1
|
Li J, Yang Z, Kawajiri A, Sato K, Tayama S, Ishii N, Zhu J, Kawabe T. Excess generation and activation of naturally arising memory-phenotype CD4 + T lymphocytes are inhibited by regulatory T cells in steady state. Front Immunol 2024; 15:1429954. [PMID: 39221254 PMCID: PMC11361994 DOI: 10.3389/fimmu.2024.1429954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 07/31/2024] [Indexed: 09/04/2024] Open
Abstract
Conventional CD4+ T lymphocytes consist of naïve, foreign antigen-specific memory, and self-antigen-driven memory-phenotype (MP) cell compartments at homeostasis. We recently showed that MP cells tonically proliferate in response to self-antigens and differentiate into the T-bet+ subset in steady state. How excess proliferation and differentiation of MP cells are inhibited remains unclear. Given immunosuppressive function of regulatory T cells (Tregs), it is possible that they are also involved in inhibition of spontaneous MP cell activation. Here we show using Foxp3-diphtheria toxin receptor-transgenic mice that both MP and naïve CD4+ T cells spontaneously proliferate and differentiate into Th1 cells upon acute Treg depletion. At an early time point post Treg depletion, MP as compared to naïve CD4+ T cells are preferentially activated while at a later stage, the response is dominated by activated cells originated from the naïve pool. Moreover, we argue that MP cell proliferation is driven by TCR and CD28 signaling whereas Th1 differentiation mediated by IL-2. Furthermore, our data indicate that such activation of MP and naïve CD4+ T lymphocytes contribute to development of multi-organ inflammation at early and later time points, respectively, after Treg ablation. Together our findings reveal that Tregs tonically inhibit early, spontaneous proliferation and Th1 differentiation of MP CD4+ T lymphocytes as well as late activation of naïve cells, thereby contributing to maintenance of T cell homeostasis.
Collapse
Affiliation(s)
- Jing Li
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Ziying Yang
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Akihisa Kawajiri
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Kosuke Sato
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Shunichi Tayama
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Naoto Ishii
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Jinfang Zhu
- Molecular and Cellular Immunoregulation Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Takeshi Kawabe
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
- Molecular and Cellular Immunoregulation Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
2
|
Kawabe T. Homeostasis and immunological function of self-driven memory-phenotype CD4 + T lymphocytes. Immunol Med 2023; 46:1-8. [PMID: 36218322 DOI: 10.1080/25785826.2022.2129370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023] Open
Abstract
CD4+ T lymphocytes play an essential role in adaptive immune responses. In pathogen infection, naïve CD4+ T cells that strongly respond to foreign antigens robustly proliferate to differentiate into effector/memory cells, contributing to elimination of the pathogen concerned. In addition to this conventional T cell activation pathway, naïve T cells can also weakly respond to self antigens in the periphery to spontaneously acquire a memory phenotype through homeostatic proliferation in steady state. Such 'memory-phenotype' (MP) CD4+ T lymphocytes are distinguishable from foreign antigen-specific memory cells in terms of marker expression. Once generated, MP cells are maintained by rapid proliferation while differentiating into the T-bet+ 'MP1' subset, with the latter response promoted by IL-12 homeostatically produced by type 1 dendritic cells. Importantly, MP1 cells possess innate immune function; they can produce IFN-γ in response to IL-12 and IL-18 to contribute to host defense against pathogens. Similarly, the presence of RORγt+ 'MP17' and Gata3hi 'MP2' cells as well as their potential immune functions have been proposed. In this review, I will discuss our current understanding on the unique mechanisms of generation, maintenance, and differentiation of MP CD4+ T lymphocytes as well as their functional significance in various disease conditions.
Collapse
Affiliation(s)
- Takeshi Kawabe
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| |
Collapse
|
3
|
Kawabe T, Sher A. Memory-phenotype CD4+ T cells: a naturally arising T lymphocyte population possessing innate immune function. Int Immunol 2021; 34:189-196. [PMID: 34897483 PMCID: PMC8962445 DOI: 10.1093/intimm/dxab108] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 12/06/2021] [Indexed: 12/15/2022] Open
Abstract
In conventional adaptive immune responses, upon recognition of foreign antigens, naive CD4+ T lymphocytes are activated to differentiate into effector/memory cells. In addition, emerging evidence suggests that in the steady state, naive CD4+ T cells spontaneously proliferate in response to self-antigens to acquire a memory phenotype (MP) through homeostatic proliferation. This expansion is particularly profound in lymphopenic environments but also occurs in lymphoreplete, normal conditions. The 'MP T lymphocytes' generated in this manner are maintained by rapid proliferation in the periphery and they tonically differentiate into T-bet-expressing 'MP1' cells. Such MP1 CD4+ T lymphocytes can exert innate effector function, producing IFN-γ in response to IL-12 in the absence of antigen recognition, thereby contributing to host defense. In this review, we will discuss our current understanding of how MP T lymphocytes are generated and persist in steady-state conditions, their populational heterogeneity as well as the evidence for their effector function. We will also compare these properties with those of a similar population of innate memory cells previously identified in the CD8+ T lymphocyte lineage.
Collapse
Affiliation(s)
- Takeshi Kawabe
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan,Correspondence to: T. Kawabe; E-mail: or A. Sher; E-mail:
| | - Alan Sher
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA,Correspondence to: T. Kawabe; E-mail: or A. Sher; E-mail:
| |
Collapse
|
4
|
Abstract
Conventional CD4+ and CD8+ T lymphocytes comprise a mixture of naive and memory cells. Generation and survival of these T-cell subsets is under strict homeostatic control and reflects contact with self-major histocompatibility complex (MHC) and certain cytokines. Naive T cells arise in the thymus via T-cell receptor (TCR)-dependent positive selection to self-peptide/MHC complexes and are then maintained in the periphery through self-MHC interaction plus stimulation via interleukin-7 (IL-7). By contrast, memory T cells are largely MHC-independent for their survival but depend strongly on stimulation via cytokines. Whereas typical memory T cells are generated in response to foreign antigens, some arise spontaneously through contact of naive precursors with self-MHC ligands; we refer to these cells as memory-phenotype (MP) T cells. In this review, we discuss the generation and homeostasis of naive T cells and these two types of memory T cells, focusing on their relative interaction with MHC ligands and cytokines.
Collapse
Affiliation(s)
- Takeshi Kawabe
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan
| | - Jaeu Yi
- Division of Rheumatology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Jonathan Sprent
- Immunology Division, Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia
- St. Vincent's Clinical School, University of New South Wales, Sydney, New South Wales 2010, Australia
| |
Collapse
|
5
|
Eldershaw S, Verma K, Croft W, Rai T, Kinsella F, Stephens C, Chen H, Nunnick J, Zuo J, Malladi R, Moss P. Lymphopenia-induced lymphoproliferation drives activation of naive T cells and expansion of regulatory populations. iScience 2021; 24:102164. [PMID: 33665580 PMCID: PMC7907823 DOI: 10.1016/j.isci.2021.102164] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 10/15/2020] [Accepted: 02/04/2021] [Indexed: 11/23/2022] Open
Abstract
Chemotherapy pre-conditioning is an essential component of chimeric antigen receptor transduced cell therapy. Acute lymphopenia-induced proliferation (LIP) is known to be driven primarily by homeostatic cytokines, but little is known on the underlying mechanisms in humans. We undertook phenotypic and transcriptional analysis of T cells undergoing LIP two weeks post-myeloablative autograft stem cell transplantation. Strong IL-7 signaling was reflected in downregulated IL-7R expression on all T cells, including naive cells, along with parallel increased IL-2Rα expression. Notably, activated residual naive cells expressed Fas indicating recent TCR engagement. Moreover, proportion of Ki67 + FoxP3+ Tregs was almost doubled. Transcriptional analysis revealed increased fatty acid metabolism and interferon signaling responses. In contrast, TGF-β signaling was strongly suppressed. Thus, human LIP response is characterized by cytokine and TCR-driven proliferation which drives global T cell activation but also preferentially triggers regulatory cell expansion which may limit tumor-specific immunity. These features indicate potential therapeutic opportunities to manipulate immunotherapy regimens incorporating LIP conditioning protocols.
Collapse
Affiliation(s)
- S. Eldershaw
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - K. Verma
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - W. Croft
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
- Centre for Computational Biology, University of Birmingham, Birmingham, UK
| | - T. Rai
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - F.A.M. Kinsella
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
- Center for clinical Haematology, Queen Elizabeth Hospital, Birmingham, UK
| | - C. Stephens
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - H. Chen
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - J. Nunnick
- Center for clinical Haematology, Queen Elizabeth Hospital, Birmingham, UK
| | - J. Zuo
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - R. Malladi
- Center for clinical Haematology, Queen Elizabeth Hospital, Birmingham, UK
| | - P. Moss
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
- Centre for Computational Biology, University of Birmingham, Birmingham, UK
| |
Collapse
|
6
|
Santos-Zas I, Lemarié J, Zlatanova I, Cachanado M, Seghezzi JC, Benamer H, Goube P, Vandestienne M, Cohen R, Ezzo M, Duval V, Zhang Y, Su JB, Bizé A, Sambin L, Bonnin P, Branchereau M, Heymes C, Tanchot C, Vilar J, Delacroix C, Hulot JS, Cochain C, Bruneval P, Danchin N, Tedgui A, Mallat Z, Simon T, Ghaleh B, Silvestre JS, Ait-Oufella H. Cytotoxic CD8 + T cells promote granzyme B-dependent adverse post-ischemic cardiac remodeling. Nat Commun 2021; 12:1483. [PMID: 33674611 PMCID: PMC7935973 DOI: 10.1038/s41467-021-21737-9] [Citation(s) in RCA: 103] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 02/09/2021] [Indexed: 12/21/2022] Open
Abstract
Acute myocardial infarction is a common condition responsible for heart failure and sudden death. Here, we show that following acute myocardial infarction in mice, CD8+ T lymphocytes are recruited and activated in the ischemic heart tissue and release Granzyme B, leading to cardiomyocyte apoptosis, adverse ventricular remodeling and deterioration of myocardial function. Depletion of CD8+ T lymphocytes decreases apoptosis within the ischemic myocardium, hampers inflammatory response, limits myocardial injury and improves heart function. These effects are recapitulated in mice with Granzyme B-deficient CD8+ T cells. The protective effect of CD8 depletion on heart function is confirmed by using a model of ischemia/reperfusion in pigs. Finally, we reveal that elevated circulating levels of GRANZYME B in patients with acute myocardial infarction predict increased risk of death at 1-year follow-up. Our work unravels a deleterious role of CD8+ T lymphocytes following acute ischemia, and suggests potential therapeutic strategies targeting pathogenic CD8+ T lymphocytes in the setting of acute myocardial infarction. Immune cells contribute to adverse remodeling following myocardial infarction. Here the authors show in mice and pigs that CD8+ lymphocytes release Granzyme B in the infarcted heart leading to cardiomyocyte death, enhanced inflammation and deterioration of cardiac function.
