potential. This review highlights recent advancements in our understanding of CD8 T-cell subsets and the contributions of these cells to various disease pathologies. Understanding the diverse roles and functions of effector CD8 T-cell subsets is crucial to discern the complex dynamics of immune responses in different disease settings. Furthermore, the development of immunotherapeutic approaches that specifically target and regulate the function of distinct CD8 T-cell subsets holds great promise for precision medicine.
Collapse
Affiliation(s)
- Choong-Hyun Koh
- Laboratory of Immune Regulation, Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Suyoung Lee
- Laboratory of Immune Regulation, Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
- BK21 Plus Program, College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea
| | - Minkyeong Kwak
- Laboratory of Immune Regulation, Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
- BK21 Plus Program, College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea
| | - Byung-Seok Kim
- Division of Life Sciences, College of Life Science and Bioengineering, Incheon National University, Incheon, 22012, Republic of Korea
| | - Yeonseok Chung
- Laboratory of Immune Regulation, Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea.
- BK21 Plus Program, College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea.
- Wide River Institute of Immunology, Seoul National University, Hongcheon, Gangwon, 25159, Republic of Korea.
| |
Collapse
|
3
|
Mangani D, Yang D, Anderson AC. Learning from the nexus of autoimmunity and cancer. Immunity 2023; 56:256-271. [PMID: 36792572 PMCID: PMC9986833 DOI: 10.1016/j.immuni.2023.01.022] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 01/13/2023] [Accepted: 01/19/2023] [Indexed: 02/16/2023]
Abstract
The immune system plays critical roles in both autoimmunity and cancer, diseases at opposite ends of the immune spectrum. Autoimmunity arises from loss of T cell tolerance against self, while in cancer, poor immunity against transformed self fails to control tumor growth. Blockade of pathways that preserve self-tolerance is being leveraged to unleash immunity against many tumors; however, widespread success is hindered by the autoimmune-like toxicities that arise in treated patients. Knowledge gained from the treatment of autoimmunity can be leveraged to treat these toxicities in patients. Further, the understanding of how T cell dysfunction arises in cancer can be leveraged to induce a similar state in autoreactive T cells. Here, we review what is known about the T cell response in autoimmunity and cancer and highlight ways in which we can learn from the nexus of these two diseases to improve the application, efficacy, and management of immunotherapies.
Collapse
Affiliation(s)
- Davide Mangani
- Evergrande Center for Immunologic Diseases, Ann Romney Center for Neurologic Diseases, Harvard Medical School and Mass General Brigham, Boston, MA 02115, USA; Institute for Research in Biomedicine, Faculty of Biomedical Sciences, Universita della Svizzera Italiana, Bellinzona 6500, Switzerland.
| | - Dandan Yang
- Evergrande Center for Immunologic Diseases, Ann Romney Center for Neurologic Diseases, Harvard Medical School and Mass General Brigham, Boston, MA 02115, USA
| | - Ana C Anderson
- Evergrande Center for Immunologic Diseases, Ann Romney Center for Neurologic Diseases, Harvard Medical School and Mass General Brigham, Boston, MA 02115, USA.
| |
Collapse
|
4
|
Lees JR. CD8+ T cells: The past and future of immune regulation. Cell Immunol 2020; 357:104212. [PMID: 32979764 DOI: 10.1016/j.cellimm.2020.104212] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 08/16/2020] [Accepted: 09/01/2020] [Indexed: 02/05/2023]
Abstract
Regulation of the adaptive immune response is critical for health. Regulatory activity can be found in multiple components of the immune system, however, the focus on particular components of the immune regulatory network has left many aspects of this critical immune component understudied. Here we review the evidence for activities of CD8+ T cells in immune homeostasis and regulation of autoimmune reactivity. The heterogeneous nature of identified CD8+ cell types are examined, and common phenotypes associated with functional activities are defined. The varying types of antigen signal crucial for CD8+ T cell regulatory activity are identified and the implications of these activation pathways for control of adaptive responses is considered. Finally, the promising capacity for transgenic antigen receptor directed cytotoxicity as a mechanism for modulation of autoimmunity is detailed.
Collapse
Affiliation(s)
- Jason R Lees
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States.
| |
Collapse
|
5
|
Benkhoucha M, Senoner I, Lalive PH. c-Met is expressed by highly autoreactive encephalitogenic CD8+ cells. J Neuroinflammation 2020; 17:68. [PMID: 32075650 PMCID: PMC7031922 DOI: 10.1186/s12974-019-1676-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 12/16/2019] [Indexed: 12/29/2022] Open
Abstract
Background CD8+ T lymphocytes are critical mediators of neuroinflammatory diseases. Understanding the mechanisms that govern the function of this T cell population is crucial to better understanding central nervous system autoimmune disease pathology. We recently identified a novel population of highly cytotoxic c-Met-expressing CD8+ T lymphocytes and found that hepatocyte growth factor (HGF) limits effective murine cytotoxic T cell responses in cancer models. Here, we examined the role of c-Met-expressing CD8+ T cells by using a MOG35–55 T cell-mediated EAE model. Methods Mice were subcutaneously immunized with myelin oligodendrocyte glycoprotein peptide (MOG)35–55 in complete Freund’s adjuvant (CFA). Peripheral and CNS inflammation was evaluated at peak disease and chronic phase, and c-Met expression by CD8 was evaluated by flow cytometry and immunofluorescence. Molecular, cellular, and killing function analysis were performed by real-time PCR, ELISA, flow cytometry, and killing assay. Results In the present study, we observed that a fraction of murine effector CD8+ T cells expressed c-Met receptor (c-Met+CD8+) in an experimental autoimmune encephalitis (EAE) model. Phenotypic and functional analysis of c-Met+CD8+ T cells revealed that they recognize the encephalitogenic epitope myelin oligodendrocyte glycoprotein37–50. We demonstrated that this T cell population produces higher levels of interferon-γ and granzyme B ex vivo and that HGF directly restrains the cytolytic function of c-Met+CD8+ T cells in cell-mediated cytotoxicity reactions Conclusions Altogether, our findings suggest that the HGF/c-Met pathway could be exploited to modulate CD8+ T cell-mediated neuroinflammation.
Collapse
Affiliation(s)
- Mahdia Benkhoucha
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Isis Senoner
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Patrice H Lalive
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland. .,Department of Neurosciences, Division of Neurology, University Hospital of Geneva, Geneva, Switzerland.
| |
Collapse
|
6
|
The immunologic considerations in human head transplantation. Int J Surg 2017; 41:196-202. [PMID: 28130190 DOI: 10.1016/j.ijsu.2017.01.084] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 01/19/2017] [Accepted: 01/20/2017] [Indexed: 11/24/2022]
Abstract
The idea of head transplantation appears at first as unrealistic, unethical, and futile. Here we discuss immunological considerations in human head transplantation. In a separate accompanying article we discuss surgical, ethical, and psychosocial issues concerned in body-to-head transplantation (BHT) [1]. The success of such an unusual allograft, where the donor and the recipient can reject each other, depends on prevention of complex immunologic reactions, especially rejection of the head by the body (graft-vs-host) or probably less likely, the possibility of the head rejecting the total body allograft (host-vs-graft). The technical and immunologic difficulties are enormous, especially since rapid nerve and cord connections and regeneration have not yet been possible to achieve. In this article we begin by briefly reviewing neuro-immunologic issues that may favor BHT such as the blood brain barrier (BBB) and point out its shortcomings. And we touch on the cellular and humoral elements in the brain proper that differ in some respects from those in other organs and in the periphery. Based on recent successes in vascular composite allografts (VCAs), we will elaborate on potential specific advantages and difficulties in BHT of various available immunosuppressive medications already utilized in VCAs. The risk/benefit ratio of these drugs will be emphasized in relation to direct brain toxicity such as seizure disorders, interference, or promotion of nerve regeneration, and potentiation of cerebral viral infections. The final portion of this article will focus on pre-transplant immunologic manipulation of the deceased donor body along with pretreatment of the recipient.
Collapse
|
7
|
Hohlfeld R, Dornmair K, Meinl E, Wekerle H. The search for the target antigens of multiple sclerosis, part 2: CD8+ T cells, B cells, and antibodies in the focus of reverse-translational research. Lancet Neurol 2015; 15:317-31. [PMID: 26724102 DOI: 10.1016/s1474-4422(15)00313-0] [Citation(s) in RCA: 143] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2015] [Revised: 10/13/2015] [Accepted: 10/22/2015] [Indexed: 01/16/2023]
Abstract
Interest in CD8+ T cells and B cells was initially inspired by observations in multiple sclerosis rather than in animal models: CD8+ T cells predominate in multiple sclerosis lesions, oligoclonal immunoglobulin bands in CSF have long been recognised as diagnostic and prognostic markers, and anti-B-cell therapies showed considerable efficacy in multiple sclerosis. Taking a reverse-translational approach, findings from human T-cell receptor (TCR) and B-cell receptor (BCR) repertoire studies provided strong evidence for antigen-driven clonal expansion in the brain and CSF. New methods allow the reconstruction of human TCRs and antibodies from tissue-infiltrating immune cells, which can be used for the unbiased screening of antigen libraries. Myelin oligodendrocyte glycoprotein (MOG) has received renewed attention as an antibody target in childhood multiple sclerosis and in a small subgroup of adult patients with multiple sclerosis. Furthermore, there is growing evidence that a separate condition in adults exists, tentatively called MOG-antibody-associated encephalomyelitis, which has clinical features that overlap with neuromyelitis optica spectrum disorder and multiple sclerosis. Although CD8+ T cells and B cells are thought to have a pathogenic role in some subgroups of patients, their target antigens have yet to be identified.
