1
|
Martins YC, Rosa-Gonçalves P, Daniel-Ribeiro CT. Theories of immune recognition: Is anybody right? Immunology 2024; 173:274-285. [PMID: 39034280 DOI: 10.1111/imm.13839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 07/04/2024] [Indexed: 07/23/2024] Open
Abstract
The clonal selection theory (CST) is the centrepiece of the current paradigm used to explain immune recognition and memory. Throughout the past decades, the original CST had been expanded and modified to explain new experimental evidences since its original publication by Burnet. This gave origin to new paradigms that govern experimental immunology nowadays, such as the associative recognition of antigen model and the stranger/danger signal model. However, these new theories also do not fully explain experimental findings such as natural autoimmune immunoglobulins, idiotypic networks, low and high dose tolerance, and dual-receptor T and B cells. To make sense of these empirical data, some authors have been trying to change the paradigm of immune cognition using a systemic approach, analogies with brain processing and concepts from second-order cybernetics. In the present paper, we review the CST and some of the theories/hypotheses derived from it, focusing on immune recognition. We point out their main weaknesses and highlight arguments made by their opponents and believers. We conclude that, until now, none of the proposed theories can fully explain the totality of immune phenomena and that a theory of everything is needed in immunology.
Collapse
Affiliation(s)
- Yuri Chaves Martins
- Department of Anesthesiology, Saint Louis University School of Medicine, Saint Louis, Missouri, USA
| | - Pamela Rosa-Gonçalves
- Laboratório de Pesquisa em Malária, Instituto Oswaldo Cruz and Centro de Pesquisa, Diagnóstico e Treinamento em Malária, Fiocruz, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Cláudio Tadeu Daniel-Ribeiro
- Laboratório de Pesquisa em Malária, Instituto Oswaldo Cruz and Centro de Pesquisa, Diagnóstico e Treinamento em Malária, Fiocruz, Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
2
|
Uhlemann H, Epp K, Klesse C, Link-Rachner CS, Surendranath V, Günther UP, Schetelig J, Heidenreich F. Shape of the art: TCR-repertoire after allogeneic hematopoietic cell transplantation. Best Pract Res Clin Haematol 2024; 37:101558. [PMID: 39098804 DOI: 10.1016/j.beha.2024.101558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 05/03/2024] [Accepted: 06/27/2024] [Indexed: 08/06/2024]
Abstract
The human adaptive immune repertoire is characterized by specificity and diversity to provide immunity against past and future tasks. Such tasks are mainly infections but also malignant transformations of cells. With its multiple lines of defense, the human immune system contains both, rapid reaction forces and the potential to capture, disassemble and analyze strange structures in order to teach the adaptive immune system and mount a specific immune response. Prevention and mitigation of autoimmunity is of equal importance. In the context of allogeneic hematopoietic cell transplantation (HCT) specific challenges exist with the transfer of cells from the adapted donor immune system to the immunosuppressed recipient. Those challenges are immunogenetic disparity between donor and host, reconstitution of immunity early after HCT by expansion of mature immune effector cells, and impaired thymic function, if the recipient is an adult (as it is the case in most HCTs). The possibility to characterize the adaptive immune repertoire by massively parallel sequencing of T-cell receptor gene rearrangements allows for a much more detailed characterization of the T-cell repertoire. In addition, high-dimensional characterization of immune effector cells based on their immunophenotype and single cell RNA sequencing allow for much deeper insights in adaptive immune responses. We here review, existing - still incomplete - information on immune reconstitution after allogeneic HCT. Building on the technological advances much deeper insights into immune recovery after HCT and adaptive immune responses and can be expected in the coming years.
Collapse
Affiliation(s)
- Heike Uhlemann
- University Hospital Carl Gustav Carus, Dresden, Germany; DKMS Group gGmbH, Clinical Trials Unit, Dresden, Germany.
| | - Katharina Epp
- University Hospital Carl Gustav Carus, Dresden, Germany
| | | | | | | | | | - Johannes Schetelig
- University Hospital Carl Gustav Carus, Dresden, Germany; DKMS Group gGmbH, Clinical Trials Unit, Dresden, Germany
| | - Falk Heidenreich
- University Hospital Carl Gustav Carus, Dresden, Germany; DKMS Group gGmbH, Clinical Trials Unit, Dresden, Germany
| |
Collapse
|
3
|
Tian G, Li M, Lv G. Analysis of T-Cell Receptor Repertoire in Transplantation: Fingerprint of T Cell-mediated Alloresponse. Front Immunol 2022; 12:778559. [PMID: 35095851 PMCID: PMC8790170 DOI: 10.3389/fimmu.2021.778559] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 12/22/2021] [Indexed: 11/13/2022] Open
Abstract
T cells play a key role in determining allograft function by mediating allogeneic immune responses to cause rejection, and recent work pointed their role in mediating tolerance in transplantation. The unique T-cell receptor (TCR) expressed on the surface of each T cell determines the antigen specificity of the cell and can be the specific fingerprint for identifying and monitoring. Next-generation sequencing (NGS) techniques provide powerful tools for deep and high-throughput TCR profiling, and facilitate to depict the entire T cell repertoire profile and trace antigen-specific T cells in circulation and local tissues. Tailing T cell transcriptomes and TCR sequences at the single cell level provides a full landscape of alloreactive T-cell clones development and biofunction in alloresponse. Here, we review the recent advances in TCR sequencing techniques and computational tools, as well as the recent discovery in overall TCR profile and antigen-specific T cells tracking in transplantation. We further discuss the challenges and potential of using TCR sequencing-based assays to profile alloreactive TCR repertoire as the fingerprint for immune monitoring and prediction of rejection and tolerance.