Collapse
Affiliation(s)
| | | | | | - Marine Cachanado
- Assistance Publique-Hôpitaux de Paris, APHP.SU; Department of Clinical Pharmacology and Clinical Research Platform (URCEST-CRB-CRC-EST), Hôpital Saint Antoine, Paris, France
| | | | - Hakim Benamer
- Service de cardiologie, Institut Cardiovasculaire Paris Sud, Paris, France
| | - Pascal Goube
- Service de cardiologie, Centre Hospitalier de Corbeil, Corbeil, France
| | | | - Raphael Cohen
- Université de Paris, PARCC, INSERM, F-75015, Paris, France
| | - Maya Ezzo
- Université de Paris, PARCC, INSERM, F-75015, Paris, France
| | - Vincent Duval
- Université de Paris, PARCC, INSERM, F-75015, Paris, France
| | - Yujiao Zhang
- Université de Paris, PARCC, INSERM, F-75015, Paris, France
| | - Jin-Bo Su
- Inserm U955-IMRB, Equipe 03, UPEC, Ecole Nationale Vétérinaire d'Alfort, Maisons-Alfort, France
| | - Alain Bizé
- Inserm U955-IMRB, Equipe 03, UPEC, Ecole Nationale Vétérinaire d'Alfort, Maisons-Alfort, France
| | - Lucien Sambin
- Inserm U955-IMRB, Equipe 03, UPEC, Ecole Nationale Vétérinaire d'Alfort, Maisons-Alfort, France
| | - Philippe Bonnin
- Inserm U965, Department of Physiology, Assistance Publique Hôpitaux de Paris, Hôpital Lariboisière, France
| | - Maxime Branchereau
- Inserm U1048-Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), université Paul Sabatier, Toulouse, France
| | - Christophe Heymes
- Inserm U1048-Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), université Paul Sabatier, Toulouse, France
| | | | - José Vilar
- Université de Paris, PARCC, INSERM, F-75015, Paris, France
| | | | | | - Clement Cochain
- Institute of Experimental Biomedicine, University Hospital Würzburg, Würzburg, Germany
| | - Patrick Bruneval
- Université de Paris, PARCC, INSERM, F-75015, Paris, France.,Service d'anatomopathologie, Hôpital Europeen G. Pompidou, Assistance Publique, Hôpitaux de Paris, Paris, France
| | - Nicolas Danchin
- Service de cardiologie, Hôpital Europeen G. Pompidou, Assistance Publique, Hôpitaux de Paris, Paris, France
| | - Alain Tedgui
- Université de Paris, PARCC, INSERM, F-75015, Paris, France
| | - Ziad Mallat
- Université de Paris, PARCC, INSERM, F-75015, Paris, France.,Division of Cardiovascular Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, CB2 2QQ, UK
| | - Tabassome Simon
- Assistance Publique-Hôpitaux de Paris, APHP.SU; Department of Clinical Pharmacology and Clinical Research Platform (URCEST-CRB-CRC-EST), Hôpital Saint Antoine, Paris, France.,Sorbonne Université, UPMC-site St Antoine, Service de Pharmacologie, Assistance Publique-Hôpitaux de Paris, APHP.SU; Department of Clinical Pharmacology and Clinical Research Platform (URCEST-CRB-CRC-EST), Hôpital Saint Antoine, Paris, France
| | - Bijan Ghaleh
- Inserm U955-IMRB, Equipe 03, UPEC, Ecole Nationale Vétérinaire d'Alfort, Maisons-Alfort, France
| | | | - Hafid Ait-Oufella
- Université de Paris, PARCC, INSERM, F-75015, Paris, France. .,Sorbonne Université, Service de médecine intensive-Réanimation, Assistance Publique, Hôpitaux de Paris, Paris, France.
| |
Collapse
|
7
|
Lupieri A, Smirnova NF, Solinhac R, Malet N, Benamar M, Saoudi A, Santos-Zas I, Zeboudj L, Ait-Oufella H, Hirsch E, Ohayon P, Lhermusier T, Carrié D, Arnal JF, Ramel D, Gayral S, Laffargue M. Smooth muscle cells-derived CXCL10 prevents endothelial healing through PI3Kγ-dependent T cells response. Cardiovasc Res 2020; 116:438-449. [PMID: 31106375 DOI: 10.1093/cvr/cvz122] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 02/25/2019] [Accepted: 05/15/2019] [Indexed: 12/14/2022] Open
Abstract
AIMS Defects in efficient endothelial healing have been associated with complication of atherosclerosis such as post-angioplasty neoatherosclerosis and plaque erosion leading to thrombus formation. However, current preventive strategies do not consider re-endothelialization in their design. Here, we investigate mechanisms linking immune processes and defect in re-endothelialization. We especially evaluate if targeting phosphoinositide 3-kinase γ immune processes could restore endothelial healing and identify immune mediators responsible for these defects. METHODS AND RESULTS Using in vivo model of endovascular injury, we showed that both ubiquitous genetic inactivation of PI3Kγ and hematopoietic cell-specific PI3Kγ deletion improved re-endothelialization and that CD4+ T-cell population drives this effect. Accordingly, absence of PI3Kγ activity correlates with a decrease in local IFNγ secretion and its downstream interferon-inducible chemokine CXCL10. CXCL10 neutralization promoted re-endothelialization in vivo as the same level than those observed in absence of PI3Kγ suggesting a role of CXCL10 in re-endothelialization defect. Using a new established ex vivo model of carotid re-endothelialization, we showed that blocking CXCL10 restore the IFNγ-induced inhibition of endothelial healing and identify smooth muscle cells as the source of CXCL10 secretion in response to Th1 cytokine. CONCLUSION Altogether, these findings expose an unforeseen cellular cross-talk within the arterial wall whereby a PI3Kγ-dependent T-cell response leads to CXCL10 production by smooth muscle cells which in turn inhibits endothelial healing. Therefore, both PI3Kγ and the IFNγ/CXCL10 axis provide novel strategies to promote endothelial healing.
Collapse
Affiliation(s)
- Adrien Lupieri
- Institute of Metabolic and Cardiovascular Diseases (I2MC), Université de Toulouse, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1048, Toulouse F-31432, France
| | - Natalia F Smirnova
- Institute of Metabolic and Cardiovascular Diseases (I2MC), Université de Toulouse, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1048, Toulouse F-31432, France
| | - Romain Solinhac
- Institute of Metabolic and Cardiovascular Diseases (I2MC), Université de Toulouse, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1048, Toulouse F-31432, France
| | - Nicole Malet
- Institute of Metabolic and Cardiovascular Diseases (I2MC), Université de Toulouse, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1048, Toulouse F-31432, France
| | - Mehdi Benamar
- Centre de Physiopathologie de Toulouse Purpan, Université de Toulouse, INSERM, Centre National de la Recherche Scientifique (CNRS), Toulouse, F 31300, France
| | - Abdel Saoudi
- Centre de Physiopathologie de Toulouse Purpan, Université de Toulouse, INSERM, Centre National de la Recherche Scientifique (CNRS), Toulouse, F 31300, France
| | - Icia Santos-Zas
- Paris-Cardiovascular Research Center (PARCC), Université Paris Descartes, Sorbonne Paris Cité, Institut National de la Santé et de la Recherche Médicale (INSERM), UMR970, Paris, France
| | - Lynda Zeboudj
- Paris-Cardiovascular Research Center (PARCC), Université Paris Descartes, Sorbonne Paris Cité, Institut National de la Santé et de la Recherche Médicale (INSERM), UMR970, Paris, France
| | - Hafid Ait-Oufella
- Paris-Cardiovascular Research Center (PARCC), Université Paris Descartes, Sorbonne Paris Cité, Institut National de la Santé et de la Recherche Médicale (INSERM), UMR970, Paris, France
| | - Emilio Hirsch
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Via Nizza 52, 10126 Torino, Italy
| | - Paul Ohayon
- Institute of Metabolic and Cardiovascular Diseases (I2MC), Université de Toulouse, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1048, Toulouse F-31432, France.,Department of Cardiology, University Hospital Rangueil, Toulouse, France
| | - Thibault Lhermusier
- Institute of Metabolic and Cardiovascular Diseases (I2MC), Université de Toulouse, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1048, Toulouse F-31432, France.,Department of Cardiology, University Hospital Rangueil, Toulouse, France
| | - Didier Carrié
- Institute of Metabolic and Cardiovascular Diseases (I2MC), Université de Toulouse, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1048, Toulouse F-31432, France.,Department of Cardiology, University Hospital Rangueil, Toulouse, France
| | - Jean-François Arnal
- Institute of Metabolic and Cardiovascular Diseases (I2MC), Université de Toulouse, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1048, Toulouse F-31432, France
| | - Damien Ramel
- Institute of Metabolic and Cardiovascular Diseases (I2MC), Université de Toulouse, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1048, Toulouse F-31432, France
| | - Stephanie Gayral
- Institute of Metabolic and Cardiovascular Diseases (I2MC), Université de Toulouse, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1048, Toulouse F-31432, France
| | - Muriel Laffargue
- Institute of Metabolic and Cardiovascular Diseases (I2MC), Université de Toulouse, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1048, Toulouse F-31432, France
| |
Collapse
|
8
|
Knipper JA, Wright D, Cope AP, Malissen B, Zamoyska R. PTPN22 Acts in a Cell Intrinsic Manner to Restrict the Proliferation and Differentiation of T Cells Following Antibody Lymphodepletion. Front Immunol 2020; 11:52. [PMID: 32047502 PMCID: PMC6997546 DOI: 10.3389/fimmu.2020.00052] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 01/09/2020] [Indexed: 11/25/2022] Open
Abstract
Lymphopenic insult has been shown to precipitate the initiation of autoimmune disease in murine models such as the Non-obese diabetic mouse. Similarly, in man lymphopenia induced by mAb therapy, for instance Alemtuzumab as treatment for Multiple Sclerosis, can precipitate development of secondary autoimmune disease in up to 30 % of patients. We asked whether an identified autoimmune susceptibility locus might increase the risk of developing autoimmunity in the context of mAb-induced lymphopenia in a mouse model. A single nucleotide polymorphism (SNP) in the gene encoding the tyrosine phosphatase PTPN22 (R620W) is associated with multiple human autoimmune diseases, and PTPN22 has been shown to modulate T cell responses, particularly to weak antigens. In keeping with this, PTPN22-deficient or PTPN22 R619W mutant murine T cells adoptively transferred into immunodeficient lymphopenic hosts showed a higher lymphopenia-induced proliferation rate than WT cells. We induced lymphopenia by treating wild-type or PTPN22 knock-out mice with T cell depleting antibodies and monitored reconstitution of the T cell pool. We found that PTPN22 deficient T cells acquired a more activated effector phenotype, with significantly more IFNγ producing cells. This resulted from expansion driven by self-peptide MHC, as it was evident when the contribution of IL-7 to lymphopenic expansion was blocked with IL-7R Ab. Interestingly, Foxp3+ Tregs were also considerably expanded in PTPN22-deficient and PTPN22 R619W mice, as was the frequency of both CD25+ and CD25- CD4 T cells that produce IL-10. Using bone marrow chimeric mice, we showed that PTPN22 influenced development of both regulatory and effector T cell functions in a cell-intrinsic manner. Overall the expansion of Tregs is likely to keep the expanded T effector populations in check and sparing Treg during therapeutic mAb depletion may be a useful strategy to prevent occurrence of secondary autoimmunity.
Collapse
Affiliation(s)
- Johanna A Knipper
- Ashworth Laboratories, Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, United Kingdom
| | - David Wright
- Ashworth Laboratories, Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Andrew P Cope
- Faculty of Life Sciences and Medicine, Centre for Inflammation Biology and Cancer Immunology, School of Immunology and Microbial Sciences, King's College London, London, United Kingdom
| | - Bernard Malissen
- Centre d'Immunologie de Marseille-Luminy, INSERM, CNRS, Aix Marseille Université, Marseille, France.,Centre d'Immunophénomique, Aix Marseille Université, INSERM, CNRS UMR, Marseille, France
| | - Rose Zamoyska
- Ashworth Laboratories, Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
9
|
Abstract
Generating and maintaining a diverse repertoire of naive T cells is essential for protection against pathogens, and developing a mechanistic and quantitative description of the processes involved lies at the heart of our understanding of vertebrate immunity. Here, we review the biology of naive T cells from birth to maturity and outline how the integration of mathematical models and experiments has helped us to develop a full picture of their life histories.