Collapse
Affiliation(s)
- Reinhard Hohlfeld
- Institute of Clinical Neuroimmunology, Biomedical Center and University Hospital, Campus Martinsried-Grosshadern, Ludwig-Maximilians University, Munich, Germany; Munich Cluster of Systems Neurology (SyNergy), Munich, Germany.
| | - Klaus Dornmair
- Institute of Clinical Neuroimmunology, Biomedical Center and University Hospital, Campus Martinsried-Grosshadern, Ludwig-Maximilians University, Munich, Germany
| | - Edgar Meinl
- Institute of Clinical Neuroimmunology, Biomedical Center and University Hospital, Campus Martinsried-Grosshadern, Ludwig-Maximilians University, Munich, Germany
| | - Hartmut Wekerle
- HERTIE Senior Professor Group Neuroimmunology, Max Planck Institute of Neurobiology, Martinsried, Germany
| |
Collapse
|
8
|
Cross-recognition of a myelin peptide by CD8+ T cells in the CNS is not sufficient to promote neuronal damage. J Neurosci 2015; 35:4837-50. [PMID: 25810515 DOI: 10.1523/jneurosci.3380-14.2015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Multiple sclerosis (MS) is an inflammatory disease of the CNS thought to be driven by CNS-specific T lymphocytes. Although CD8(+) T cells are frequently found in multiple sclerosis lesions, their distinct role remains controversial because direct signs of cytotoxicity have not been confirmed in vivo. In the present work, we determined that murine ovalbumin-transgenic (OT-1) CD8(+) T cells recognize the myelin peptide myelin oligodendrocyte glycoprotein 40-54 (MOG40-54) both in vitro and in vivo. The aim of this study was to investigate whether such cross-recognizing CD8(+) T cells are capable of inducing CNS damage in vivo. Using intravital two-photon microscopy in the mouse model of multiple sclerosis, we detected antigen recognition motility of the OT-1 CD8(+) T cells within the CNS leading to a selective enrichment in inflammatory lesions. However, this cross-reactivity of OT-1 CD8(+) T cells with MOG peptide in the CNS did not result in clinically or subclinically significant damage, which is different from myelin-specific CD4(+) Th17-mediated autoimmune pathology. Therefore, intravital imaging demonstrates that local myelin recognition by autoreactive CD8(+) T cells in inflammatory CNS lesions alone is not sufficient to induce disability or increase axonal injury.
Collapse
|
9
|
Phenotypic changes in immune cell subsets reflect increased infarct volume in male vs. female mice. Transl Stroke Res 2014; 4:554-63. [PMID: 24187596 DOI: 10.1007/s12975-013-0268-z] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Inflammatory responses in the brain after cerebral ischemia have been studied extensively in male mice, but not female mice, thus potentially giving a less-than-accurate view of gender associated pathological processes. In humans, cerebral infarcts are typically smaller in premenopausal females than in age-matched males. In the current study, we confirmed smaller infarcts in female vs. male mice after middle cerebral artery occlusion and 96 h of reperfusion. Moreover, we explored immunological alterations related to this difference and found that the percentage of CD4+ T lymphocytes was significantly higher in spleens in males than females, with increased expression of the activation markers, CD69 and CD44. In contrast, the percentage of CD8+ T lymphocytes was significantly higher in spleens of females than males, leading to the identification of a small but distinct population of IL-10-secreting CD8+CD122+ suppressor T cells that were also increased in females. Finally, we observed that males have a greater percentage of activated macrophages/microglia in the brain than females, as well as increased expression of the VLA-4 adhesion molecule in both brain and spleen. This new information suggesting gender-dependent immunological mechanisms in stroke implies that effective treatments for human stroke may also be gender specific.
Collapse
|
10
|
Leuenberger T, Paterka M, Reuter E, Herz J, Niesner RA, Radbruch H, Bopp T, Zipp F, Siffrin V. The role of CD8+ T cells and their local interaction with CD4+ T cells in myelin oligodendrocyte glycoprotein35-55-induced experimental autoimmune encephalomyelitis. THE JOURNAL OF IMMUNOLOGY 2013; 191:4960-8. [PMID: 24123686 DOI: 10.4049/jimmunol.1300822] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
T cells have an essential role in the induction of multiple sclerosis and its animal model experimental autoimmune encephalomyelitis (EAE). Although for CD4(+) T cells it is well established that they contribute to the disease, less is known about the role of CD8(+) T cells. Our aim was to determine the individual contribution of CD4(+) and CD8(+) T cells in myelin oligodendrocyte glycoprotein (MOG)35-55-induced EAE. We investigated MOG35-55-activated CD8(+) T cells to clarify their potential to induce or attenuate EAE. We monitored the behavior of CD8(+) T cells and their interaction with CD4(+) T cells directly at the site of inflammation in the CNS using intravital imaging of the brainstem of EAE-affected living anesthetized mice. We found that mice without CD4(+) T cells did not develop relevant clinical signs of disease, although CD8(+) T cells were present in the CNS of these mice. These CD8(+) T cells displayed reduced motility compared with those in the presence of CD4(+) T cells. In mice that harbored CD4(+) and CD8(+) T cells, we saw a similar extent of clinical signs of EAE as in mice with only CD4(+) T cells. Furthermore, the dynamic motility and viability of CD4(+) T cells were not disturbed by CD8(+) T cells in the lesions of these mice. Therefore, we conclude that in MOG35-55-induced EAE, CD8(+) T cell accumulation in the CNS represents instead an epiphenomenon with no impact on clinical disease or on the effects of CD4(+) T cells, the latter being the true inducers of the disease.
Collapse
Affiliation(s)
- Tina Leuenberger
- Department of Neurology, Focus Program Translational Neurosciences, Rhine Main Neuroscience Network, Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Luessi F, Siffrin V, Zipp F. Neurodegeneration in multiple sclerosis: novel treatment strategies. Expert Rev Neurother 2013; 12:1061-76; quiz 1077. [PMID: 23039386 DOI: 10.1586/ern.12.59] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
In recent years it has become clear that the neuronal compartment already plays an important role early in the pathology of multiple sclerosis (MS). Neuronal injury in the course of chronic neuroinflammation is a key factor in determining long-term disability in patients. Viewing MS as both inflammatory and neurodegenerative has major implications for therapy, with CNS protection and repair needed in addition to controlling inflammation. Here, the authors' review recently elucidated molecular insights into inflammatory neuronal/axonal pathology in MS and discuss the resulting options regarding neuroprotective and regenerative treatment strategies.
Collapse
Affiliation(s)
- Felix Luessi
- Focus Program Translational Neuroscience, Rhine Main Neuroscience Network, Department of Neurology, University Medical Center Mainz, Johannes Gutenberg University Mainz, Langenbeckstr 1, 55131 Mainz, Germany
| | | | | |
Collapse
|
12
|
Carvalheiro H, da Silva JAP, Souto-Carneiro MM. Potential roles for CD8+ T cells in rheumatoid arthritis. Autoimmun Rev 2013; 12:401-9. [DOI: 10.1016/j.autrev.2012.07.011] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2012] [Accepted: 07/18/2012] [Indexed: 02/06/2023]
|
13
|
Progesterone down-regulates spinal cord inflammatory mediators and increases myelination in experimental autoimmune encephalomyelitis. Neuroscience 2012; 226:40-50. [PMID: 23000619 DOI: 10.1016/j.neuroscience.2012.09.032] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2012] [Revised: 09/10/2012] [Accepted: 09/11/2012] [Indexed: 11/22/2022]
Abstract
In mice with experimental autoimmune encephalomyelitis (EAE) pretreatment with progesterone improves clinical signs and decreases the loss of myelin basic protein (MBP) and proteolipid protein (PLP) measured by immunohistochemistry and in situ hybridization. Presently, we analyzed if progesterone effects in the spinal cord of EAE mice involved the decreased transcription of local inflammatory mediators and the increased transcription of myelin proteins and myelin transcription factors. C57Bl/6 female mice were divided into controls, EAE and EAE receiving progesterone (100mg implant) 7 days before EAE induction. Tissues were collected on day 17 post-immunization. Real time PCR technology demonstrated that progesterone blocked the EAE-induced increase of the proinflammatory mediators tumor necrosis factor alpha (TNFα) and its receptor TNFR1, the microglial marker CD11b and toll-like receptor 4 (TLR4) mRNAs, and increased mRNA expression of PLP and MBP, the myelin transcription factors NKx2.2 and Olig1 and enhanced CC1+oligodendrocyte density respect of untreated EAE mice. Immunocytochemistry demonstrated decreased Iba1+microglial cells. Confocal microscopy demonstrated that TNFα colocalized with glial-fibrillary acidic protein+astrocytes and OX-42+microglial cells. Therefore, progesterone treatment improved the clinical signs of EAE, decreased inflammatory glial reactivity and increased myelination. Data suggest that progesterone neuroprotection involves the modulation of transcriptional events in the spinal cord of EAE mice.
Collapse
|
14
|
Bahri R, Bollinger A, Bollinger T, Orinska Z, Bulfone-Paus S. Ectonucleotidase CD38 demarcates regulatory, memory-like CD8+ T cells with IFN-γ-mediated suppressor activities. PLoS One 2012; 7:e45234. [PMID: 23028866 PMCID: PMC3444472 DOI: 10.1371/journal.pone.0045234] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2012] [Accepted: 08/20/2012] [Indexed: 01/16/2023] Open
Abstract
Regulatory CD8(+) T cells are critical for self-tolerance and restricting excessive immune responses. The variety of immune functions they fulfill, the heterogeneity of their phenotype, and the mechanism of action are still poorly understood. Here we describe that regulatory CD8(+) T cells exhibiting immunosuppressive actions in vitro and in vivo are recognized as CD38(high) T cells and present in naive mice. CD38 is a glycosylated membrane protein with ectonucleotidase properties. CD8(+)CD38(high) (CD44(+)CD122(+)CD62L(high)) lymphocytes suppress CD4(+) effector T-cell proliferation in an antigen-non specific manner via IFN-γ. While direct cell-to-cell contact is needed for this suppressor activity, it is independent of membrane-bound TGF-β and granzyme B release. IL-15 potentiates the suppressive activity of CD8(+)CD38(high) T cells and controls their survival and expansion. In humans CD8(+)CD38(high) T cells inhibit CD4(+) effector T cell proliferation. In vivo, CD8(+)CD38(high), but not CD8(+)CD38(-) T cells mitigate murine experimental autoimmune encephalomyelitis (EAE) by reducing the clinical score and delaying disease occurrence. EAE suppression is enhanced by pre-treatment of CD8(+)CD38(high) T cells with IL-15. These findings add evidence that the expression of ectoenzyme receptor family members positively correlates with suppressor functions and identifies CD8(+)CD38(high) T cells as potential inhibitors of excessive immune responses.