Collapse
Affiliation(s)
| | - Mingqian Li
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun, China
| | - Guoyue Lv
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
4
|
TCRα reporter mice reveal contribution of dual TCRα expression to T cell repertoire and function. Proc Natl Acad Sci U S A 2020; 117:32574-32583. [PMID: 33288689 DOI: 10.1073/pnas.2013188117] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
It is known that a subpopulation of T cells expresses two T cell receptor (TCR) clonotypes, though the extent and functional significance of this is not established. To definitively evaluate dual TCRα cells, we generated mice with green fluorescent protein and red fluorescent protein reporters linked to TCRα, revealing that ∼16% of T cells express dual TCRs, notably higher than prior estimates. Importantly, dual TCR expression has functional consequences, as dual TCR cells predominated response to lymphocytic choriomeningitis virus infection, comprising up to 60% of virus-specific CD4+ and CD8+ T cells during acute responses. Dual receptor expression selectively influenced immune memory, as postinfection memory CD4+ populations contained significantly increased frequencies of dual TCR cells. These data reveal a previously unappreciated contribution of dual TCR cells to the immune repertoire and highlight their potential effects on immune responses.
Collapse
|
5
|
Arrieta-Bolaños E, Fleischhauer K. Learning the next-generation sequencing alphabet of immune reconstitution: factors determining CD8 + T-cell receptor α-chain repertoire dynamics after hematopoietic stem cell transplantation. Haematologica 2019; 104:422-425. [PMID: 30819834 DOI: 10.3324/haematol.2018.209130] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Affiliation(s)
| | - Katharina Fleischhauer
- Institute for Experimental Cellular Therapy, University Hospital Essen.,German Cancer Consortium, Heidelberg, Germany
| |
Collapse
|
6
|
Carter JA, Preall JB, Atwal GS. Bayesian Inference of Allelic Inclusion Rates in the Human T Cell Receptor Repertoire. Cell Syst 2019; 9:475-482.e4. [PMID: 31677971 DOI: 10.1016/j.cels.2019.09.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 08/04/2019] [Accepted: 09/17/2019] [Indexed: 01/09/2023]
Abstract
A small population of αβ T cells is characterized by the expression of more than one unique T cell receptor (TCR); this outcome is the result of "allelic inclusion," that is, inclusion of both α- or β-chain alleles during V(D)J recombination. Limitations in single-cell sequencing technology, however, have precluded comprehensive enumeration of these dual receptor T cells. Here, we develop and experimentally validate a fully Bayesian inference model capable of reliably estimating the true rate of α and β TCR allelic inclusion across two different emulsion-barcoding single-cell sequencing platforms. We provide a database composed of over 51,000 previously unpublished allelic inclusion TCR sequence sets drawn from eight healthy individuals and show that allelic inclusion contributes a distinct and functionally important set of sequences to the human TCR repertoire. This database and a Python implementation of our statistical inference model are freely available at our Github repository (https://github.com/JasonACarter/Allelic_inclusion).
Collapse
Affiliation(s)
- Jason A Carter
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, NY 11794, USA; Cold Spring Harbor Laboratory, Cold Spring Harbor, Stony Brook, NY 11724, USA.
| | - Jonathan B Preall
- Cold Spring Harbor Laboratory, Cold Spring Harbor, Stony Brook, NY 11724, USA
| | - Gurinder S Atwal
- Cold Spring Harbor Laboratory, Cold Spring Harbor, Stony Brook, NY 11724, USA.
| |
Collapse
|
7
|
Schuldt NJ, Binstadt BA. Dual TCR T Cells: Identity Crisis or Multitaskers? JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2019; 202:637-644. [PMID: 30670579 PMCID: PMC11112972 DOI: 10.4049/jimmunol.1800904] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 08/21/2018] [Indexed: 05/25/2024]
Abstract
Dual TCR T cells are a common and natural product of TCR gene rearrangement and thymocyte development. As much as one third of the T cell population may have the capability to express two different TCR specificities on the cell surface. This discovery provoked a reconsideration of the classic model of thymic selection. Many potential roles for dual TCR T cells have since been hypothesized, including posing an autoimmune hazard, dominating alloreactive T cell responses, inducing allergy, and expanding the TCR repertoire to improve protective immunity. Yet, since the initial wave of publications following the discovery of dual TCR T cells, research in the area has slowed. In this study, we aim to provide a brief but comprehensive history of dual TCR T cell research, re-evaluate past observations in the context of current knowledge of the immune system, and identify key issues for future study.