Collapse
Affiliation(s)
- Benedict Seddon
- Institute of Immunity and Transplantation, Division of Infection and Immunity, UCL, Royal Free Hospital, London, UK
| | - Andrew J Yates
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York
| |
Collapse
|
10
|
Cytotoxic CD8 + T lymphocytes expressing ALS-causing SOD1 mutant selectively trigger death of spinal motoneurons. Proc Natl Acad Sci U S A 2019; 116:2312-2317. [PMID: 30674678 PMCID: PMC6369778 DOI: 10.1073/pnas.1815961116] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
CD8+ T lymphocytes, which are typically devoted to eliminate malignant and infected cells, have been described in the central nervous system (CNS) of patients and mice with amyotrophic lateral sclerosis (ALS). However, their role in ALS pathogenesis has yet to be unraveled. Here, we show that ablation of CD8+ T cells in ALS mice increased the number of surviving motoneurons. CD8+ T cells expressing the ALS-causing superoxide dismutase-1 mutant protein recognize and selectively kill motoneurons in vitro. To exert their cytotoxic function, mutant CD8+ T cells required presentation of the antigen-MHC-I complex at the surface of the motoneurons. Analysis of T cell receptor diversity supports the evidence that self-reactive CD8+ T lymphocytes infiltrate the CNS of ALS mice to exert cytotoxic function. Adaptive immune response is part of the dynamic changes that accompany motoneuron loss in amyotrophic lateral sclerosis (ALS). CD4+ T cells that regulate a protective immunity during the neurodegenerative process have received the most attention. CD8+ T cells are also observed in the spinal cord of patients and ALS mice although their contribution to the disease still remains elusive. Here, we found that activated CD8+ T lymphocytes infiltrate the central nervous system (CNS) of a mouse model of ALS at the symptomatic stage. Selective ablation of CD8+ T cells in mice expressing the ALS-associated superoxide dismutase-1 (SOD1)G93A mutant decreased spinal motoneuron loss. Using motoneuron-CD8+ T cell coculture systems, we found that mutant SOD1-expressing CD8+ T lymphocytes selectively kill motoneurons. This cytotoxicity activity requires the recognition of the peptide-MHC-I complex (where MHC-I represents major histocompatibility complex class I). Measurement of interaction strength by atomic force microscopy-based single-cell force spectroscopy demonstrated a specific MHC-I-dependent interaction between motoneuron and SOD1G93A CD8+ T cells. Activated mutant SOD1 CD8+ T cells produce interferon-γ, which elicits the expression of the MHC-I complex in motoneurons and exerts their cytotoxic function through Fas and granzyme pathways. In addition, analysis of the clonal diversity of CD8+ T cells in the periphery and CNS of ALS mice identified an antigen-restricted repertoire of their T cell receptor in the CNS. Our results suggest that self-directed immune response takes place during the course of the disease, contributing to the selective elimination of a subset of motoneurons in ALS.
Collapse
|
11
|
Rowe JH, Delmonte OM, Keles S, Stadinski BD, Dobbs AK, Henderson LA, Yamazaki Y, Allende LM, Bonilla FA, Gonzalez-Granado LI, Celikbilek Celik S, Guner SN, Kapakli H, Yee C, Pai SY, Huseby ES, Reisli I, Regueiro JR, Notarangelo LD. Patients with CD3G mutations reveal a role for human CD3γ in T reg diversity and suppressive function. Blood 2018; 131:2335-2344. [PMID: 29653965 PMCID: PMC5969384 DOI: 10.1182/blood-2018-02-835561] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 04/09/2018] [Indexed: 02/05/2023] Open
Abstract
Integrity of the T-cell receptor/CD3 complex is crucial for positive and negative selection of T cells in the thymus and for effector and regulatory functions of peripheral T lymphocytes. In humans, CD3D, CD3E, and CD3Z gene defects are a cause of severe immune deficiency and present early in life with increased susceptibility to infections. By contrast, CD3G mutations lead to milder phenotypes, mainly characterized by autoimmunity. However, the role of CD3γ in establishing and maintaining immune tolerance has not been elucidated. In this manuscript, we aimed to investigate abnormalities of T-cell repertoire and function in patients with genetic defects in CD3G associated with autoimmunity. High throughput sequencing was used to study composition and diversity of the T-cell receptor β (TRB) repertoire in regulatory T cells (Tregs), conventional CD4+ (Tconv), and CD8+ T cells from 6 patients with CD3G mutations and healthy controls. Treg function was assessed by studying its ability to suppress proliferation of Tconv cells. Treg cells of patients with CD3G defects had reduced diversity, increased clonality, and reduced suppressive function. The TRB repertoire of Tconv cells from patients with CD3G deficiency was enriched for hydrophobic amino acids at positions 6 and 7 of the CDR3, a biomarker of self-reactivity. These data demonstrate that the T-cell repertoire of patients with CD3G mutations is characterized by a molecular signature that may contribute to the increased rate of autoimmunity associated with this condition.
Collapse
Affiliation(s)
- Jared H Rowe
- Division of Hematology-Oncology, Boston Children's Hospital, Boston, MA
| | - Ottavia M Delmonte
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Sevgi Keles
- Division of Pediatric Immunology and Allergy, Meram Medical Faculty, Necmettin Erbakan University, Konya, Turkey
| | - Brian D Stadinski
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA
| | - Adam K Dobbs
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | | | - Yasuhiro Yamazaki
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | | | | | - Luis I Gonzalez-Granado
- Primary Immunodeficiencies Unit, Department of Pediatrics, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Seyma Celikbilek Celik
- Division of Pediatric Immunology and Allergy, Meram Medical Faculty, Necmettin Erbakan University, Konya, Turkey
| | - Sukru N Guner
- Division of Pediatric Immunology and Allergy, Meram Medical Faculty, Necmettin Erbakan University, Konya, Turkey
| | - Hasan Kapakli
- Division of Pediatric Immunology and Allergy, Meram Medical Faculty, Necmettin Erbakan University, Konya, Turkey
| | - Christina Yee
- Division of Immunology, Boston Children's Hospital, Boston, MA
| | - Sung-Yun Pai
- Division of Hematology-Oncology, Boston Children's Hospital, Boston, MA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA; and
| | - Eric S Huseby
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA
| | - Ismail Reisli
- Division of Pediatric Immunology and Allergy, Meram Medical Faculty, Necmettin Erbakan University, Konya, Turkey
| | - Jose R Regueiro
- Department of Immunology, Complutense University School of Medicine, imas12 Research Institute, Madrid, Spain
| | - Luigi D Notarangelo
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| |
Collapse
|
12
|
Rane S, Hogan T, Seddon B, Yates AJ. Age is not just a number: Naive T cells increase their ability to persist in the circulation over time. PLoS Biol 2018; 16:e2003949. [PMID: 29641514 PMCID: PMC5894957 DOI: 10.1371/journal.pbio.2003949] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 03/02/2018] [Indexed: 12/22/2022] Open
Abstract
The processes regulating peripheral naive T-cell numbers and clonal diversity remain poorly understood. Conceptually, homeostatic mechanisms must fall into the broad categories of neutral (simple random birth–death models), competition (regulation of cell numbers through quorum-sensing, perhaps via limiting shared resources), adaptation (involving cell-intrinsic changes in homeostatic fitness, defined as net growth rate over time), or selection (involving the loss or outgrowth of cell populations deriving from intercellular variation in fitness). There may also be stably maintained heterogeneity within the naive T-cell pool. To distinguish between these mechanisms, we confront very general models of these processes with an array of experimental data, both new and published. While reduced competition for homeostatic stimuli may impact cell survival or proliferation in neonates or under moderate to severe lymphopenia, we show that the only mechanism capable of explaining multiple, independent experimental studies of naive CD4+ and CD8+ T-cell homeostasis in mice from young adulthood into old age is one of adaptation, in which cells act independently and accrue a survival or proliferative advantage continuously with their post-thymic age. However, aged naive T cells may also be functionally impaired, and so the accumulation of older cells via ‘conditioning through experience’ may contribute to reduced immune responsiveness in the elderly. The body maintains large populations of naive T cells, a type of white blood cell that is able to respond specifically to pathogens. This arsenal is essential for our capacity to fight novel infections throughout our lifespan, and their numbers remain quite stable despite a gradual decline in the production of new naive T cells as we age. However, the mechanisms that underlie this stability are not well understood. In this study, we address this problem by testing a variety of potential mechanisms, each framed as a mathematical model, against multiple datasets obtained from experiments performed in mice. Our analysis supports a mechanism by which naïve T cells gradually increase their ability to survive the longer they reside in the circulation. Paradoxically, however, naïve T cells may also lose their ability to respond effectively to infections as they age. Together, these processes may drive the accumulation of older, functionally impaired T cells, potentially at the expense of younger and more immunologically potent cells, as we age.
Collapse
Affiliation(s)
- Sanket Rane
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York, United States of America
- Institute of Infection, Immunity & Inflammation, College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Thea Hogan
- Institute of Immunity and Transplantation, Division of Infection and Immunity, UCL, Royal Free Hospital, London, United Kingdom
| | - Benedict Seddon
- Institute of Immunity and Transplantation, Division of Infection and Immunity, UCL, Royal Free Hospital, London, United Kingdom
| | - Andrew J. Yates
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York, United States of America
- Institute of Infection, Immunity & Inflammation, College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, United Kingdom
- * E-mail:
| |
Collapse
|
13
|
Ellestad KK, Thangavelu G, Haile Y, Lin J, Boon L, Anderson CC. Prior to Peripheral Tolerance, Newly Generated CD4 T Cells Maintain Dangerous Autoimmune Potential: Fas- and Perforin-Independent Autoimmunity Controlled by Programmed Death-1. Front Immunol 2018; 9:12. [PMID: 29416537 PMCID: PMC5787554 DOI: 10.3389/fimmu.2018.00012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2017] [Accepted: 01/04/2018] [Indexed: 12/31/2022] Open
Abstract
Lymphopenia can result from various factors, including viral infections, clinical interventions, or as a normal property of the fetal/neonatal period. T cells in a lymphopenic environment undergo lymphopenia-induced proliferation (LIP) to fill the available “niche” as defined by peptide–MHC and homeostatic cytokine resources. We recently reported systemic autoimmunity following reconstitution of the lymphoid compartment of Rag1−/− mice with PD-1−/− hematopoietic stem cells or by transfer of thymocytes, but not splenocytes, suggesting that programmed death-1 (PD-1) plays a crucial role in controlling recent thymic emigrants (RTE) and preventing autoimmunity upon their LIP. However, it is unclear whether RTE residing within the periphery of a lymphoreplete host maintain enhanced autoimmune generating potential or if this property only manifests if RTE experience a lymphopenic periphery immediately after export from the thymus. Furthermore, it is unclear which of a variety of T cell effector mechanisms generate pathology when control of RTE by PD-1 is lacking. Herein, we determined that PD-1 is upregulated on CD4 T cells undergoing the natural LIP characteristic of the neonatal period. Newly generated T cells lacking PD-1 maintained an enhanced autoimmune potential even after residence in a lymphoreplete periphery, emphasizing the importance of PD-1 in the establishment of peripheral tolerance. Neither Fas nor perforin-dependent killing mechanisms were required for autoimmunity, while host MHC-II expression was critical, suggesting that LIP-driven autoimmunity in the absence of PD-1 may primarily result from a CD4 T cell-mediated systemic cytokinemia, a feature potentially shared by other autoimmune or inflammatory syndromes associated with immune reconstitution and LIP.