Collapse
MESH Headings
- ADP-ribosyl Cyclase 1/immunology
- ADP-ribosyl Cyclase 1/metabolism
- Animals
- Antigens, CD/immunology
- Antigens, CD/metabolism
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- CD8-Positive T-Lymphocytes/pathology
- Cell Communication/immunology
- Cell Proliferation
- Cell Survival
- Cells, Cultured
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Female
- Flow Cytometry
- Immunologic Memory
- Immunophenotyping
- Interferon-gamma/immunology
- Interleukin-15/immunology
- Lymphocyte Activation
- Membrane Glycoproteins/immunology
- Membrane Glycoproteins/metabolism
- Mice
- Mice, Inbred C57BL
- Nucleotidases/immunology
- Nucleotidases/metabolism
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- T-Lymphocytes, Regulatory/pathology
Collapse
Affiliation(s)
- Rajia Bahri
- Department of Immunology and Cell Biology, Research Center Borstel, Borstel, Germany
| | - Annalena Bollinger
- Department of Immunology and Cell Biology, Research Center Borstel, Borstel, Germany
| | - Thomas Bollinger
- Institut für Medizinische Mikrobiologie und Hygiene, University of Lübeck, Lübeck, Germany
| | - Zane Orinska
- Department of Immunology and Cell Biology, Research Center Borstel, Borstel, Germany
| | - Silvia Bulfone-Paus
- Department of Immunology and Cell Biology, Research Center Borstel, Borstel, Germany
- Faculty of Medical and Human Sciences, University of Manchester, Manchester, United Kingdom
- * E-mail:
| |
Collapse
|
15
|
The immunomodulatory and neuroprotective effects of mesenchymal stem cells (MSCs) in experimental autoimmune encephalomyelitis (EAE): a model of multiple sclerosis (MS). Int J Mol Sci 2012; 13:9298-9331. [PMID: 22942767 PMCID: PMC3430298 DOI: 10.3390/ijms13079298] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2012] [Revised: 07/11/2012] [Accepted: 07/11/2012] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent cells that differentiate into the mesenchymal lineages of adipocytes, osteocytes and chondrocytes. MSCs can also transdifferentiate and thereby cross lineage barriers, differentiating for example into neurons under certain experimental conditions. MSCs have anti-proliferative, anti-inflammatory and anti-apoptotic effects on neurons. Therefore, MSCs were tested in experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS), for their effectiveness in modulating the pathogenic process in EAE to develop effective therapies for MS. The data in the literature have shown that MSCs can inhibit the functions of autoreactive T cells in EAE and that this immunomodulation can be neuroprotective. In addition, MSCs can rescue neural cells via a mechanism that is mediated by soluble factors, which provide a suitable environment for neuron regeneration, remyelination and cerebral blood flow improvement. In this review, we discuss the effectiveness of MSCs in modulating the immunopathogenic process and in providing neuroprotection in EAE.
Collapse
|
16
|
Kim HJ, Cantor H. Regulation of self-tolerance by Qa-1-restricted CD8(+) regulatory T cells. Semin Immunol 2012; 23:446-52. [PMID: 22136694 DOI: 10.1016/j.smim.2011.06.001] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2011] [Accepted: 06/07/2011] [Indexed: 10/14/2022]
Abstract
Mounting an efficient immune response to pathogens while avoiding damage to host tissues is the central task of the immune system. Emerging evidence has highlighted the contribution of the CD8(+) lineage of regulatory T cells to the maintenance of self-tolerance. Specific recognition of the MHC class Ib molecule Qa-1 complexed to peptides expressed by activated CD4(+) T cells by regulatory CD8(+) T cells triggers an inhibitory interaction that prevents autoimmune responses. Conversely, defective Qa-1-restricted CD8(+) regulatory activity can result in development of systemic autoimmune disease. Here, we review recent research into the cellular and molecular basis of these regulatory T cells, their mechanism of suppressive activity and the potential application of these insights into new treatments for autoimmune disease and cancer.
Collapse
Affiliation(s)
- Hye-Jung Kim
- Department of Cancer Immunology & AIDS, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA 02215, USA
| | | |
Collapse
|
17
|
Ifergan I, Kebir H, Alvarez JI, Marceau G, Bernard M, Bourbonnière L, Poirier J, Duquette P, Talbot PJ, Arbour N, Prat A. Central nervous system recruitment of effector memory CD8+ T lymphocytes during neuroinflammation is dependent on α4 integrin. Brain 2011; 134:3560-77. [PMID: 22058139 PMCID: PMC7110084 DOI: 10.1093/brain/awr268] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Clonally expanded CD8+ T lymphocytes are present in multiple sclerosis lesions, as well as in the cerebrospinal fluid of patients with multiple sclerosis. In experimental autoimmune encephalomyelitis, CD8+ T lymphocytes are found in spinal cord and brainstem lesions. However, the exact phenotype of central nervous system-infiltrating CD8+ T lymphocytes and the mechanism by which these cells cross the blood–brain barrier remain largely unknown. Using cerebrospinal fluid from patients with multiple sclerosis, spinal cord from experimental autoimmune encephalomyelitis and coronavirus-induced encephalitis, we demonstrate that central nervous system-infiltrating CD8+ T lymphocytes are mostly of the effector memory phenotype (CD62L− CCR7− granzymeBhi). We further show that purified human effector memory CD8+ T lymphocytes transmigrate more readily across blood-brain barrier-endothelial cells than non-effector memory CD8+ T lymphocytes, and that blood-brain barrier endothelium promotes the selective recruitment of effector memory CD8+ T lymphocytes. Furthermore, we provide evidence for the recruitment of interferon-γ- and interleukin-17-secreting CD8+ T lymphocytes by human and mouse blood-brain barrier endothelium. Finally, we show that in vitro migration of CD8+ T lymphocytes across blood-brain barrier-endothelial cells is dependent on α4 integrin, but independent of intercellular adhesion molecule-1/leucocyte function-associated antigen-1, activated leucocyte cell adhesion molecule/CD6 and the chemokine monocyte chemotactic protein-1/CCL2. We also demonstrate that in vivo neutralization of very late antigen-4 restricts central nervous system infiltration of CD8+ T lymphocytes in active immunization and adoptive transfer experimental autoimmune encephalomyelitis, and in coronavirus-induced encephalitis. Our study thus demonstrates an active role of the blood-brain barrier in the recruitment of effector memory CD8+ T lymphocytes to the CNS compartment and defines α4 integrin as a major contributor of CD8+ T lymphocyte entry into the brain.
Collapse
Affiliation(s)
- Igal Ifergan
- Neuroimmunology Research Unit, Centre for Excellence in Neuromics, CRCHUM-Notre-Dame Hospital, Université de Montréal, Montréal, QC, H2L 4M1, Canada
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Sajic M, Gregson N, Linington C, Hughes RAC, Smith KJ. The role of CD8(+) T cells in a model of multiple sclerosis induced with recombinant myelin oligodendrocyte glycoprotein. Mult Scler 2011; 18:286-98. [PMID: 21952095 DOI: 10.1177/1352458511424309] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND AND OBJECTIVES Since CD8(+) T cells may be important in the pathogenesis of multiple sclerosis (MS), we examined their role in the DA rat experimental autoimmune encephalomyelitis (EAE) model induced by immunization with recombinant myelin oligodendrocyte glycoprotein (rMOG). METHODS The inflammatory infiltrate in the spinal cord of affected animals was assessed by histology, electrophysiology and flow cytometry during the course of the disease (the first peak, remission and the second peak). The proportions of activated/memory effector (CD8(+)CD44(+)) and putative suppressor (CD8(+)CD28(-), CD8(+)CD25(high)) CD8(+) T cells in the draining lymph nodes were determined. To explore the role of CD8(+) T cells, similar experiments were performed in CD8(+) T cell depleted rats, before, during and after the first peak of the disease. RESULTS Throughout the disease, both CD4(+) T cells and macrophages/activated microglia outnumbered CD8(+) T cells within the spinal cord. The number of putative suppressor CD8(+) T cells increased significantly both during and after the first peak suggesting the induction of a regulatory CD8(+) T-cell response. However, antibody-mediated depletion of CD8(+) T cells before induction of the disease, or after the first peak, did not significantly alter the incidence, severity or course of rMOG-induced EAE. CONCLUSIONS The findings suggest that CD8(+) T cells do not play a significant role in the pathogenesis or regulation of EAE induced by rMOG in DA rats. In this respect, rMOG-induced EAE is not an appropriate model for studying the role of CD8(+) T cells in MS.
Collapse
Affiliation(s)
- Marija Sajic
- Department of Neuroinflammation, UCL Institute of Neurology, London, UK.