Collapse
Affiliation(s)
- Kristofor K Ellestad
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, Canada.,Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
| | - Govindarajan Thangavelu
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada.,Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Yohannes Haile
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
| | - Jiaxin Lin
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada.,Department of Surgery, University of Alberta, Edmonton, AB, Canada.,Alberta Transplant Institute, University of Alberta, Edmonton, AB, Canada
| | | | - Colin C Anderson
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, Canada.,Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada.,Department of Surgery, University of Alberta, Edmonton, AB, Canada.,Alberta Transplant Institute, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
14
|
Chuang E, Augustine M, Jung M, Schwartz RH, Singh NJ. Density dependent re-tuning of autoreactive T cells alleviates their pathogenicity in a lymphopenic environment. Immunol Lett 2017; 192:61-71. [PMID: 29111199 DOI: 10.1016/j.imlet.2017.10.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 10/04/2017] [Accepted: 10/09/2017] [Indexed: 10/18/2022]
Abstract
Peripheral T cell tolerance is challenging to induce in partially lymphopenic hosts and this is relevant for clinical situations involving transplant tolerance. While the shortage of regulatory cells is thought to be one reason for this, T cell-intrinsic tolerance processes such as anergy are also poorly triggered in such hosts. In order to understand the latter, we used a T cell deficient mouse model system where adoptively transferred autoreactive T cells are significantly tolerized in a cell intrinsic fashion, without differentiation to regulatory T cells. Intriguingly these T cells often retain sufficient effector functions to trigger autoimmune pathology. Here we find that the high population density of the autoreactive T cells that accumulated in such a host limits the progression of the cell-intrinsic tolerance process in T cells. Accordingly, reducing the cell density during a second transfer allowed T cells to further tune down their responsiveness to antigenic stimulation. The retuning of T cells was reflected by a loss of the T cell's abilities to proliferate, produces cytokines or help B cells. We further suggest, based on altering the levels of chronic antigen using miniosmotic pumps, that the effects of cell-density on T cell re-tuning may reflect the effective changes in the antigen dose perceived by individual T cells. This could proportionally elicit more negative feedback downstream of the TCR. Consistent with this, the retuned T cells showed signaling defects both proximal and distal to the TCR. Therefore, similar to the immunogenic activation of T cells, cell-intrinsic T cell tolerance may also involve a quantitative and progressive process of tuning down its antigen-responsiveness. The progress of such tuning seems to be stabilized at multiple intermediate stages by factors such as cell density, rather than just absolute antigen levels.
Collapse
Affiliation(s)
- Eleanore Chuang
- Laboratory of Cellular & Molecular Immunology, National Institute of Allergy & Infectious Diseases (NIAID), National Institutes of Health (NIH), 4/211, Center Drive, Bethesda, MD 20892-0420, United States; John A. Burns School of Medicine, Department of Tropical Medicine, Hawaii Center for AIDS, University of Hawaii, Honolulu, HI 96813, United States
| | - Marilyn Augustine
- Laboratory of Cellular & Molecular Immunology, National Institute of Allergy & Infectious Diseases (NIAID), National Institutes of Health (NIH), 4/211, Center Drive, Bethesda, MD 20892-0420, United States; University of Pittsburgh, Division of Endocrinology and Metabolism, 200 Lothrop Street, E1140 BST, Pittsburgh, PA 15261, United States
| | - Matthew Jung
- Laboratory of Cellular & Molecular Immunology, National Institute of Allergy & Infectious Diseases (NIAID), National Institutes of Health (NIH), 4/211, Center Drive, Bethesda, MD 20892-0420, United States; Washington University School of Medicine, Deptartment of Otolaryngology Head and Neck Surgery, 660 South Euclid Ave, St. Louis, MO 63110, United States
| | - Ronald H Schwartz
- Laboratory of Cellular & Molecular Immunology, National Institute of Allergy & Infectious Diseases (NIAID), National Institutes of Health (NIH), 4/211, Center Drive, Bethesda, MD 20892-0420, United States
| | - Nevil J Singh
- Laboratory of Cellular & Molecular Immunology, National Institute of Allergy & Infectious Diseases (NIAID), National Institutes of Health (NIH), 4/211, Center Drive, Bethesda, MD 20892-0420, United States; Department of Microbiology and Immunology, University of Maryland School of Medicine, HH 320A, 660 W Baltimore Street, Baltimore, MD 21201, United States.
| |
Collapse
|
15
|
Ellestad KK, Lin J, Boon L, Anderson CC. PD-1 Controls Tonic Signaling and Lymphopenia-Induced Proliferation of T Lymphocytes. Front Immunol 2017; 8:1289. [PMID: 29075267 PMCID: PMC5643416 DOI: 10.3389/fimmu.2017.01289] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 09/26/2017] [Indexed: 01/22/2023] Open
Abstract
Recovery of the T lymphocyte compartment within a lymphopenic host by lymphopenia-induced proliferation (LIP) is regulated by inter- and intraclonal competition for limited resources, including homeostatic cytokines and peptide:MHC (pMHC) complexes with which the TCR can interact at least weakly to yield a tonic signal. Importantly, the process of LIP can synergize with other factors that promote T cell activation to drive inflammatory disease. While reconstitution of the lymphoid compartment of immune deficient Rag-/- mice by transfer of wild-type hematopoietic stem cells (HSC) does not generally result in an overt disease phenotype, transfer of HSC deficient in expression of the co-inhibitory molecule PD-1 results in severe systemic autoimmunity driven by newly generated T cells that emerge from the thymus into the periphery and undergo LIP. Importantly, autoimmunity does not appear to depend on a response to exogenous (i.e., gut flora-derived) antigens. PD-1 is well known to be upregulated during T cell activation in response to cognate antigens, but it is unclear whether PD-1 has a role in controlling LIP of T cells in the absence of cognate antigen, i.e., in response to tonic pMHC. We examined whether PD-1 controls LIP of newly generated T cells by controlling the response to tonic pMHC or the homeostatic cytokine IL-7. We found that PD-1-deficient T cells have a proliferative advantage over WT T cells during LIP and this effect is MHC-II dependent and independent of IL-7Rα signaling. Furthermore, our data suggest that signals through IL-7Rα can be dispensable for LIP and may instead be of increased importance for T cell survival in conditions of high competition for limited pMHC (e.g., post-LIP, in a lymphoreplete host). We hypothesize that autoimmunity post-PD-1-/- HSC transplant is the result of an overzealous T cell response to normally tonic self-pMHC precipitated by the synergy of LIP and PD-1 deficiency. Furthermore, potentiation of TCR signals in response to normally tonic self-pMHC may contribute to the success of PD-1 blockade in cancer immunotherapy.
Collapse
Affiliation(s)
- Kristofor K Ellestad
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, Canada.,Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
| | - Jiaxin Lin
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada.,Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | | | - Colin C Anderson
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, Canada.,Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada.,Department of Surgery, University of Alberta, Edmonton, AB, Canada.,Alberta Transplant Institute, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
16
|
Zamora AE, Aguilar EG, Sungur CM, Khuat LT, Dunai C, Lochhead GR, Du J, Pomeroy C, Blazar BR, Longo DL, Venstrom JM, Baumgarth N, Murphy WJ. Licensing delineates helper and effector NK cell subsets during viral infection. JCI Insight 2017; 2:87032. [PMID: 28515356 DOI: 10.1172/jci.insight.87032] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 04/18/2017] [Indexed: 11/17/2022] Open
Abstract
Natural killer (NK) cells can be divided into phenotypic subsets based on expression of receptors that bind self-MHC-I molecules, a concept termed licensing or education. Here we show NK cell subsets with different migratory, effector, and immunoregulatory functions in dendritic cell and antigen (ag)-specific CD8+ T cell responses during influenza and murine cytomegalovirus infections. Shortly after infection, unlicensed NK cells localized in draining lymph nodes and produced GM-CSF, which correlated with the expansion and activation of dendritic cells, and resulted in greater and sustained ag-specific T cell responses. In contrast, licensed NK cells preferentially migrated to infected tissues and produced IFN-γ. Importantly, human NK cell subsets exhibited similar phenotypic characteristics. Collectively, our studies demonstrate a critical demarcation between the functions of licensed and unlicensed NK cell subsets, with the former functioning as the classical effector subset and the latter as the stimulator of adaptive immunity helping to prime immune responses.
Collapse
Affiliation(s)
| | | | | | | | | | - G Raymond Lochhead
- Department of Internal Medicine, UC Davis School of Medicine, Sacramento, California, USA
| | - Juan Du
- Department of Medicine, UCSF, San Francisco, California, USA
| | - Claire Pomeroy
- President of Lasker Foundation, Albert and Mary Lasker Foundation, New York City, New York, USA
| | - Bruce R Blazar
- Masonic Cancer Center and Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis, Minnesota, USA
| | - Dan L Longo
- National Institute on Aging, NIH, Baltimore, Maryland, USA
| | | | - Nicole Baumgarth
- Center for Comparative Medicine, UC Davis, Davis, California, USA
| | - William J Murphy
- Department of Dermatology.,Department of Internal Medicine, UC Davis School of Medicine, Sacramento, California, USA
| |
Collapse
|
17
|
Ellestad KK, Anderson CC. Two Strikes and You’re Out? The Pathogenic Interplay of Coinhibitor Deficiency and Lymphopenia-Induced Proliferation. THE JOURNAL OF IMMUNOLOGY 2017; 198:2534-2541. [DOI: 10.4049/jimmunol.1601884] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 01/10/2017] [Indexed: 12/15/2022]
|
18
|
Mellak S, Ait-Oufella H, Esposito B, Loyer X, Poirier M, Tedder TF, Tedgui A, Mallat Z, Potteaux S. Angiotensin II mobilizes spleen monocytes to promote the development of abdominal aortic aneurysm in Apoe-/- mice. Arterioscler Thromb Vasc Biol 2014; 35:378-88. [PMID: 25524776 DOI: 10.1161/atvbaha.114.304389] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
OBJECTIVE Abdominal aortic aneurysm (AAA) is widespread among elderly people and results in progressive expansion and rupture of the aorta with high mortality. Macrophages, which are the main population observed within the site of aneurysm, are thought to derive from circulating monocytes although no direct evidence has been provided to date. In this study, we were particularly interested in understanding the trafficking behavior of monocyte subsets in AAA and their role in disease pathogenesis. APPROACH AND RESULTS Using bone marrow transplantation in Apoe(-/-) mice, we showed that circulating monocytes give rise to abdominal aortic macrophages in hypercholesterolemic mice submitted to angiotensin II (AngII). Detailed monitoring of monocyte compartmentalization revealed that lymphocyte antigen 6C(high) and lymphocyte antigen 6C(low) monocytes transiently increase in blood early after AngII infusion and differentially infiltrate the abdominal aorta. The splenic reservoir accounted for the mobilization of the 2 monocyte subsets after 3 days of AngII infusion. Spleen removal or lymphocyte deficiency in Apoe(-/-) Rag2(-/-) mice similarly impaired early monocyte increase in blood in response to AngII and protected against AAA development, independently of blood pressure. Reconstitution of Apoe(-/-) Rag2(-/-) mice with total splenocytes but not with B-cell-depleted splenocytes restored monocyte mobilization in response to AngII and enhanced susceptibility to AAA. CONCLUSIONS Taken together, the data show that lymphocyte antigen 6C(high) and lymphocyte antigen 6C(low) monocytes are mobilized from the spleen in response to AngII. Intriguingly, the process is dependent on the presence of B cells and significantly contributes to the development of AAA and the occurrence of aortic rupture.
Collapse
Affiliation(s)
- Safa Mellak
- From the INSERM Unit UMR-S 970, Paris Cardiovascular Research Center (PARCC), Université Paris Descartes, Sorbonne Paris Cité, Paris, France (S.M., H.A.-O., B.E., X.L., M.P., A.T., Z.M., S.P.); Réanimation Médicale, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Saint-Antoine, Université Pierre-et-Marie Curie, Université Pierre-et-Marie Curie, Paris, France (H.A.-O.); Department of Immunology, Duke University Medical Center, Durham, NC (T.F.T.); and Department of Medicine, University of Cambridge, Cambridge, United Kingdom (Z.M.)
| | - Hafid Ait-Oufella
- From the INSERM Unit UMR-S 970, Paris Cardiovascular Research Center (PARCC), Université Paris Descartes, Sorbonne Paris Cité, Paris, France (S.M., H.A.-O., B.E., X.L., M.P., A.T., Z.M., S.P.); Réanimation Médicale, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Saint-Antoine, Université Pierre-et-Marie Curie, Université Pierre-et-Marie Curie, Paris, France (H.A.-O.); Department of Immunology, Duke University Medical Center, Durham, NC (T.F.T.); and Department of Medicine, University of Cambridge, Cambridge, United Kingdom (Z.M.)
| | - Bruno Esposito
- From the INSERM Unit UMR-S 970, Paris Cardiovascular Research Center (PARCC), Université Paris Descartes, Sorbonne Paris Cité, Paris, France (S.M., H.A.-O., B.E., X.L., M.P., A.T., Z.M., S.P.); Réanimation Médicale, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Saint-Antoine, Université Pierre-et-Marie Curie, Université Pierre-et-Marie Curie, Paris, France (H.A.-O.); Department of Immunology, Duke University Medical Center, Durham, NC (T.F.T.); and Department of Medicine, University of Cambridge, Cambridge, United Kingdom (Z.M.)
| | - Xavier Loyer
- From the INSERM Unit UMR-S 970, Paris Cardiovascular Research Center (PARCC), Université Paris Descartes, Sorbonne Paris Cité, Paris, France (S.M., H.A.-O., B.E., X.L., M.P., A.T., Z.M., S.P.); Réanimation Médicale, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Saint-Antoine, Université Pierre-et-Marie Curie, Université Pierre-et-Marie Curie, Paris, France (H.A.-O.); Department of Immunology, Duke University Medical Center, Durham, NC (T.F.T.); and Department of Medicine, University of Cambridge, Cambridge, United Kingdom (Z.M.)
| | - Maxime Poirier
- From the INSERM Unit UMR-S 970, Paris Cardiovascular Research Center (PARCC), Université Paris Descartes, Sorbonne Paris Cité, Paris, France (S.M., H.A.-O., B.E., X.L., M.P., A.T., Z.M., S.P.); Réanimation Médicale, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Saint-Antoine, Université Pierre-et-Marie Curie, Université Pierre-et-Marie Curie, Paris, France (H.A.-O.); Department of Immunology, Duke University Medical Center, Durham, NC (T.F.T.); and Department of Medicine, University of Cambridge, Cambridge, United Kingdom (Z.M.)
| | - Thomas F Tedder
- From the INSERM Unit UMR-S 970, Paris Cardiovascular Research Center (PARCC), Université Paris Descartes, Sorbonne Paris Cité, Paris, France (S.M., H.A.-O., B.E., X.L., M.P., A.T., Z.M., S.P.); Réanimation Médicale, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Saint-Antoine, Université Pierre-et-Marie Curie, Université Pierre-et-Marie Curie, Paris, France (H.A.-O.); Department of Immunology, Duke University Medical Center, Durham, NC (T.F.T.); and Department of Medicine, University of Cambridge, Cambridge, United Kingdom (Z.M.)
| | - Alain Tedgui
- From the INSERM Unit UMR-S 970, Paris Cardiovascular Research Center (PARCC), Université Paris Descartes, Sorbonne Paris Cité, Paris, France (S.M., H.A.-O., B.E., X.L., M.P., A.T., Z.M., S.P.); Réanimation Médicale, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Saint-Antoine, Université Pierre-et-Marie Curie, Université Pierre-et-Marie Curie, Paris, France (H.A.-O.); Department of Immunology, Duke University Medical Center, Durham, NC (T.F.T.); and Department of Medicine, University of Cambridge, Cambridge, United Kingdom (Z.M.)
| | - Ziad Mallat
- From the INSERM Unit UMR-S 970, Paris Cardiovascular Research Center (PARCC), Université Paris Descartes, Sorbonne Paris Cité, Paris, France (S.M., H.A.-O., B.E., X.L., M.P., A.T., Z.M., S.P.); Réanimation Médicale, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Saint-Antoine, Université Pierre-et-Marie Curie, Université Pierre-et-Marie Curie, Paris, France (H.A.-O.); Department of Immunology, Duke University Medical Center, Durham, NC (T.F.T.); and Department of Medicine, University of Cambridge, Cambridge, United Kingdom (Z.M.)
| | - Stéphane Potteaux
- From the INSERM Unit UMR-S 970, Paris Cardiovascular Research Center (PARCC), Université Paris Descartes, Sorbonne Paris Cité, Paris, France (S.M., H.A.-O., B.E., X.L., M.P., A.T., Z.M., S.P.); Réanimation Médicale, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Saint-Antoine, Université Pierre-et-Marie Curie, Université Pierre-et-Marie Curie, Paris, France (H.A.-O.); Department of Immunology, Duke University Medical Center, Durham, NC (T.F.T.); and Department of Medicine, University of Cambridge, Cambridge, United Kingdom (Z.M.).
| |
Collapse
|
19
|
Kaminitz A, Mizrahi K, Ash S, Ben-Nun A, Askenasy N. Stable activity of diabetogenic cells with age in NOD mice: dynamics of reconstitution and adoptive diabetes transfer in immunocompromised mice. Immunology 2014; 142:465-73. [PMID: 24601987 DOI: 10.1111/imm.12277] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2013] [Revised: 02/22/2014] [Accepted: 02/28/2014] [Indexed: 12/23/2022] Open
Abstract
The non-obese diabetic (NOD) mouse is a prevalent disease model of type 1 diabetes. Immune aberrations that cause and propagate autoimmune insulitis in these mice are being continually debated, with evidence supporting both dominance of effector cells and insufficiency of suppressor mechanisms. In this study we assessed the behaviour of NOD lymphocytes under extreme expansion conditions using adoptive transfer into immunocompromised NOD.SCID (severe combined immunodeficiency) mice. CD4(+) CD25(+) T cells do not cause islet inflammation, whereas splenocytes and CD4(+) CD25(-) T cells induce pancreatic inflammation and hyperglycaemia in 80-100% of the NOD.SCID recipients. Adoptively transferred effector T cells migrate to the lymphoid organs and pancreas, proliferate, are activated in the target organ in situ and initiate inflammatory insulitis. Reconstitution of all components of the CD4(+) subset emphasizes the plastic capacity of different cell types to adopt effector and suppressor phenotypes. Furthermore, similar immune profiles of diabetic and euglycaemic NOD.SCID recipients demonstrate dissociation between fractional expression of CD25 and FoxP3 and the severity of insulitis. There were no evident and consistent differences in diabetogenic activity and immune reconstituting activity of T cells from pre-diabetic (11 weeks) and new onset diabetic NOD females. Similarities in immune phenotypes and variable distribution of effector and suppressor subsets in various stages of inflammation commend caution in interpretation of quantitative and qualitative aberrations as markers of disease severity in adoptive transfer experiments.
Collapse
Affiliation(s)
- Ayelet Kaminitz
- Frankel Laboratory, Centre for Stem Cell Research, Schneider Children's Medical Centre of Israel, Petach Tikva, Israel; Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel
| | | | | | | | | |
Collapse
|
20
|
Martinet KZ, Bloquet S, Bourgeois C. Ageing combines CD4 T cell lymphopenia in secondary lymphoid organs and T cell accumulation in gut associated lymphoid tissue. IMMUNITY & AGEING 2014; 11:8. [PMID: 24829607 PMCID: PMC4020584 DOI: 10.1186/1742-4933-11-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Accepted: 04/29/2014] [Indexed: 12/11/2022]
Abstract
Background CD4 T cell lymphopenia is an important T cell defect associated to ageing. Higher susceptibility to infections, cancer, or autoimmune pathologies described in aged individuals is thought to partly rely on T cell lymphopenia. We hypothesize that such diverse effects may reflect anatomical heterogeneity of age related T cell lymphopenia. Indeed, no data are currently available on the impact of ageing on T cell pool recovered from gut associated lymphoid tissue (GALT), a crucial site of CD4 T cell accumulation. Results Primary, secondary and tertiary lymphoid organs of C57BL/6 animals were analysed at three intervals of ages: 2 to 6 months (young), 10 to 14 months (middle-aged) and 22 to 26 months (old). We confirmed that ageing preferentially impacted CD4 T cell compartment in secondary lymphoid organs. Importantly, a different picture emerged from gut associated mucosal sites: during ageing, CD4 T cell accumulation was progressively developing in colon and small intestine lamina propria and Peyer’s patches. Similar trend was also observed in middle-aged SJL/B6 F1 mice. Interestingly, an inverse correlation was detected between CD4 T cell numbers in secondary lymphoid organs and colonic lamina propria of C57BL/6 mice whereas no increase in proliferation rate of GALT CD4 T cells was detected. In contrast to GALT, no CD4 T cell accumulation was detected in lungs and liver in middle-aged animals. Finally, the concomitant accumulation of CD4 T cell in GALT and depletion in secondary lymphoid organs during ageing was detected both in male and female animals. Conclusions Our data thus demonstrate that T cell lymphopenia in secondary lymphoid organs currently associated to ageing is not sustained in gut or lung mucosa associated lymphoid tissues or non-lymphoid sites such as the liver. The inverse correlation between CD4 T cell numbers in secondary lymphoid organs and colonic lamina propria and the absence of overt proliferation in GALT suggest that marked CD4 T cell decay in secondary lymphoid organs during ageing reflect redistribution of CD4 T cells rather than generalized CD4 T cell decay. Such anatomical heterogeneity may provide an important rationale for the diversity of immune defects observed during ageing.