| | | | | | | | | |
Collapse
|
19
|
Kim YH, Choi BK, Shin SM, Kim CH, Oh HS, Park SH, Lee DG, Lee MJ, Kim KH, Vinay DS, Kwon BS. 4-1BB triggering ameliorates experimental autoimmune encephalomyelitis by modulating the balance between Th17 and regulatory T cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2011; 187:1120-8. [PMID: 21715692 DOI: 10.4049/jimmunol.1002681] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Agonistic anti-4-1BB Ab is known to ameliorate experimental autoimmune encephalomyelitis. 4-1BB triggering typically leads to the expansion of CD8(+) T cells, which produce abundant IFN-γ, and this in turn results in IDO-dependent suppression of autoimmune responses. However, because neutralization of IFN-γ or depletion of CD8(+) T cell only partially abrogates the effect of 4-1BB triggering, we sought to identify an additional mechanism of 4-1BB-triggered suppression of autoimmune responses using IFN-γ- or IFN-γR-deficient mice. 4-1BB triggering inhibited the generation of Th17 cells that is responsible for experimental autoimmune encephalomyelitis induction and progression, and increased Foxp3(+)CD4(+) regulatory T (Treg) cells, particularly among CD4(+) T cells. This was not due to a direct effect of 4-1BB signaling on CD4(+) T cell differentiation: 4-1BB signaling not only reduced Th17 cells and increased Treg cells in wild-type mice, which could be due to IFN-γ production by the CD8(+) T cells, but also did so in IFN-γ-deficient mice, in that case by downregulating IL-6 production. These results show that although secondary suppressive mechanisms evoked by 4-1BB triggering are usually masked by the strong effects of IFN-γ, 4-1BB signaling seems to modulate autoimmune responses by a number of mechanisms, and modulation of the Th17 versus Treg cell balance is one of those mechanisms.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- CD4 Lymphocyte Count
- Cell Differentiation/genetics
- Cell Differentiation/immunology
- Cells, Cultured
- Disease Progression
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/therapy
- Interferon-gamma/deficiency
- Interferon-gamma/metabolism
- Interferon-gamma/physiology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Molecular Sequence Data
- Receptors, Interferon/deficiency
- Receptors, Interferon/genetics
- Receptors, Interferon/physiology
- Signal Transduction/genetics
- Signal Transduction/immunology
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- T-Lymphocytes, Regulatory/pathology
- Th17 Cells/immunology
- Th17 Cells/metabolism
- Th17 Cells/pathology
- Tumor Necrosis Factor Receptor Superfamily, Member 9/antagonists & inhibitors
- Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology
- Tumor Necrosis Factor Receptor Superfamily, Member 9/physiology
- Interferon gamma Receptor
Collapse
Affiliation(s)
- Young H Kim
- Immune and Cell Therapy Branch, Division of Cancer Biology, National Cancer Center, Goyang-si, Gyeongi-do 410-769, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Jiang H, Canfield SM, Gallagher MP, Jiang HH, Jiang Y, Zheng Z, Chess L. HLA-E-restricted regulatory CD8(+) T cells are involved in development and control of human autoimmune type 1 diabetes. J Clin Invest 2010; 120:3641-50. [PMID: 20877010 PMCID: PMC2947239 DOI: 10.1172/jci43522] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2010] [Accepted: 08/18/2010] [Indexed: 11/17/2022] Open
Abstract
A key feature of the immune system is its ability to discriminate self from nonself. Breakdown in any of the mechanisms that maintain unresponsiveness to self (a state known as self-tolerance) contributes to the development of autoimmune conditions. Recent studies in mice show that CD8(+) T cells specific for the unconventional MHC class I molecule Qa-1 bound to peptides derived from the signal sequence of Hsp60 (Hsp60sp) contribute to self/nonself discrimination. However, it is unclear whether they exist in humans and play a role in human autoimmune diseases. Here we have shown that CD8(+) T cells specific for Hsp60sp bound to HLA-E (the human homolog of Qa-1) exist and play an important role in maintaining peripheral self-tolerance by discriminating self from nonself in humans. Furthermore, in the majority of type 1 diabetes (T1D) patients tested, there was a specific defect in CD8(+) T cell recognition of HLA-E/Hsp60sp, which was associated with failure of self/nonself discrimination. However, the defect in the CD8(+) T cells from most of the T1D patients tested could be corrected in vitro by exposure to autologous immature DCs loaded with the Hsp60sp peptide. These data suggest that HLA-E-restricted CD8(+) T cells may play an important role in keeping self-reactive T cells in check. Thus, correction of this defect could be a potentially effective and safe approach in the therapy of T1D.
Collapse
Affiliation(s)
- Hong Jiang
- Department of Medicine, Naomi Berrie Diabetes Center (NBDC), Columbia University, College of Physicians and Surgeons, New York, New York 10032, USA.
| | | | | | | | | | | | | |
Collapse
|
21
|
Mars LT, Saikali P, Liblau RS, Arbour N. Contribution of CD8 T lymphocytes to the immuno-pathogenesis of multiple sclerosis and its animal models. Biochim Biophys Acta Mol Basis Dis 2010; 1812:151-61. [PMID: 20637863 DOI: 10.1016/j.bbadis.2010.07.006] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2009] [Revised: 06/21/2010] [Accepted: 07/06/2010] [Indexed: 12/17/2022]
Abstract
Multiple sclerosis (MS) is an inflammatory disease of the central nervous system (CNS) characterized by multi-focal demyelination, axonal loss, and immune cell infiltration. Numerous immune mediators are detected within MS lesions, including CD4(+) and CD8(+) T lymphocytes suggesting that they participate in the related pathogenesis. Although CD4(+) T lymphocytes are traditionally considered the main actors in MS immunopathology, multiple lines of evidence suggest that CD8(+) T lymphocytes are also implicated in the pathogenesis. In this review, we outline the recent literature pertaining to the potential roles of CD8(+) T lymphocytes both in MS and its animal models. The CD8(+) T lymphocytes detected in MS lesions demonstrate characteristics of activated and clonally expanded cells supporting the notion that these cells actively contribute to the observed injury. Moreover, several experimental in vivo models mediated by CD8(+) T lymphocytes recapitulate important features of the human disease. Whether the CD8(+) T cells can induce or aggravate tissue destruction in the CNS needs to be fully explored. Strengthening our understanding of the pathogenic potential of CD8(+) T cells in MS should provide promising new avenues for the treatment of this disabling inflammatory disease.
Collapse
Affiliation(s)
- Lennart T Mars
- INSERM, U563, Centre de Physiopathologie de Toulouse Purpan, Hôpital Purpan, Toulouse, F-31300, France; Université Toulouse III, Paul-Sabatier, Toulouse, F-31400, France
| | | | | | | |
Collapse
|
22
|
Chen ML, Yan BS, Kozoriz D, Weiner HL. Novel CD8+ Treg suppress EAE by TGF-beta- and IFN-gamma-dependent mechanisms. Eur J Immunol 2009; 39:3423-35. [PMID: 19768696 PMCID: PMC2814307 DOI: 10.1002/eji.200939441] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Although CD8+ Treg-mediated suppression has been described, CD8+ Treg remain poorly characterized. Here we identify a novel subset of CD8+ Treg that express latency-associated peptide (LAP) on their cell surface (CD8+LAP+ cells) and exhibit regulatory activity in vitro and in vivo. Only a small fraction of CD8+LAP+ cells express Foxp3 or CD25, although the expression levels of Foxp3 for these cells are higher than their LAP- counterparts. In addition to TGF-beta, CD8+LAP+ cells produce IFN-gamma, and these cells suppress EAE that is dependent on both TGF-beta and IFN-gamma. In an adoptive co-transfer model, CD8+LAP+ cells suppress myelin oligodendrocyte glycoprotein (MOG)-specific immune responses by inducing or expanding Foxp3+ cells and by inhibiting proliferation and IFN-gamma production in vivo. Furthermore, in vivo neutralization of IFN-gamma and studies with IFN-gamma-deficient mice demonstrate an important role for IFN-gamma production in the function of CD8+LAP+ cells. Our findings identify the underlying mechanisms that account for the immunoregulatory activity of CD8+ T cells and suggest that induction or amplification of CD8+LAP+ cells may be a therapeutic strategy to help control autoimmune processes.
Collapse
MESH Headings
- Adoptive Transfer
- Analysis of Variance
- Animals
- CD8 Antigens/metabolism
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- CD8-Positive T-Lymphocytes/transplantation
- Cytokines/metabolism
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Enzyme-Linked Immunosorbent Assay
- Female
- Forkhead Transcription Factors/metabolism
- Glycoproteins/immunology
- Interferon-gamma/genetics
- Interferon-gamma/metabolism
- Interleukin-2 Receptor alpha Subunit/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Inbred Strains
- Mice, Knockout
- Myelin Proteins
- Myelin-Associated Glycoprotein/chemistry
- Myelin-Associated Glycoprotein/immunology
- Myelin-Oligodendrocyte Glycoprotein
- Peptide Fragments/immunology
- Receptors, Interferon/genetics
- Receptors, Interferon/metabolism
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- T-Lymphocytes, Regulatory/transplantation
- Transforming Growth Factor beta/genetics
- Transforming Growth Factor beta/metabolism
- Interferon gamma Receptor
Collapse
Affiliation(s)
- Mei-Ling Chen
- Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | | | | | | |
Collapse
|
23
|
Schreiner B, Heppner FL, Becher B. Modeling multiple sclerosis in laboratory animals. Semin Immunopathol 2009; 31:479-95. [PMID: 19802608 DOI: 10.1007/s00281-009-0181-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2009] [Accepted: 08/13/2009] [Indexed: 12/18/2022]
Abstract
Inflammatory demyelinating disease of the central nervous system is one of the most frequent causes of neurological disability in young adults. While in situ analysis and in vitro models do shed some light onto the processes of tissue damage and cellular interactions, the development of neuroinflammation and demyelination is a far too complex process to be adequately modeled by simple test tube systems. Thus, animal models using primarily genetically modified mice have been proven to be of paramount importance. In this chapter, we discuss recent advances in modeling brain diseases focusing on murine models and report on new tools to study the pathogenesis of complex diseases such as multiple sclerosis.
Collapse
|
24
|
Abstract
Autoreactive T cell responses have a crucial role in central nervous system (CNS) diseases such as multiple sclerosis. Recent data indicate that CNS autoimmunity can be mediated by two distinct lineages of CD4+ T cells that are defined by the production of either interferon-gamma or interleukin-17. The activity of these CD4+ T cell subsets within the CNS influences the pathology and clinical course of disease. New animal models show that myelin-specific CD8+ T cells can also mediate CNS autoimmunity. This Review focuses on recent progress in delineating the pathogenic mechanisms, regulation and interplay between these different T cell subsets in CNS autoimmunity.
Collapse
Affiliation(s)
- Joan Goverman
- Department of Immunology, University of Washington, Seattle, Washington 98195-7650, USA.
| |
Collapse
|
25
|
Kiesel JR, Buchwald ZS, Aurora R. Cross-Presentation by Osteoclasts Induces FoxP3 in CD8+T Cells. THE JOURNAL OF IMMUNOLOGY 2009; 182:5477-87. [DOI: 10.4049/jimmunol.0803897] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
26
|
Wu Y, Zheng Z, Jiang Y, Chess L, Jiang H. The specificity of T cell regulation that enables self-nonself discrimination in the periphery. Proc Natl Acad Sci U S A 2009; 106:534-9. [PMID: 19118203 PMCID: PMC2613037 DOI: 10.1073/pnas.0811843106] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2008] [Indexed: 11/18/2022] Open
Abstract
It was recently shown that perceiving the avidity of T cell activation can be translated into peripheral T cell regulation to control autoimmune disease. This regulation is achieved by CD8(+) T cells that recognize a common surrogate target structure, Qa-1/Hsp60sp, preferentially expressed by activated T cells of intermediate but not high avidity. A truncated self-reactive repertoire, devoid of high-avidity T cells, generated by thymic negative selection, allows selective down-regulation of intermediate-avidity T cells to accomplish self-nonself discrimination in the periphery. Identification of the common surrogate target structure expressed on intermediate-avidity T cells opens up a conceptual theme to understand the relationship between the specificity of peripheral immune regulation and self-nonself discrimination. Here, we investigated peptide vaccination induced cross-protection mediated by CD8(+) T cells in two autoimmune disease models, experimental allergic encephalomyelitis (EAE) and type 1 diabetes (T1D). We show that Qa-1 restricted CD8(+) T cells cross-protect animals from either EAE or T1D without abrogating the immune response to foreign antigens. Cross-protection occurs because potentially pathogenic self-reactive T cells included in the pool of intermediate-avidity T cells are capable of preferentially expressing common surrogate target structures on their surface to render themselves subject to the down-regulation, independent of the specificity of the antigens that they are triggered by. Thus, like in the thymus, the immune system discriminates self from nonself, during adaptive immunity in the periphery, not by recognizing the structural differences between self and foreign antigens, but rather by perceiving the avidity of T cell activation.