Collapse
Affiliation(s)
- Kim Zita Martinet
- INSERM U1012, Faculté de Médecine Paris-Sud, 63 rue Gabriel Péri, 94276 Le Kremlin-Bicêtre, France ; Univ Paris-Sud, UMR-S1012, Le Kremlin-Bicêtre, France
| | - Stéphane Bloquet
- Univ Paris-Sud, UMR-S1012, Le Kremlin-Bicêtre, France ; Animalerie centrale, Faculté de Médecine Paris-Sud, Univ Paris-Sud, Le Kremlin-Bicêtre, France
| | - Christine Bourgeois
- INSERM U1012, Faculté de Médecine Paris-Sud, 63 rue Gabriel Péri, 94276 Le Kremlin-Bicêtre, France ; Univ Paris-Sud, UMR-S1012, Le Kremlin-Bicêtre, France
| |
Collapse
|
21
|
Sckisel GD, Tietze JK, Zamora AE, Hsiao HH, Priest SO, Wilkins DEC, Lanier LL, Blazar BR, Baumgarth N, Murphy WJ. Influenza infection results in local expansion of memory CD8(+) T cells with antigen non-specific phenotype and function. Clin Exp Immunol 2014; 175:79-91. [PMID: 23937663 DOI: 10.1111/cei.12186] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/07/2013] [Indexed: 12/30/2022] Open
Abstract
Primary viral infections induce activation of CD8(+) T cells responsible for effective resistance. We sought to characterize the nature of the CD8(+) T cell expansion observed after primary viral infection with influenza. Infection of naive mice with different strains of influenza resulted in the rapid expansion of memory CD8(+) T cells exhibiting a unique bystander phenotype with significant up-regulation of natural killer group 2D (NKG2D), but not CD25, on the CD44(high) CD8(+) T cells, suggesting an antigen non-specific phenotype. We further confirmed the non-specificity of this phenotype on ovalbumin-specific (OT-I) CD8(+) T cells, which are not specific to influenza. These non-specific CD8(+) T cells also displayed increased lytic capabilities and were observed primarily in the lung. Thus, influenza infection was shown to induce a rapid, antigen non-specific memory T cell expansion which is restricted to the specific site of inflammation. In contrast, CD8(+) T cells of a similar phenotype could be observed in other organs following administration of systemic agonistic anti-CD40 and interleukin-2 immunotherapy, demonstrating that bystander expansion in multiple sites is possible depending on whether the nature of activation is either acute or systemic. Finally, intranasal blockade of NKG2D resulted in a significant increase in viral replication early during the course of infection, suggesting that NKG2D is a critical mediator of anti-influenza responses prior to the initiation of adaptive immunity. These results characterize further the local bystander expansion of tissue-resident, memory CD8(+) T cells which, due to their early induction, may play an important NKG2D-mediated, antigen non-specific role during the early stages of viral infection.
Collapse
Affiliation(s)
- Gail D Sckisel
- Department of Dermatology, School of Medicine, University of California, Davis, Sacramento, CA, USA; Graduate Group in Immunology, University of California, Davis, Davis, CA, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Pedroza-Pacheco I, Madrigal A, Saudemont A. Interaction between natural killer cells and regulatory T cells: perspectives for immunotherapy. Cell Mol Immunol 2013; 10:222-9. [PMID: 23524654 DOI: 10.1038/cmi.2013.2] [Citation(s) in RCA: 125] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Regulatory T (Treg) cells and natural killer (NK) cells are key players in the immune system. The interaction between these two cell types has been reported to be beneficial in healthy conditions such as pregnancy. However, in the case of certain pathologies such as autoimmune diseases and cancer this interaction can become detrimental, as Treg cells have been described to suppress NK cells and in particular to impair NK cell effector functions. This review aims to discuss the recent information on the interaction between Treg cells and NK cells under healthy and pathologic conditions, to describe the specific conditions in which this interaction takes place, the effect of Treg cells on hematopoietic stem cell differentiation and the consequences of this interaction on the optimization of immunotherapeutic protocols.
Collapse
Affiliation(s)
- Isabela Pedroza-Pacheco
- Anthony Nolan Research Institute and University College London, Royal Free Campus, London, UK
| | | | | |
Collapse
|
23
|
Méndez-Lagares G, Pozo-Balado MM, del Mar del Pozo Balado M, Genebat M, Genebat González M, García Pergañeda A, García Pergañeda Sánchez A, Leal M, Leal Noval M, Pacheco YM, Pacheco López YM. Severe immune dysregulation affects CD4⁺CD25(hi)FoxP3⁺ regulatory T cells in HIV-infected patients with low-level CD4 T-cell repopulation despite suppressive highly active antiretroviral therapy. J Infect Dis 2012; 205:1501-9. [PMID: 22457273 DOI: 10.1093/infdis/jis230] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
We hypothesized that CD4(+)CD25(hi)FoxP3(+) regulatory T cells (Tregs) could be involved in the high immune activation existing in patients with low-level CD4 T-cell repopulation under suppressive high active antiretroviral therapy (hereafter, "LLR patients"). Sixteen LLR patients, 18 human immunodeficiency virus (HIV)-infected controls (hereafter, "HIV controls"), and 16 healthy subjects were included. The frequency of CD4(+)CD25(hi)FoxP3(+) and HIV-specific Treg suppressive function were assessed. Relationships between Treg and CD4/CD8 activation (HLA-DR/CD38) and the frequency of naive CD4 T-cells were assessed. Low-level patients showed a higher Treg frequency but reduced HIV-specific immunosuppressive functions than HIV controls. Whereas in healthy subjects a strong negative correlation between Tregs and activated CD8 T cells emerged (r = -0.75, P < .001), it appeared disrupted in both HIV-infected groups (r = -0.06 and P = .83 for LLR patients; r = -0.11 and P = .68 for and HIV controls). Nevertheless, in LLR patients, Tregs negatively correlated with naive CD4 T cells (r = -0.60, P = .01), whereas there was no such correlation in HIV controls (r = -0.19, P = .46) or healthy subjects (r = -0.10, P = .73). Remarkably, a higher ratio of Tregs to naive CD4 T cells was observed in LLR patients than in HIV controls (P = .001) and healthy subjects (P < .001). We conclude that LLR patients have important alterations in immunoregulation involving CD4(+)CD25(hi)FoxP3(+) Tregs. In this scenario, the role of Tregs seems to be more related to the control of the naive CD4 T-cell homeostatic proliferation, rather than to the immune activation.
Collapse
Affiliation(s)
- Gema Méndez-Lagares
- Immunovirology Laboratory, Institute of Biomedicine of Seville, Infectious Diseases Service and Department of Clinical Biochemistry, Virgen del Rocío University Hospital, IBIS/CSIC/University of Seville, Spain
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
External influences on the immune system via activation of the aryl hydrocarbon receptor. Semin Immunol 2011; 23:99-105. [PMID: 21288737 DOI: 10.1016/j.smim.2011.01.008] [Citation(s) in RCA: 135] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2010] [Accepted: 01/10/2011] [Indexed: 02/06/2023]
Abstract
The aryl hydrocarbon receptor (AhR), subject of intensive research over three decades by the pharmacology/toxicology field has recently made its entry into mainstream immunology research and is set to continue to intrigue with ever more complex modes of modulating immune responses. The discovery of high and selective AhR expression on Th17 cells and its role in induction of the cytokine IL-22 attributed new immunological functions to this transcription factor and stimulated further research into physiological functions of the AhR in the immune system. A number of recent reviews have highlighted potential new avenues of research. This review addresses recent new insight into physiological roles of AhR in the immune system.
Collapse
|
25
|
Osborne LC, Patton DT, Seo JH, Abraham N. Elevated IL-7 Availability Does Not Account for T Cell Proliferation in Moderate Lymphopenia. THE JOURNAL OF IMMUNOLOGY 2011; 186:1981-8. [DOI: 10.4049/jimmunol.1002224] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
26
|
Simonetta F, Chiali A, Cordier C, Urrutia A, Girault I, Bloquet S, Tanchot C, Bourgeois C. Increased CD127 expression on activated FOXP3+CD4+ regulatory T cells. Eur J Immunol 2010; 40:2528-38. [PMID: 20690182 DOI: 10.1002/eji.201040531] [Citation(s) in RCA: 103] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Regulatory T cells (Treg) are commonly identified by CD25 (IL-2R alpha) surface expression and/or intracellular expression of the FOXP3 transcription factor. In addition, Treg are also characterized by low CD127 (IL-7R alpha) expression when compared to conventional T cells and their biology in the periphery is considered essentially independent of IL-7. We further investigated CD127 expression on Treg and we demonstrated differential CD127 expression depending on Treg subsets considered. Notably, we observed high CD127 expression on inducible costimulatory molecule (ICOS)- and CD103-expressing Treg subsets. Since these two markers reflect activation status, we addressed whether Treg activation modulated CD127 expression. We demonstrated that in contrast to conventional T cells, Treg significantly upregulated CD127 expression during in vitro and in vivo activation using adoptive transfer and contact dermatitis models. High CD127 expression on Treg was also predominantly detected ex vivo in some specific sites, notably bone marrow and skin. Importantly, higher CD127 expression on Treg correlated with higher phosphorylation of STAT5 upon IL-7 exposure. High CD127 expression on Treg also provided survival advantage upon in vitro incubation with IL-7. We thus demonstrated that low CD127 expression is not an intrinsic characteristic of Treg and we identified activated Treg as a potential target of endogenous or therapeutic IL-7.
Collapse
|
27
|
Askenasy EM, Askenasy N, Askenasy JJ. Does lymphopenia preclude restoration of immune homeostasis? The particular case of type 1 diabetes. Autoimmun Rev 2010; 9:687-90. [DOI: 10.1016/j.autrev.2010.05.019] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2010] [Accepted: 05/24/2010] [Indexed: 11/27/2022]
|
28
|
Immunosuppressive therapy exacerbates autoimmunity in NOD mice and diminishes the protective activity of regulatory T cells. J Autoimmun 2010; 35:145-52. [PMID: 20638242 DOI: 10.1016/j.jaut.2010.06.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2010] [Accepted: 06/13/2010] [Indexed: 01/29/2023]
Abstract
Mounting evidence indicates that immunosuppressive therapy and autologous bone marrow transplantation are relatively inefficient approaches to treat autoimmune diabetes. In this study we assessed the impact of immunosuppression on inflammatory insulitis in NOD mice, and the effect of radiation on immunomodulation mediated by adoptive transfer of various cell subsets. Sublethal radiation of NOD females at the age of 14 weeks (onset of hyperglycemia) delayed the onset of hyperglycemia, however two thirds of the mice became diabetic. Adoptive transfer of splenocytes into irradiated NON and NOD mice precipitated disease onset despite increased contents of CD25(+)FoxP3(+) T cells in the pancreas and regional lymphatics. Similar phenotypic changes were observed when CD25(+) T cells were infused after radiation, which also delayed disease onset without affecting its incidence. Importantly, irradiation increased the susceptibility to diabetes in NOD and NON mice (71-84%) as compared to immunomodulation with splenocytes and CD25(+) T cells in naïve recipients (44-50%). Although irradiation had significant and durable influence on pancreatic infiltrates and the fractions of functional CD25(+)FoxP3(+) Treg cells were elevated by adoptive cell transfer, this approach conferred no protection from disease progression. Irradiation was ineffective both in debulking of pathogenic clones and in restoring immune homeostasis, and the consequent homeostatic expansion evolves as an unfavorable factor in attempts to restore self-tolerance and might even provoke uncontrolled proliferation of pathogenic clones. The obstacles imposed by immunosuppression on abrogation of autoimmune insulitis require replacement of non-specific immunosuppressive therapy by selective immunomodulation that does not cause lymphopenia.
Collapse
|
29
|
Onoe T, Chittenden M, Zhao G, Yang YG, Sykes M. Homeostatic expansion and phenotypic conversion of human T cells depend on peripheral interactions with APCs. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2010; 184:6756-65. [PMID: 20483739 PMCID: PMC3086709 DOI: 10.4049/jimmunol.0901711] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Immune recovery in lymphopenic hosts depends largely on homeostatic peripheral expansion, especially when thymopoiesis is insufficient, as is often the case in human adults. Although it has been well studied in mice, the study of homeostatic peripheral expansion of human T cells has been limited by the lack of an appropriate in vivo model. In this study, we use T cell-deficient humanized mice and an adoptive transfer approach to demonstrate that two distinct proliferative responses of autologous T cells occur in vivo in a lymphopenic setting. Human naive CD4 and CD8 T cells that undergo rapid proliferation acquire a memory-like phenotype and the ability to rapidly produce IFN-gamma, whereas those undergoing slow proliferation retain naive phenotypic and functional characteristics. Recovery of both populations depends on the extent of human non-T cell chimerism in the periphery of recipient humanized mice. Furthermore, memory conversion of CD4 and CD8 T cells correlates with the level of human CD14+ and CD19+ chimerism in recipient mice, respectively, suggesting that different types of APCs support memory conversion of CD4 and CD8 T cells. Because lymphopenia affects clinical outcomes, this model, which will allow detailed investigation of the effects of lymphopenia in patients, is of clinical significance.
Collapse
Affiliation(s)
- Takashi Onoe
- Transplantation Biology Research Center, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
| | - Meredith Chittenden
- Transplantation Biology Research Center, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
| | - Guiling Zhao
- Transplantation Biology Research Center, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
| | - Yong-Guang Yang
- Transplantation Biology Research Center, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
| | - Megan Sykes
- Transplantation Biology Research Center, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
| |
Collapse
|
30
|
Feng T, Wang L, Schoeb TR, Elson CO, Cong Y. Microbiota innate stimulation is a prerequisite for T cell spontaneous proliferation and induction of experimental colitis. J Exp Med 2010; 207:1321-32. [PMID: 20498021 PMCID: PMC2882839 DOI: 10.1084/jem.20092253] [Citation(s) in RCA: 183] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2009] [Accepted: 04/15/2010] [Indexed: 11/04/2022] Open
Abstract
Little is known about how the microbiota regulates T cell proliferation and whether spontaneous T cell proliferation is involved in the pathogenesis of inflammatory bowel disease. In this study, we show that stimulation of innate pathways by microbiota-derived ligands and antigen-specific T cell stimulation are both required for intestinal inflammation. Microbiota-derived ligands promoted spontaneous T cell proliferation by activating dendritic cells (DCs) to produce IL-6 via Myd88, as shown by the spontaneous proliferation of T cells adoptively transferred into specific pathogen-free (SPF) RAG-/- mice, but not in germfree RAG-/- mice. Reconstitution of germfree RAG-/- mice with cecal bacterial lysate-pulsed DCs, but not with IL-6-/- or Myd88-/- DCs, restored spontaneous T cell proliferation. CBir1 TCR transgenic (CBir1 Tg) T cells, which are specific for an immunodominant microbiota antigen, induced colitis in SPF RAG-/- mice. Blocking the spontaneous proliferation of CBir1 Tg T cells by co-transferring bulk OT II CD4+ T cells abrogated colitis development. Although transferred OT II T cells underwent spontaneous proliferation in RAG-/- mice, the recipients failed to develop colitis because of the lack of cognate antigen in the intestinal lumen. Collectively, our data demonstrate that induction of colitis requires both spontaneous proliferation of T cells driven by microbiota-derived innate signals and antigen-specific T cell proliferation.
Collapse
Affiliation(s)
- Ting Feng
- Department of Microbiology, Division of Gastroenterology and Hepatology, Department of Medicine, and Department of Genetics, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Lanfang Wang
- Department of Microbiology, Division of Gastroenterology and Hepatology, Department of Medicine, and Department of Genetics, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Trenton R. Schoeb
- Department of Microbiology, Division of Gastroenterology and Hepatology, Department of Medicine, and Department of Genetics, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Charles O. Elson
- Department of Microbiology, Division of Gastroenterology and Hepatology, Department of Medicine, and Department of Genetics, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Yingzi Cong
- Department of Microbiology, Division of Gastroenterology and Hepatology, Department of Medicine, and Department of Genetics, University of Alabama at Birmingham, Birmingham, AL 35294
| |
Collapse
|
31
|
Winstead CJ, Reilly CS, Moon JJ, Jenkins MK, Hamilton SE, Jameson SC, Way SS, Khoruts A. CD4+CD25+Foxp3+ regulatory T cells optimize diversity of the conventional T cell repertoire during reconstitution from lymphopenia. THE JOURNAL OF IMMUNOLOGY 2010; 184:4749-60. [PMID: 20357265 DOI: 10.4049/jimmunol.0904076] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The functional capacity of the adaptive immune system is dependent on the size and the diversity of the T cell population. In states of lymphopenia, T cells are driven to proliferate to restore the T cell population size. However, different T cell clones proliferate at different rates, and some T cells experience burst-like expansion called spontaneous lymphopenia-induced proliferation (LIP). These T cells are likely receiving stimulation from cognate Ags and are most responsible for inflammatory pathology that can emerge in lymphopenic states. Foxp3(+) regulatory T cells (Tregs) selectively inhibit spontaneous LIP, which may contribute to their ability to prevent lymphopenia-associated autoimmunity. We hypothesized that another potential negative consequence of unrestrained spontaneous LIP is constriction of the total T cell repertoire. We demonstrate that the absence of Foxp3(+) Tregs during the period of immune reconstitution results in the development of TCR repertoire "holes" and the loss of Ag-specific responsiveness to infectious microorganisms. In contrast, the presence of Tregs during the period of immune reconstitution preserves optimal TCR diversity and foreign Ag responsiveness. This finding contrasts with the generally accepted immunosuppressive role of Tregs and provides another example of Treg activity that actually enhances immune function.
Collapse
Affiliation(s)
- Colleen J Winstead
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55414, USA
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Bosco N, Swee LK, Bénard A, Ceredig R, Rolink A. Auto-reconstitution of the T-cell compartment by radioresistant hematopoietic cells following lethal irradiation and bone marrow transplantation. Exp Hematol 2010; 38:222-232.e2. [DOI: 10.1016/j.exphem.2009.12.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2009] [Revised: 11/24/2009] [Accepted: 12/23/2009] [Indexed: 10/20/2022]
|
33
|
Lymphocyte proliferation in immune-mediated diseases. Trends Immunol 2009; 30:430-8. [PMID: 19699149 DOI: 10.1016/j.it.2009.06.002] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2009] [Revised: 06/21/2009] [Accepted: 06/24/2009] [Indexed: 12/21/2022]
Abstract
Defects in T cell homeostatic mechanisms can result in T cell lymphopenia, defined as decreased numbers of lymphocytes. Lymphopenia results in homeostatic proliferation in order to maintain T cell homeostasis. It has been proposed that homeostatic proliferation can expand the pool of autoreactive T cells that promote autoimmunity, and indeed recent studies have further substantiated this observation in both animal models and humans. Conversely, homeostatic proliferation can promote tumor immunity by allowing tumor-specific T cells to accumulate. In this review, we discuss how the outcome of homeostatic proliferation can function both in a deleterious manner in autoimmunity and a beneficial way in tumor immunity. We also discuss the roles of various cytokines and T regulatory cells that control homeostatic proliferation.
Collapse
|
34
|
Sojka DK, Hughson A, Fowell DJ. CTLA-4 is required by CD4+CD25+ Treg to control CD4+ T-cell lymphopenia-induced proliferation. Eur J Immunol 2009; 39:1544-51. [PMID: 19462377 PMCID: PMC2835300 DOI: 10.1002/eji.200838603] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
CTLA-4 is constitutively expressed by CD4(+)CD25(+)Foxp3(+) Treg but its precise role in Treg function is not clear. Although blockade of CTLA-4 interferes with Treg function, studies using CTLA-4-deficient Treg have failed to reveal an essential requirement for CTLA-4 in Treg suppression in vivo. Conditional deletion of CTLA-4 in Foxp3(+) T cells disrupts immune homeostasis in vivo but the immune processes disrupted by CTLA-4 deletion have not been determined. We demonstrate that Treg expression of CTLA-4 is essential for Treg control of lymphopenia-induced CD4 T-cell expansion. Despite IL-10 expression, CTLA-4-deficient Treg were unable to control the expansion of CD4(+) target cells in a lymphopenic environment. Moreover, unlike their WT counterparts, CTLA-4-deficient Treg failed to inhibit cytokine production associated with homeostatic expansion and were unable to prevent colitis. Thus, while Treg developing in the absence of CTLA-4 appear to acquire some compensatory suppressive mechanisms in vitro, we identify a non-redundant role for CTLA-4 in Treg function in vivo.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- Antigens, CD/metabolism
- Antigens, CD/physiology
- Antigens, Surface/metabolism
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- CD4-Positive T-Lymphocytes/transplantation
- CTLA-4 Antigen
- Cell Proliferation/drug effects
- Colitis/immunology
- Colitis/prevention & control
- Cytokines/genetics
- Cytokines/metabolism
- DNA-Binding Proteins/genetics
- Enzyme Inhibitors/pharmacology
- Immune Tolerance
- Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors
- Interleukin-2 Receptor alpha Subunit/metabolism
- Lymph Nodes/cytology
- Lymph Nodes/immunology
- Lymphopenia/genetics
- Lymphopenia/immunology
- Lymphopenia/pathology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Receptors, Transforming Growth Factor beta/genetics
- Spleen/cytology
- Spleen/immunology
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Tryptophan/analogs & derivatives
- Tryptophan/pharmacology
- Lymphocyte Activation Gene 3 Protein
Collapse
Affiliation(s)
- Dorothy K Sojka
- David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, Department of Microbiology and Immunology, University of Rochester, Rochester, NY 14642, USA
| | | | | |
Collapse
|
35
|
Depletion of CD4 T cells enhances immunotherapy for neuroblastoma after syngeneic HSCT but compromises development of antitumor immune memory. Blood 2009; 113:4449-57. [PMID: 19182203 DOI: 10.1182/blood-2008-11-190827] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
High-risk neuroblastoma remains a clinically challenging disease. Here, we report that a multifaceted immunotherapeutic approach including syngeneic hematopoietic stem cell transplantation (HSCT), adoptive transfer of sensitized T cells (from syngeneic donors vaccinated to tumor antigens), and early posttransplantation tumor vaccination can effectively treat mice with established neuroblastoma. Vaccination was an important component of this immunotherapy, as it resulted in enhanced and prolonged tumor-specific CD8 T-cell activity and improved antitumor efficacy. Surprisingly, CD4 cell depletion of mice given sensitized T cells resulted in better tumor-free survival, which was associated with an early increased expansion of CD8 T cells with an effector phenotype, increased numbers of tumor-reactive CD8 T cells, and increased tumor infiltration by CD8 T cells. However, in the absence of CD4 T cells, development of long-term tumor immunity (memory) was severely compromised as reflected by diminished CD8 T-cell recall responses and an inability to resist tumor rechallenge in vivo. Based on these results, a major challenge with this immunotherapeutic approach is how to obtain the ideal initial antitumor response but still preserve antitumor immune memory. These data suggest that identification and selective depletion of immune inhibitory CD4 T cells may be a strategy to enhance early antitumor immunity and induce a long-lasting tumor response after HSCT.
Collapse
|
36
|
Burchell JT, Wikstrom ME, Stumbles PA, Sly PD, Turner DJ. Attenuation of allergen-induced airway hyperresponsiveness is mediated by airway regulatory T cells. Am J Physiol Lung Cell Mol Physiol 2008; 296:L307-19. [PMID: 19028981 DOI: 10.1152/ajplung.00521.2007] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Understanding the mechanisms involved in respiratory tolerance to inhaled allergens could potentially result in improved therapies for asthma and allergic diseases. Airway hyperresponsiveness (AHR) is a major feature of allergic asthma, thus the aim of the current study was to investigate mechanisms underlying suppression of allergen-induced AHR during chronic allergen exposure. Adult BALB/c mice were systemically sensitized with ovalbumin (OVA) in adjuvant and then challenged with a single 3 or 6 wk of OVA aerosols. Airway and parenchymal responses to inhaled methacholine (MCh), inflammatory cell counts, cytokines, OVA-specific IgE and IgG(1), parenchymal histology, and numbers of airway CD4(+)69(+) activated and CD4(+)25(+)FoxP3(+) regulatory T (Treg) cells were assessed 24 h after the final aerosol. Single OVA challenge resulted in AHR, eosinophilia, increased serum OVA-specific IgE, and T helper 2 (Th2) cytokines in bronchoalveolar lavage (BAL) but no difference in numbers of Treg compared with control mice. Three weeks of OVA challenges resulted in suppression of AHR and greater numbers of airway Treg cells and increased transforming growth factor-beta(1) (TGFbeta(1)) compared with control mice despite the presence of increased eosinophilia, OVA-specific IgE and IgG(1), and airway remodeling. Six weeks of OVA challenges restored AHR, whereas airway Treg numbers, TGFbeta(1), BAL eosinophilia, and Th2 cytokines returned to control levels. Partial in vivo depletion or adoptive transfer of Treg cells restored or inhibited AHR, respectively, but did not affect TGFbeta(1) or Th2 cytokine production. In conclusion, AHR suppression is mediated by airway Treg cells and potentially via a paracrine induction of TGFbeta(1) in the airways.