Collapse
Affiliation(s)
- Yilun Wu
- Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY 10032
| | - Zongyu Zheng
- Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY 10032
| | - Yihua Jiang
- Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY 10032
| | - Leonard Chess
- Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY 10032
| | - Hong Jiang
- Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY 10032
| |
Collapse
|
27
|
Jiang H, Chess L. How the immune system achieves self-nonself discrimination during adaptive immunity. Adv Immunol 2009; 102:95-133. [PMID: 19477320 DOI: 10.1016/s0065-2776(09)01202-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
We propose an "Avidity Model of Self-Nonself Discrimination" in which self-nonself discrimination is achieved by both central thymic selection and peripheral immune regulation. The conceptual framework that links these two events is the understanding that both in the thymus and in the periphery the survival or the fate of T cells is determined by the avidity of the interactions between T cell receptors (TCRs) on T cells, specific to any antigens and MHC/antigen peptides presented by antigen-presenting cells (APCs). We envision that the immune system achieves self-nonself discrimination, during adaptive immunity, not by recognizing the structural differences between self versus foreign antigens, but rather by perceiving the avidity of T cell activation. Intrathymic deletion of high avidity T cell clones responding to the majority of self-antigens generates a truncated peripheral self-reactive repertoire composed of mainly intermediate and low but devoid of high avidity T cells compared with the foreign-reactive repertoire. The existence of intermediate avidity self-reactive T cells in the periphery represents a potential danger of pathogenic autoimmunity inherited in each individual because potentially pathogenic self-reactive T cells are included in the pool of intermediate avidity T cells and can often be functionally activated to elicit autoimmune diseases. The distinct composition of peripheral T cell repertoires to self versus to foreign antigens provides a unique opportunity for the immune system to discriminate self from nonself, in the periphery, by selectively downregulating intermediate avidity T cells to both self and foreign antigens. Selective downregulation of the intermediate avidity T cell populations containing the potentially pathogenic self-reactive T cells enables the immune system to specifically control autoimmune diseases without damaging the effective anti-infection immunity, which is, largely, mediated by high avidity T cells specific to the infectious pathogens. In this regard, it has been recently shown that Qa-1-restricted CD8(+) T cells selectively downregulate intermediate avidity T cells, to both self and foreign antigens, and as a consequence, specifically dampen autoimmunity yet optimize the immune response to foreign antigens. Selective downregulation of intermediate avidity T cells is accomplished via specific recognition, by the Qa-1-restricted CD8(+) T cells, of particular Qa-1/self-peptide complexes, such as Qa-1/Hsp60sp, which function as a common surrogate target structure and preferentially expressed on the activated intermediate avidity T cells. This regulatory pathway thus represents one example of the peripheral mechanisms that the immune system evolved to complete self-nonself discrimination that is achieved, imperfectly, by thymic negative selection, in order to maintain self-tolerance. The conceptual framework of the "Avidity Model" differs from, but contains intellectual wisdom of certain conceptual elements of, the "Tunable Activation Thresholds Hypothesis," the "Danger Model," and the "Ergotypic Regulation Phenomenon." It provides a unified and simple paradigm to explain various seemingly unrelated biomedical problems inherent in immunological disorders that cannot be uniformly interpreted by any currently existing paradigms. The potential impact of the conceptual framework of the "Avidity Model" on our understanding of the development and control of commonly seen autoimmune diseases is also discussed.
Collapse
Affiliation(s)
- Hong Jiang
- Institute of Immunology, School of Medicine, Shanghai Jiaotong University, Shanghai, PR China
| | | |
Collapse
|
28
|
Regulation of CD8+ regulatory T cells: Interruption of the NKG2A-Qa-1 interaction allows robust suppressive activity and resolution of autoimmune disease. Proc Natl Acad Sci U S A 2008; 105:19420-5. [PMID: 19047627 DOI: 10.1073/pnas.0810383105] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Regulation of autoreactive CD4 T cells is essential to maintain self-tolerance and prevent autoimmune disease. Although CD8 T regulatory (Treg) cells that recognize self-peptides restricted by Qa-1 (HLA-E in humans) inhibit autoreactive CD4 cells and attenuate experimental autoimmune encephalomyelitis (EAE), the mechanism of this interaction is unclear. We generated Qa-1 mutant knock-in mice that impair Qa-1 binding to the T cell receptor (TCR) and CD94/NKG2A receptors. Analysis of these mice showed that TCR-dependent recognition of Qa-1-peptide complexes on target CD4 cells is essential for suppression by CD8 Treg cells. Further analysis revealed that genetic disruption of the Qa-1-CD94/NKG2A interaction unleashes robust CD8 Treg cell activity that completely abolishes development of EAE.
Collapse
|
29
|
Jiang H, Chess L. Qa-1/HLA-E-restricted regulatory CD8+ T cells and self-nonself discrimination: an essay on peripheral T-cell regulation. Hum Immunol 2008; 69:721-7. [PMID: 18822330 DOI: 10.1016/j.humimm.2008.08.279] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2008] [Revised: 08/10/2008] [Accepted: 08/12/2008] [Indexed: 01/14/2023]
Abstract
By discriminating self from nonself and controlling the magnitude and class of immune responses, the immune system mounts effective immunity against virtually any foreign antigens but avoids harmful immune responses to self. These are two equally important and related but distinct processes, which function in concert to ensure an optimal function of the immune system. Immunologically relevant clinical problems often occur because of failure of either process, especially the former. Currently, there is no unified conceptual framework to characterize the precise relationship between thymic negative selection and peripheral immune regulation, which is the basis for understanding self-non-self discrimination versus control of magnitude and class of immune responses. In this article, we explore a novel hypothesis of how the immune system discriminates self from nonself in the periphery during adaptive immunity. This hypothesis permits rational analysis of various seemingly unrelated biomedical problems inherent in immunologic disorders that cannot be uniformly interpreted by any currently existing paradigms. The proposed hypothesis is based on a unified conceptual framework of the "avidity model of peripheral T-cell regulation" that we originally proposed and tested, in both basic and clinical immunology, to understand how the immune system achieves self-nonself discrimination in the periphery.
Collapse
Affiliation(s)
- Hong Jiang
- Department of Medicine, College of Physicians & Surgeons, Columbia University, New York, NY, USA.
| | | |
Collapse
|
30
|
Perceiving the avidity of T cell activation can be translated into peripheral T cell regulation. Proc Natl Acad Sci U S A 2007; 104:20472-7. [PMID: 18077361 DOI: 10.1073/pnas.0709878104] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The structure recognized by regulatory T cells that enables them to discriminate self from nonself in the periphery is one of the central issues of regulatory T cell biology. A link between immunoregulation and self-nonself discrimination has emerged from experiments showing that Qa-1-restricted CD8(+) T cells selectively down-regulate target T cells activated by the intermediate avidity of their own T cell antigen receptor-ligand interactions. Because the peripheral self-reactive T cell repertoire is devoid of high-avidity T cells compared with the foreign-reactive repertoire, as a result of thymic negative selection, the selective down-regulation of intermediate but not high-avidity T cells enables the immune system to suppress autoimmunity without damaging the ongoing immune response to foreign pathogens. However, the molecular mechanism delineating how avidity of T cell activation is perceived by the regulatory T cells has not been elucidated. Here we show that a heat shock peptide (Hsp60sp), coupled with the MHC class Ib molecule Qa-1, is a surrogate target structure that is preferentially expressed at a higher level on the intermediate avidity T cells and specifically recognized by the Qa-1-restricted CD8(+) T cells. The biological significance of this observation was confirmed by the ability of Hsp60sp-loaded relevant dendritic cells to induce a Qa-1-restricted CD8(+) T cell-mediated protection from autoimmune encephalopathy in the experimental allergic encephalomyelitis model. Thus, perceiving the avidity of T cell activation can be translated into peripheral T cell regulation to discriminate self from nonself in the periphery to maintain self-tolerance.
Collapse
|
31
|
Menezes JS, van den Elzen P, Thornes J, Huffman D, Droin NM, Maverakis E, Sercarz EE. A public T cell clonotype within a heterogeneous autoreactive repertoire is dominant in driving EAE. J Clin Invest 2007; 117:2176-85. [PMID: 17627303 PMCID: PMC1906731 DOI: 10.1172/jci28277] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2006] [Accepted: 04/30/2007] [Indexed: 01/03/2023] Open
Abstract
Experimental autoimmune encephalomyelitis (EAE) is an animal model of multiple sclerosis. Immunization of B10.PL mice with the Ac1-9 peptide, the immunodominant determinant of myelin basic protein (MBP), produced a single episode of EAE followed by recovery and resistance to reinduction of disease. Using the CDR3 length spectratyping technique, we characterized the clonal composition of the Ac1-9-specific T cell repertoire from induction through onset and resolution of disease. Two clonally restricted subsets within a heterogeneous self-reactive repertoire were found in mouse lymph nodes, spleen, and spinal cord soon after immunization, before any sign of EAE. These clonotypes, designated BV8S2/BJ2S7 and BV16/BJ2S5, were present in all mice examined and thus considered public. BV8S2/BJ2S7 was found in far greater excess; was exclusively Th1 polarized; disappeared from the spinal cord, spleen, and lymph nodes concomitantly with recovery; and transferred disease to naive recipients. In contrast, BV16/BJ2S5 and numerous private clonotypes were either Th1 or Th2 and persisted following recovery. These results are consistent with the hypothesis that the public clonotype BV8S2/BJ2S7 is a driver of disease and necessary for its propagation.
Collapse
MESH Headings
- Animals
- Complementarity Determining Regions/genetics
- Complementarity Determining Regions/immunology
- Encephalomyelitis, Autoimmune, Experimental/chemically induced
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Epitopes, T-Lymphocyte/genetics
- Epitopes, T-Lymphocyte/immunology
- Mice
- Multiple Sclerosis/chemically induced
- Multiple Sclerosis/genetics
- Multiple Sclerosis/immunology
- Multiple Sclerosis/pathology
- Myelin Basic Protein/genetics
- Myelin Basic Protein/immunology
- Myelin Basic Protein/toxicity
- Organ Specificity/genetics
- Organ Specificity/immunology
- Peptide Fragments/genetics
- Peptide Fragments/immunology
- Peptide Fragments/toxicity
- Recovery of Function/genetics
- Recovery of Function/immunology
- Th1 Cells/immunology
- Th1 Cells/pathology
- Th2 Cells/immunology
- Th2 Cells/pathology
Collapse
Affiliation(s)
- Juscilene S. Menezes
- Division of Immune Regulation, Torrey Pines Institute for Molecular Studies, San Diego, California, USA.