Collapse
|
37
|
Host CD4+CD25+ T cells can expand and comprise a major component of the Treg compartment after experimental HCT. Blood 2008; 113:733-43. [PMID: 18832651 DOI: 10.1182/blood-2008-08-173179] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Reconstitution of the recipient lymphoid compartment following hematopoietic cell transplantation (HCT) is typically delayed. The present studies investigated the residual host CD4(+)CD25(+)Foxp3(+) (Treg) compartment after several conditioning regimens, including T cell-depleted and T cell-replete HCT and observed (1) a small number of recipient Treg cells survived aggressive conditioning; (2) the surviving, that is, residual Tregs underwent marked expansion; and (3) recipient CD4(+)FoxP3(+) cells composed the majority of the Treg compartment for several months post-syngeneic HCT. Notably, residual Tregs also dominated the compartment post-HCT with T cell-depleted (TCD) major histocompatibility complex-matched allogeneic bone marrow but not following T cell-replete transplantations. The residual Treg cell compartment was functionally competent as assessed by in vitro lymphoid suppression and in vivo autoimmune disease transfer assay. These observations support the notion that functional host Tregs initially occupy a niche in lymphopenic transplantation recipients, undergo significant expansion, and contribute to the compartment for an extended period before donor-derived CD4(+)FoxP3(+) T cells eventually compose the majority of the compartment. In total, the findings suggest that the presence of host Tregs may be important to consider regarding elicitation of immune (eg, antitumor, vaccine) responses in recipients during the early post-transplant period involving autologous and certain allogeneic HCT regimens.
Collapse
|
38
|
Bourgeois C, Hao Z, Rajewsky K, Potocnik AJ, Stockinger B. Ablation of thymic export causes accelerated decay of naive CD4 T cells in the periphery because of activation by environmental antigen. Proc Natl Acad Sci U S A 2008; 105:8691-6. [PMID: 18562288 PMCID: PMC2438401 DOI: 10.1073/pnas.0803732105] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2008] [Indexed: 12/30/2022] Open
Abstract
A model of chemical thymectomy by inducible Rag ablation was used to study peripheral T cell homeostasis. Induction of Rag ablation was efficient and complete, leading to cessation of thymic T cell production within 3-4 weeks. The decay of peripheral T cells became apparent with a delay of an additional 2-3 weeks and was entirely accounted for by loss of naïve T cells, whereas numbers of memory phenotype and regulatory T cells were not decreased. Naïve CD4 T cells decayed with an average half-life of 50 days, whereas naïve CD8 T cells exhibited a considerably longer half-life. The rapid decay of naïve CD4 T cells was not caused by intrinsic survival differences compared with naïve CD8 T cells, but was caused by changes in the lymphopenic environment resulting in higher microbial load and consequential activation. This finding suggests that in lymphopenic conditions involving compromised thymic function replenishment and survival of a naïve CD4 T cell repertoire may be severely curtailed because of chronic activation. Such a scenario might play a role in the aging immune system and chronic viral infection, such as HIV infection, and contribute to loss of CD4 T cells and impaired immune function. As our data show, continued replenishment with cells from the thymus seems to be required to maintain efficient gut mucosal defense.
Collapse
Affiliation(s)
- Christine Bourgeois
- *Division of Molecular Immunology, The National Institute for Medical Research, The Ridgeway, Mill Hill, London NW7 1AA, United Kingdom
| | - Zhenyue Hao
- The Campbell Family Institute for Breast Cancer Research, Ontario Cancer Research Institute, Toronto, ON, Canada M5G2C1; and
| | - Klaus Rajewsky
- Immune Disease Institute, Harvard Medical School, 200 Longwood Avenue, Boston, MA 02115
| | - Alexandre J. Potocnik
- *Division of Molecular Immunology, The National Institute for Medical Research, The Ridgeway, Mill Hill, London NW7 1AA, United Kingdom
| | - Brigitta Stockinger
- *Division of Molecular Immunology, The National Institute for Medical Research, The Ridgeway, Mill Hill, London NW7 1AA, United Kingdom
| |
Collapse
|
39
|
Thomas-Vaslin V, Altes HK, de Boer RJ, Klatzmann D. Comprehensive assessment and mathematical modeling of T cell population dynamics and homeostasis. THE JOURNAL OF IMMUNOLOGY 2008; 180:2240-50. [PMID: 18250431 DOI: 10.4049/jimmunol.180.4.2240] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Our current view of T cell differentiation and population dynamics is assembled from pieces of data obtained from separate experimental systems and is thus patchy. We reassessed homeostasis and dynamics of T cells 1) by generating a mathematical model describing the spatiotemporal features of T cell differentiation, and 2) by fitting this model to experimental data generated by disturbing T cell differentiation through transient depletion of dividing T cells in mice. This specific depletion was obtained by administration of ganciclovir to mice expressing the conditional thymidine kinase suicide gene in T cells. With this experimental approach, we could derive quantitative parameters describing the cell fluxes, residence times, and rates of import, export, proliferation, and death across cell compartments for thymocytes and recent thymic emigrants (RTEs). Among other parameters, we show that 93% of thymocytes produced before single-positive stages are eliminated through the selection process. Then, a postselection peripheral expansion of naive T cells contributes three times more to naive T cell production than the thymus, with half of the naive T cells consisting of dividing RTEs. Altogether, this work provides a quantitative population dynamical framework of thymocyte development, RTEs, and naive T cells.
Collapse
Affiliation(s)
- Véronique Thomas-Vaslin
- Unité Mixte de Recherche 7087, Biologie et Thérapeutique des Pathologies Immunitaires, Université Pierre et Marie Curie-Paris 06, 83 Boulevard de l'Hôpital, Paris, France.
| | | | | | | |
Collapse
|
40
|
Wan N, Dai H, Wang T, Moore Y, Zheng XX, Dai Z. Bystander central memory but not effector memory CD8+ T cells suppress allograft rejection. THE JOURNAL OF IMMUNOLOGY 2008; 180:113-21. [PMID: 18097010 DOI: 10.4049/jimmunol.180.1.113] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Memory T cells respond faster and more vigorously than their naive counterparts and are critical for adaptive immunity. However, it is unknown whether and how memory T cells react in the face of irrelevant Ags. It is generally accepted that bystander memory T cells are neutral in immune responsiveness. In this study, we present the first evidence that bystander central memory (TCM), but not effector memory (TEM), CD8+ T cells suppress allograft rejection as well as T cell proliferation in the draining lymph nodes (DLN) of recipient mice. Both bystander TCM and naive T cells, but fewer TEM cells, migrated to DLN, whereas TCM cells exhibited faster turnover than their naive counterparts, suggesting that bystander TCM cells have an advantage over their naive counterparts in suppression. However, bystander TEM cells migrated to inflammatory graft sites, but not DLN, and yet failed to exert their suppression. These findings indicate that bystander memory T cells need to migrate to lymph nodes to exert their suppression by inhibiting responder T cell activation or homeostatic proliferation. Moreover, the suppression mediated by bystander TCM cells was largely dependent on IL-15, as IL-15 was required for their homeostatic proliferation and TCM-mediated suppression of allograft rejection. This suppression also required the presence of TGFbeta1, as TCM cells expressed TGFbeta1 while neutralizing TGFbeta1 abolished their suppression. Thus, bystander TCM, but not TEM, CD8+ T cells are potent suppressors rather than bystanders. This new finding will have an impact on cellular immunology and may have clinic implications for tolerance induction.
Collapse
Affiliation(s)
- Ni Wan
- Center for Biomedical Research, University of Texas Health Center, Tyler, TX 75708, USA
| | | | | | | | | | | |
Collapse
|
41
|
Brinkman DMC, Jol-van der Zijde CM, ten Dam MM, te Boekhorst PAW, ten Cate R, Wulffraat NM, Hintzen RQ, Vossen JM, van Tol MJD. Resetting the adaptive immune system after autologous stem cell transplantation: lessons from responses to vaccines. J Clin Immunol 2007; 27:647-58. [PMID: 17690955 PMCID: PMC2075528 DOI: 10.1007/s10875-007-9120-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2007] [Accepted: 07/06/2007] [Indexed: 12/29/2022]
Abstract
Autologous stem cell transplantation (ASCT) to treat autoimmune diseases (AID) is thought to reset immunological memory directed against autoantigens. This hypothesis can only be studied indirectly because the exact nature of the pathogenetic autoantigens is unknown in most AID. Therefore, 19 children with juvenile idiopathic arthritis (JIA) or systemic lupus erythematodes (SLE) and 10 adults with multiple sclerosis (MS) were vaccinated with the T-cell-dependent neoantigen rabies and the recall antigen tetanus toxoid after, respectively before, bone marrow harvest. Both vaccinations were repeated after ASCT. All except two of the responders mounted a primary antibody response to rabies after revaccination, and 44% of the responders mounted a primary antibody response to tetanus boost after ASCT. These data show that immunological memory to a neoantigen is lost in most patients with AID after immunoablative pretreatment; however, memory to a recall antigen boosted before bone marrow harvest is only lost in part of the patients. Disease progression was arrested in all patients with JIA/SLE except one, but only in a minority of MS patients. Clinical outcome on a per case basis was not associated with the profile of the immune response toward the vaccination antigens after ASCT.
Collapse
Affiliation(s)
- D M C Brinkman
- Department of Pediatrics, Leiden University Medical Center, 2300 RC, Leiden, The Netherlands.
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Bourgeois C, Stockinger B. T cell homeostasis in steady state and lymphopenic conditions. Immunol Lett 2006; 107:89-92. [PMID: 16963128 DOI: 10.1016/j.imlet.2006.08.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2006] [Revised: 08/02/2006] [Accepted: 08/02/2006] [Indexed: 11/23/2022]
Abstract
Homeostasis in the immune system is an important principle ensuring that the numbers of peripheral lymphocytes are kept more or less constant despite numerous disturbances in the immune system during the lifetime of an organism. Mechanisms relating to maintenance of homeostasis have mainly been investigated in experimental systems exhibiting extreme lymphopenic conditions in which the behavior of adoptively transferred lymphocytes is assessed in the absence of endogenous lymphocytes. While these experimental systems have yielded important insight into mechanisms that shape the survival and expansion potential of T cells, their extrapolation to normal steady state conditions that do not involve extreme lymphopenia has sometimes been difficult. We review here the different scenarios of homeostatic control mechanisms in steady state as well as severely or partially lymphopenic environments.
Collapse
Affiliation(s)
- Christine Bourgeois
- Division of Molecular Immunology, MRC National Institute for Medical Research, The Ridgeway, Mill Hill, London NW7 1AA, UK
| | | |
Collapse
|