La Jolla Institute for Allergy and Immunology, San Diego, California, USA
| | - Peter van den Elzen
- Division of Immune Regulation, Torrey Pines Institute for Molecular Studies, San Diego, California, USA.
La Jolla Institute for Allergy and Immunology, San Diego, California, USA
| | - Jordan Thornes
- Division of Immune Regulation, Torrey Pines Institute for Molecular Studies, San Diego, California, USA.
La Jolla Institute for Allergy and Immunology, San Diego, California, USA
| | - Donald Huffman
- Division of Immune Regulation, Torrey Pines Institute for Molecular Studies, San Diego, California, USA.
La Jolla Institute for Allergy and Immunology, San Diego, California, USA
| | - Nathalie M. Droin
- Division of Immune Regulation, Torrey Pines Institute for Molecular Studies, San Diego, California, USA.
La Jolla Institute for Allergy and Immunology, San Diego, California, USA
| | - Emanual Maverakis
- Division of Immune Regulation, Torrey Pines Institute for Molecular Studies, San Diego, California, USA.
La Jolla Institute for Allergy and Immunology, San Diego, California, USA
| | - Eli E. Sercarz
- Division of Immune Regulation, Torrey Pines Institute for Molecular Studies, San Diego, California, USA.
La Jolla Institute for Allergy and Immunology, San Diego, California, USA
| |
Collapse
|
32
|
|
33
|
Izawa A, Yamaura K, Albin MJ, Jurewicz M, Tanaka K, Clarkson MR, Ueno T, Habicht A, Freeman GJ, Yagita H, Abdi R, Pearson T, Greiner DL, Sayegh MH, Najafian N. A novel alloantigen-specific CD8+PD1+ regulatory T cell induced by ICOS-B7h blockade in vivo. THE JOURNAL OF IMMUNOLOGY 2007; 179:786-96. [PMID: 17617568 DOI: 10.4049/jimmunol.179.2.786] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Delayed ICOS-B7h signal blockade promotes significant prolongation of cardiac allograft survival in wild-type but not in CD8-deficient C57BL/6 recipients of fully MHC-mismatched BALB/c heart allografts, suggesting the possible generation of CD8(+) regulatory T cells in vivo. We now show that the administration of a blocking anti-ICOS mAb results in the generation of regulatory CD8(+) T cells. These cells can transfer protection and prolong the survival of donor-specific BALB/c, but not third party C3H, heart grafts in CD8-deficient C57BL/6 recipients. This is unique to ICOS-B7h blockade, because B7 blockade by CTLA4-Ig prolongs graft survival in CD8-deficient mice and does not result in the generation of regulatory CD8(+) T cells. Those cells localize to the graft, produce both IFN-gamma and IL-4 after allostimulation in vitro, prohibit the expansion of alloreactive CD4(+) T cells, and appear to mediate a Th2 switch of recipient CD4(+) T cells after adoptive transfer in vivo. Finally, these cells are not confined to the CD28-negative population but express programmed death 1, a molecule required for their regulatory function in vivo. CD8(+)PD1(+) T cells suppress alloreactive CD4(+) T cells but do not inhibit the functions by alloreactive CD8(+) T cells in vitro. These results describe a novel allospecific regulatory CD8(+)PD1(+) T cell induced by ICOS-B7h blockade in vivo.
Collapse
Affiliation(s)
- Atsushi Izawa
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Fazilleau N, Delarasse C, Motta I, Fillatreau S, Gougeon ML, Kourilsky P, Pham-Dinh D, Kanellopoulos JM. T cell repertoire diversity is required for relapses in myelin oligodendrocyte glycoprotein-induced experimental autoimmune encephalomyelitis. THE JOURNAL OF IMMUNOLOGY 2007; 178:4865-75. [PMID: 17404267 DOI: 10.4049/jimmunol.178.8.4865] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Comparison of TCRalphabeta repertoires of myelin oligodendrocyte glycoprotein (MOG)-specific T lymphocytes in C57BL/6 and TdT-deficient littermates (TdT(-/-)) generated during experimental autoimmune encephalomyelitis (EAE) highlights a link between a diversified TCRalphabeta repertoire and EAE relapses. At the onset of the disease, the EAE-severity is identical in TdT(+/-) and TdT(-/-) mice and the neuropathologic public MOG-specific T cell repertoires express closely similar public Valpha-Jalpha and Vbeta-Jbeta rearrangements in both strains. However, whereas TdT(+/+) and TdT(+/-) mice undergo successive EAE relapses, TdT(-/-) mice recover definitively and the lack of relapses does not stem from dominant regulatory mechanisms. During the first relapse of the disease in TdT(+/-) mice, new public Valpha-Jalpha and Vbeta-Jbeta rearrangements emerge that are distinct from those detected at the onset of the disease. Most of these rearrangements contain N additions and are found in CNS-infiltrating T lymphocytes. Furthermore, CD4(+) T splenocytes bearing these rearrangements proliferate to the immunodominant epitope of MOG and not to other immunodominant epitopes of proteolipid protein and myelin basic protein autoantigens, excluding epitope spreading to these myelin proteins. Thus, in addition to epitope spreading, a novel mechanism involving TCRalphabeta repertoire diversification contributes to autoimmune progression.
Collapse
Affiliation(s)
- Nicolas Fazilleau
- Institut National de la Santé et de la Recherche Médicale, Unité 277, Institut Pasteur, Paris, France
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system that is believed to have an autoimmune origin. CD4(+) T cells have been well studied for their involvement in the pathogenesis of MS and its animal model, experimental autoimmune encephalomyelitis (EAE). CD8(+) T cells, however, have been overlooked until recently, when more attention has focused on their potential role in pathogenic mechanisms in MS. Here we summarize our work in generating a CD8(+) T cell-mediated EAE model. We discuss immune tolerance mechanisms that regulate CD8(+) T cells specific for myelin basic protein (MBP), and describe initial results regarding triggers of CD8(+) T cell-mediated disease. The availability of CD8(+) T cell-mediated EAE models will help to elucidate the pathogenic roles of CD8(+) T cells in MS, and provide tools for development of novel therapies for MS.
Collapse
Affiliation(s)
- Qingyong Ji
- Department of Immunology, University of Washington, 1959 NE Pacific Street Seattle, WA 98195-7650, USA
| | | |
Collapse
|
36
|
Abstract
The immune system is not only well equipped to control infections but also tightly controlled to prevent autoimmune disease. Despite the negative selection of T-cell clones in the thymus, mature T cells capable of recognizing self-antigens are present in every individual. Several types of specialized regulatory cells maintain homeostasis and prevent expansion of autoreactive T cells. In this issue of Immunological Reviews, the role of CD4+ regulatory T cells is extensively discussed. Suppression of T-cell responses by CD8+ T cells has received less attention. Here, we review research on Qa-1-restricted CD8+ regulatory T cells. We focus on the role of this class Ib major histocompatibility complex (MHC) molecule in both CD8+ regulatory T-cell activity and protection of activated T cells.
Collapse
Affiliation(s)
- Linrong Lu
- Department of Cancer Immunology & AIDS, Dana Farber Cancer Institute, Boston, MA, USA
| | | | | |
Collapse
|
37
|
Marrero I, Huffman D, Kalil J, Sercarz EE, Coelho V. TAP1-/- mice present oligoclonal BV-BJ expansions following the rejection of grafts bearing self antigens. Immunology 2006; 118:461-71. [PMID: 16895555 PMCID: PMC1782321 DOI: 10.1111/j.1365-2567.2006.02387.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Our previous work showed that transporter associated with antigen processing 1 (TAP1)-/- (H-2b) mice rejected grafts from H-2b mice which display a normal density of class I major histocompatibility complex (MHC) molecules at the cell surface. Our results indicated that H-2b molecules themselves may be a target in this kind of rejection and that CD4+ T cells play a major role in this autoreactive process. Our data also suggested that TAP1-/- mice, in addition to the well-recognized phenotype of class I and CD8+ T-cell deficiency, present a functional alteration in their autoreactive CD4+ T-cell repertoires. In this model of inflammatory autoreactivity to modified self, we have analysed T-cell receptor (TCR) V-beta-J-beta (BV-BJ) usage by complementarity determining region 3 (CDR3) length spectratyping in splenocytes from naïve TAP1-/- mice and transplanted TAP1-/- mice that rejected B6 heart grafts or responded to synthetic self H-2Kb peptides. Importantly, oligoclonal T-cell expansions shared by different animals were detected in the peripheral T-cell repertoire of transplanted TAP1-/- mice. Such public expansions were also induced in vitro by H-2Kb peptides, suggesting that dominant class I peptides can induce preferential expansions of restricted T-cell populations during rejection. Some of these public T-cell expansions were also detected in transplanted mice even before in vitro stimulation with peptides, indicating that post-transplantation expansion of these populations had occurred in vivo. The functional activity of these T-cell populations awaits elucidation, as do the underlying mechanisms involved in the inflammatory autoreactive process, in TAP1-/- mice.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B, Member 2
- ATP-Binding Cassette Transporters/genetics
- Animals
- Antigen Presentation
- Autoantigens
- Autoimmunity
- CD4-Positive T-Lymphocytes/immunology
- Cell Proliferation
- Complementarity Determining Regions/immunology
- Cytotoxicity, Immunologic
- Graft Rejection/immunology
- H-2 Antigens/immunology
- Heart Transplantation
- In Situ Hybridization, Fluorescence
- Lymphocyte Activation
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Models, Animal
- Receptors, Antigen, T-Cell, alpha-beta/immunology
Collapse
Affiliation(s)
- Idania Marrero
- Immunology Laboratory, Heart Institute (InCor), São Paulo University School of MedicineSão Paulo, Brazil
- Institute for Investigation in Immunology (iii), Millennium InstituteSão Paulo, Brazil
| | - Donald Huffman
- Division of Immune Regulation, La Jolla Institute for Allergy and ImmunologySan Diego, CA, USA
| | - Jorge Kalil
- Immunology Laboratory, Heart Institute (InCor), São Paulo University School of MedicineSão Paulo, Brazil
- Institute for Investigation in Immunology (iii), Millennium InstituteSão Paulo, Brazil
| | - Eli E Sercarz
- Department of Immune Regulation, Torrey Pines Institute for Molecular StudiesSan Diego, CA, USA
| | - Verônica Coelho
- Immunology Laboratory, Heart Institute (InCor), São Paulo University School of MedicineSão Paulo, Brazil
- Institute for Investigation in Immunology (iii), Millennium InstituteSão Paulo, Brazil
| |
Collapse
|
38
|
Brisebois M, Zehntner SP, Estrada J, Owens T, Fournier S. A Pathogenic Role for CD8+ T Cells in a Spontaneous Model of Demyelinating Disease. THE JOURNAL OF IMMUNOLOGY 2006; 177:2403-11. [PMID: 16888002 DOI: 10.4049/jimmunol.177.4.2403] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Transgenic (Tg) mice that overexpress the costimulatory ligand B7.2/CD86 on microglia spontaneously develop a T cell-mediated demyelinating disease. Characterization of the inflammatory infiltrates in the nervous tissue revealed a predominance of CD8+ T cells, suggesting a prominent role of this T cell subset in the pathology. In this study, we show that the same neurological disease occurred in Tg mice deficient in the generation of CD4+ T cells, with an earlier time of onset. Analysis of the CD8+ T cell repertoire at early stage of disease revealed the presence of selected clonal expansions in the CNS but not in peripheral lymphoid organs. We further show that Tg animals deficient in IFN-gamma receptor expression were completely resistant to disease development. Microglia activation that is an early event in disease development is IFN-gamma dependent and thus appears as a key element in disease pathogenesis. Collectively, our data indicate that the spontaneous demyelinating disease in this animal model occurs as a consequence of an inflammatory response initiated through the activation of CNS-specific CD8+ T cells by Tg expression of B7.2 within the target organ. Thus, autoreactive CD8+ T cells can contribute directly to the pathogenesis of neuroinflammatory diseases such as multiple sclerosis.
Collapse
Affiliation(s)
- Marcel Brisebois
- Department of Microbiology and Immunology, McGill University, 3775 University Street, Montreal, Quebec, Canada
| | | | | | | | | |
Collapse
|
39
|
Franco A, Albani S. Translating the concept of suppressor/regulatory T cells to clinical applications. Int Rev Immunol 2006; 25:27-47. [PMID: 16669133 DOI: 10.1080/08830180500544506] [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: 01/12/2023]
Abstract
The in vivo expansion of suppressor/regulatory T cells (Tregs) is a desirable event in autoimmunity and transplantation. Here we summarize the general rules involved in antigen recognition by T cells and describe Tregs and their requirements, discussing different levels of immune intervention.
Collapse
Affiliation(s)
- Alessandra Franco
- Department of Medicine and Pediatrics, University of California, San Diego, La Jolla, California 92093-0731, USA
| | | |
Collapse
|
40
|
Hahn BH, Ebling F, Singh RR, Singh RP, Karpouzas G, La Cava A. Cellular and molecular mechanisms of regulation of autoantibody production in lupus. Ann N Y Acad Sci 2006; 1051:433-41. [PMID: 16126985 PMCID: PMC2291525 DOI: 10.1196/annals.1361.085] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The hyperactive interaction between helper T cells and autoimmune B cells in individuals predisposed to systemic lupus erythematosus (SLE) can be interrupted by induction of regulatory and suppressor T cells. Using two strategies-high dose tolerance to an immunoglobulin-derived peptide, and minigene vaccination with DNA encoding T cell epitopes presented by MHC class I molecules-our group has induced at least three types of regulatory/suppressive T cells. They include CD8+ T cells that suppress helper T cells by cytokine secretion, CD8+ T suppressors that kill B cells making anti-DNA antibodies, and peptide-binding CD4+CD25+ regulatory T cells that suppress B cells by direct cell contact. Each of these lymphocyte subsets suppresses anti-DNA antibody production and delays the onset of nephritis in BWF1 lupus-prone mice. Patients with SLE have amino acid sequences similar to those from murine anti-DNA antibodies used in these studies, and at similar locations in the VH regions of anti-DNA immunoglobulins. Therefore, strategies described here might ultimately be useful in therapy of the human disease.
Collapse
Affiliation(s)
- Bevra H Hahn
- Division of Rheumatology, David Geffen School of Medicine at the University of California-Los Angeles, Los Angeles, CA 91436, USA.
| | | | | | | | | | | |
Collapse
|
41
|
Hasler P. Biological therapies directed against cells in autoimmune disease. ACTA ACUST UNITED AC 2006; 27:443-56. [PMID: 16738955 DOI: 10.1007/s00281-006-0013-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2006] [Accepted: 02/02/2006] [Indexed: 10/24/2022]
Abstract
Among the cells of the immune system involved in the pathogenesis of autoimmune disease, T cells have received the most attention. The central role of these cells in several animal models of autoimmune diseases and in human disease counterparts has provided the rationale for specific therapeutic targeting of T cell subsets, especially CD4 T cells. So far, the applicability of this approach has not been clearly evident in clinical trials, which was also the case when nondepleting "coating" anti-CD4 monoclonal antibodies was used. In the past several years, experimental evidence supporting a major role of B cells in systemic autoimmune disease has grown. This includes the pathogenicity of certain autoantibodies, the potential of B cells to present antigen in the context of MHC Class II and to signal via costimulatory molecules, and to secrete proinflammatory cytokines. In some instances, engagement of the B cell receptor and other surface receptors is sufficient to stimulate B cells to produce antibodies. The depletion of B cells by targeting the surface marker CD20 has been shown to be effective in treating rheumatoid arthritis with a good side effect profile. Series of cases with other systemic autoimmune diseases indicate that this strategy may be effective in these conditions too. The clinical data add weight to the importance of B cells in the pathogenesis of autoimmune diseases.
Collapse
Affiliation(s)
- Paul Hasler
- Rheumaklinik, Kantonsspital Aarau, Tellstrasse, 5001 Aarau, Switzerland.
| |
Collapse
|
42
|
Affiliation(s)
- Hong Jiang
- Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA.
| | | |
Collapse
|
43
|
Kwon SS, Kim N, Yoo TJ. The effects of intradermal vaccination with DNA encoding for the T-cell receptor on the induction of experimental autoimmune encephalomyelitis in B10.PL mice. J Korean Med Sci 2005; 20:1039-45. [PMID: 16361819 PMCID: PMC2779306 DOI: 10.3346/jkms.2005.20.6.1039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Intradermal gene administration was found to induce a more profound immune response than direct intramuscular gene injection. We performed intradermal vaccination of B10.PL mice with DNA encoding for the V 8.2 region of the T-cell receptors (TCR). Three weeks later, these mice were immunized with rat myelin basic protein (MBP). Daily mean clinical scores and mortality rate were lower in this group compared with controls. The proliferative responses of lymph node cells to rat MBP were slightly less in the vaccination groups than in the control groups (p < 0.05). However, we detected no differences between the two groups with regard to the production of MBP-specific IgG, IgG1, & IgG2a antibodies. The levels of cytokine mRNA expression in the vaccination groups were observed higher than in the control groups without antigen-specific stimulation, but all of cytokine expressions between the vaccination and control groups after antigen-specific stimulation were identical. These results demonstrate that intradermal DNA vaccines encoding for TCR might prove to be useful in the control of autoimmune disease.
Collapse
MESH Headings
- Animals
- Autoantibodies/blood
- Base Sequence
- Cytokines/genetics
- DNA, Complementary/genetics
- Encephalomyelitis, Autoimmune, Experimental/etiology
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/prevention & control
- Female
- Gene Expression
- Genes, T-Cell Receptor beta
- In Vitro Techniques
- Injections, Intradermal
- Lymphocyte Activation
- Mice
- Myelin Basic Protein/immunology
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Rats
- Vaccines, DNA/administration & dosage
- Vaccines, DNA/genetics
Collapse
Affiliation(s)
- Soon Seog Kwon
- Department of Internal Medicine, The Catholic University of Korea, College of Medicine, Seoul, Korea.
| | | | | |
Collapse
|
44
|
Walter U, Santamaria P. CD8+ T cells in autoimmunity. Curr Opin Immunol 2005; 17:624-31. [PMID: 16226438 DOI: 10.1016/j.coi.2005.09.014] [Citation(s) in RCA: 102] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2005] [Accepted: 09/20/2005] [Indexed: 10/25/2022]
Abstract
Mounting evidence shows that CD8(+) T cells contribute to the initiation, progression and regulation of several pathogenic autoimmune responses in which these cells were not previously thought to play a major role. CD8(+) T cells can kill target cells directly, by recognizing peptide-MHC complexes on target cells, or indirectly, by secreting cytokines capable of signaling through death receptors expressed on the target cell surface. Autoreactive CD8(+) T cells can also contribute to autoimmunity by releasing cytokines capable of increasing the susceptibility of target cells to cytotoxicity, or by secreting chemokines that attract other immune cells to the site of autoimmunity. Autoreactive CD8(+) T cells can also downregulate autoimmune responses. Recent important advances include a mechanistic understanding of events leading to the activation and recruitment of autoreactive CD8(+) T cells in certain autoimmune responses and a greater appreciation of the diverse roles that these T cells play in autoimmunity.
Collapse
Affiliation(s)
- Ulrich Walter
- Julia McFarlane Diabetes Research Centre and Department of Microbiology and Infectious Diseases, Faculty of Medicine, The University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta T2N 4N1, Canada
| | | |
Collapse
|
45
|
Marrero I, Benvenutti LA, Kalil J, Coelho V. Autoreactivity to self H-2Kb peptides in TAP1 mice. Intravenous administration of H-2Kb class I-derived peptides induces long-term survival of grafts from C57BL/6 donors. Immunology 2005; 115:484-94. [PMID: 16011517 PMCID: PMC1782177 DOI: 10.1111/j.1365-2567.2005.02182.x] [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: 01/22/2023] Open
Abstract
We and others have previously shown that TAP1-/- mice (H-2b) reject grafts from donors without major histocompatibility complex (MHC) disparity that express wild-type levels of H-2b class I molecules (C57BL/6, TAP1+/+ mice). In this same model, we also showed that subcutaneous priming of TAP1-/- mice with synthetic peptides derived from the H-2Kb molecule accelerated graft rejection and that in vivo depletion of CD4+ T cells induced a significant prolongation of graft survival, suggesting an important role for CD4 T cells. We hypothesize that, in this model, rejection is triggered by the recognition of class I molecules or derived peptides, in an inflammatory microenvironment, by a functionally altered autoreactive T-cell repertoire that escapes the control of peripheral regulatory mechanisms. In the present study, we analysed the cellular autoreactivity induced by synthetic peptides derived from the H-2Kb sequence in naive and TAP1-/- mice transplanted with C57BL/6 grafts, and investigated whether intravenous modulation of autoreactivity to these peptides induced transplantation tolerance. We showed that TAP1-/- mice have peripheral autoreactive T cells that recognize H-2Kb peptides. A significant amplification of proliferation against these peptides was detected in TAP1-/- mice that rejected grafts, indicating that the inflammatory context of transplantation induced peripheral expansion of these autoreactive T cells. Furthermore, intravenous injection of H-2Kb-derived peptides significantly prolonged graft survival in some animals. In these mice (> 100 days graft survival), we observed intragraft inhibition of interferon-gamma and interleukin-10 expression, suggesting that these cytokines have an active role during the rejection. In conclusion, our present data indicate that inflammatory autoreactive T cells directed against H-2Kb peptides can be inhibited in the periphery to prolong graft survival in TAP1-/- mice.
Collapse
Affiliation(s)
- Idania Marrero
- Immunology Laboratory, Heart Institute (InCor), São Paulo University School of MedicineSão Paulo, Brazil
- Institute for Investigation in Immunology (iii), Millennium InstituteSão Paulo, Brazil
| | - Luiz Alberto Benvenutti
- Department of Pathology, Heart Institute (InCor), São Paulo University School of MedicineSão Paulo, Brazil
| | - Jorge Kalil
- Immunology Laboratory, Heart Institute (InCor), São Paulo University School of MedicineSão Paulo, Brazil
- Institute for Investigation in Immunology (iii), Millennium InstituteSão Paulo, Brazil
| | - Verônica Coelho
- Immunology Laboratory, Heart Institute (InCor), São Paulo University School of MedicineSão Paulo, Brazil
- Institute for Investigation in Immunology (iii), Millennium InstituteSão Paulo, Brazil
| |
Collapse
|
46
|
Friese MA, Fugger L. Autoreactive CD8+ T cells in multiple sclerosis: a new target for therapy? Brain 2005; 128:1747-63. [PMID: 15975943 DOI: 10.1093/brain/awh578] [Citation(s) in RCA: 176] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Multiple sclerosis afflicts more than 1 million individuals worldwide and is widely considered to be an autoimmune disease. Traditionally, CD4(+) T helper cells have almost exclusively been held responsible for its immunopathogenesis, partly because certain MHC class II alleles clearly predispose for developing multiple sclerosis and also, because of their importance in inducing experimental autoimmune encephalomyelitis (EAE), the animal model for multiple sclerosis. However, several strategies that target CD4(+) T cells beneficially in EAE have failed to ameliorate disease activity in multiple sclerosis, and some have even triggered exacerbations. Recently, the potential importance of CD8(+) T cells has begun to emerge. Physiologically, CD8(+) T cells are essential for detecting and eliminating abnormal cells, whether infected or neoplastic. In multiple sclerosis, genetic associations with MHC class I alleles have now been established, and CD8(+) as well as CD4(+) T cells have been found to invade and clonally expand in inflammatory central nervous system plaques. Recent animal models induced by CD8(+) T cells show interesting similarities to multiple sclerosis, in particular, in lesion distribution (more inflammation in the brain relative to the spinal cord), although not all of the features of the human disease are recapitulated. Here we outline the arguments for a possible role for CD8(+) T cells, a lymphocyte subset that has long been underrated in multiple sclerosis and should now be considered in new therapeutic approaches.
Collapse
Affiliation(s)
- Manuel A Friese
- MRC Human Immunology Unit and Department of Clinical Neurology, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK
| | | |
Collapse
|
47
|
Bradl M, Bauer J, Flügel A, Wekerle H, Lassmann H. Complementary contribution of CD4 and CD8 T lymphocytes to T-cell infiltration of the intact and the degenerative spinal cord. THE AMERICAN JOURNAL OF PATHOLOGY 2005; 166:1441-50. [PMID: 15855644 PMCID: PMC1606398 DOI: 10.1016/s0002-9440(10)62361-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The central role of T cells in inflammatory reactions of the central nervous system (CNS) is well documented. However, there is little information about the few T cells found within the noninflamed CNS. In particular, the contribution of CD4+ and CD8+ T cells to the lymphocyte pool infiltrating the intact CNS, the location of these cells in CNS white and gray matter, and changes in the cellular composition of T-cell infiltrates coinciding with degeneration are primarily undefined. To address these points, we studied T cells in the intact and degenerative rat spinal cord. In the intact spinal cord, T cells were preferentially located within the gray matter. CD8+ T cells were more numerous than CD4+ lymphocytes. In cases of neuroaxonal degeneration or myelin degeneration/oligodendrocyte death, T cells were predominantly seen in areas of degeneration and were present in increased numbers. These effects were more pronounced for the CD4+ than for the CD8+ T-cell subset. Collectively, these data provide evidence for a clear cellular and compartmental bias in T-cell infiltration of the intact and degenerative spinal cord. This could indicate that CD4+ and CD8+ T cells might fulfill complementary roles in the intact and the diseased organ.
Collapse
Affiliation(s)
- Monika Bradl
- Department of Neuroimmunology, Brain Research Institute, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, A-1090 Vienna, Austria.
| | | | | | | | | |
Collapse
|
48
|
Reed-Loisel LM, Sullivan BA, Laur O, Jensen PE. An MHC Class Ib-Restricted TCR That Cross-Reacts with an MHC Class Ia Molecule. THE JOURNAL OF IMMUNOLOGY 2005; 174:7746-52. [PMID: 15944277 DOI: 10.4049/jimmunol.174.12.7746] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
TCR transgenic 6C5 T cells recognize an insulin B chain epitope presented by the nonclassical class I MHC molecule, Qa-1(b). Positive selection of these T cells was shown previously to require Qa-1(b). Despite dedicated specificity for Qa-1(b), evidence presented in the current study indicates that 6C5 T cells can cross-recognize a classical class I molecule. Clonal deletion was observed unexpectedly in 6C5.H-2(bxq) mice, which do not express I-E MHC class II molecules and thus should not be subject to superantigen-mediated negative selection. 6C5 T cells were observed to respond in vivo and in vitro to spleen cells from allogeneic H-2(q) mice, and specificity was mapped to D(q). Evidence was obtained for direct recognition of D(q), rather than indirect presentation of a D(q)-derived peptide presented by Qa-1(b). Polyclonal CD8(+) T cells from class Ia-deficient K(b)D(b-/-) mice reacted in vitro to allogeneic spleen cells with an apparent frequency comparable to conventional class Ia-restricted T cells. Our results provide a clear example of a Qa-1-specific TCR that can cross-react with a class Ia molecule and evidence supporting the idea that this may be a common property of T cells selected by class Ib molecules.
Collapse
MESH Headings
- Animals
- Cell Differentiation/genetics
- Cell Differentiation/immunology
- Cell Line, Transformed
- Cell Line, Tumor
- Cell Lineage/genetics
- Cell Lineage/immunology
- Clonal Deletion
- Crosses, Genetic
- H-2 Antigens/genetics
- H-2 Antigens/immunology
- H-2 Antigens/metabolism
- Histocompatibility Antigens Class I/genetics
- Histocompatibility Antigens Class I/immunology
- Histocompatibility Antigens Class I/metabolism
- Mammary Tumor Virus, Mouse/immunology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C3H
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/metabolism
- Superantigens/genetics
- Superantigens/immunology
- Superantigens/metabolism
- T-Lymphocyte Subsets/cytology
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
Collapse
Affiliation(s)
- Lisa M Reed-Loisel
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | | | | |
Collapse
|
49
|
Jiang H, Wu Y, Liang B, Zheng Z, Tang G, Kanellopoulos J, Soloski M, Winchester R, Goldstein I, Chess L. An affinity/avidity model of peripheral T cell regulation. J Clin Invest 2005; 115:302-12. [PMID: 15668735 PMCID: PMC544609 DOI: 10.1172/jci23879] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2004] [Accepted: 12/01/2004] [Indexed: 01/20/2023] Open
Abstract
We show in these studies that Qa-1-dependent CD8+ T cells are involved in the establishment and maintenance of peripheral self tolerance as well as facilitating affinity maturation of CD4+ T cells responding to foreign antigen. We provide experimental evidence that the strategy used by the Qa-1-dependent CD8+ T cells to accomplish both these tasks in vivo is to selectively downregulate T cell clones that respond to both self and foreign antigens with intermediate, not high or low, affinity/avidity. Thus, the immune system evolved to regulate peripheral immunity using a unified mechanism that efficiently and effectively permits the system to safeguard peripheral self tolerance yet promote the capacity to deal with foreign invaders.
Collapse
Affiliation(s)
- Hong Jiang
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, New York 10032, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Abstract
Regulation of the immune response is a multifaceted process involving lymphocytes that function to maintain both self tolerance as well as homeostasis following productive immunity against microbes. There are 2 broad categories of Tregs that function in different immunological settings depending upon the context of antigen exposure and the nature of the inflammatory response. During massive inflammatory conditions such as microbial exposure in the gut or tissue transplantation, regulatory CD4+CD25+ Tregs broadly suppress priming and/or expansion of polyclonal autoreactive responses nonspecifically. In other immune settings where initially a limited repertoire of antigen-reactive T cells is activated and expanded, TCR-specific negative feedback mechanisms are able to achieve a fine homeostatic balance. Here I will describe experimental evidence for the existence of a Treg population specific for determinants that are derived from the TCR and are expressed by expanding myelin basic protein-reactive T cells mediating experimental autoimmune encephalomyelitis, an animal prototype for multiple sclerosis. These mechanisms ensure induction of effective but appropriately limited responses against foreign antigens while preventing autoreactivity from inflicting escalating damage. In contrast to CD25+ Tregs, which are most efficient at suppressing priming or activation, these specific Tregs are most efficient in controlling T cells following their activation.
Collapse
Affiliation(s)
- Vipin Kumar
- Laboratory of Autoimmunity, Torrey Pines Institute for Molecular Studies, San Diego, California 92121, USA.
| |
Collapse
|