Collapse
Affiliation(s)
- Nathaniel J Schuldt
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55454; and Center for Immunology, University of Minnesota, Minneapolis, MN 55455
| | - Bryce A Binstadt
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55454; and Center for Immunology, University of Minnesota, Minneapolis, MN 55455
| |
Collapse
|
8
|
Bystander T Cells: A Balancing Act of Friends and Foes. Trends Immunol 2018; 39:1021-1035. [PMID: 30413351 DOI: 10.1016/j.it.2018.10.003] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 09/27/2018] [Accepted: 10/04/2018] [Indexed: 02/07/2023]
Abstract
T cell responses are essential for appropriate protection against pathogens. T cell immunity is achieved through the ability to discriminate between foreign and self-molecules, and this relies heavily on stringent T cell receptor (TCR) specificity. Recently, bystander activated T lymphocytes, that are specific for unrelated epitopes during an antigen-specific response, have been implicated in diverse diseases. Numerous infection models have challenged the classic dogma of T cell activation as being solely dependent on TCR and major histocompatibility complex (MHC) interactions, indicating an unappreciated role for pathogen-associated receptors on T cells. We discuss here the specific roles of bystander activated T cells in pathogenesis, shedding light on the ability of these cells to modulate disease severity independently from TCR recognition.
Collapse
|
9
|
Pando A, Reagan JL, Nevola M, Fast LD. Induction of anti-leukemic responses by stimulation of leukemic CD3+ cells with allogeneic stimulator cells. Exp Hematol Oncol 2018; 7:25. [PMID: 30323982 PMCID: PMC6172765 DOI: 10.1186/s40164-018-0118-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 09/30/2018] [Indexed: 01/13/2023] Open
Abstract
Background Immunotherapeutic protocols have focused on identification of stimuli that induce effective anti-leukemic immune responses. One potent immune stimulus is the encounter with allogeneic cells. Our group previously showed that the infusion of haploidentical donor white blood cells (1-2 × 108 CD3+ cells/kg) into patients with refractory hematological malignancies induced responses of varying magnitude in over half of the patients. Because donor cells were eliminated within 2 weeks in these patients, it is presumed that the responses of recipient lymphocytes were critically important in achieving prolonged anti-leukemic responses. Methods The role of patient CD3+ cells in anti-leukemic responses was examined by isolating peripheral blood mononuclear cells from newly diagnosed leukemic patients. Immunophenotyping was performed on these peripheral blood mononuclear cells. CD3+ cells were isolated from the peripheral blood mononuclear cells and tested for their ability to proliferate and lyse autologous leukemic cells when stimulated with unrelated allogeneic cells. Results Allostimulated CD3+ cells effectively generated cytolytic responses to autologous CD3-cells in 11/21 patients. Increased numbers of CD4+ cells expressing high levels of granzyme A, B and perforin and CD8+CD39+ cells were found in nonresponsive CD3+ cells. Conclusions These results indicate that CD3+ cells from leukemic patients are capable of generating anti-leukemic responses when stimulated with unrelated allogeneic cells. This model can be used to identify approaches using alloreactive responses by patient lymphocytes to enhance in vivo anti-leukemic responses.
Collapse
Affiliation(s)
- Alejandro Pando
- Division of Hematology/Oncology, Rhode Island Hospital and the Warren Alpert School of Medicine at Brown University, One Hoppin Street, Coro West Suite 5.0.1, Providence, RI 02903 USA
| | - John L Reagan
- Division of Hematology/Oncology, Rhode Island Hospital and the Warren Alpert School of Medicine at Brown University, One Hoppin Street, Coro West Suite 5.0.1, Providence, RI 02903 USA
| | - Martha Nevola
- Division of Hematology/Oncology, Rhode Island Hospital and the Warren Alpert School of Medicine at Brown University, One Hoppin Street, Coro West Suite 5.0.1, Providence, RI 02903 USA
| | - Loren D Fast
- Division of Hematology/Oncology, Rhode Island Hospital and the Warren Alpert School of Medicine at Brown University, One Hoppin Street, Coro West Suite 5.0.1, Providence, RI 02903 USA
| |
Collapse
|
10
|
Balakrishnan A, Jama B, Morris GP. Endogenous co‐expression of two T cell receptors promotes lymphopenia‐induced proliferation via increased affinity for self‐antigen. J Leukoc Biol 2018; 104:1097-1104. [DOI: 10.1002/jlb.1ab0618-214rrr] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 08/08/2018] [Accepted: 08/10/2018] [Indexed: 11/11/2022] Open
Affiliation(s)
- Amritha Balakrishnan
- Department of PathologyUniversity of California San Diego La Jolla California USA
| | - Burhan Jama
- Department of PathologyUniversity of California San Diego La Jolla California USA
| | - Gerald P. Morris
- Department of PathologyUniversity of California San Diego La Jolla California USA
| |
Collapse
|
11
|
MHC-mismatched mixed chimerism restores peripheral tolerance of noncross-reactive autoreactive T cells in NOD mice. Proc Natl Acad Sci U S A 2018; 115:E2329-E2337. [PMID: 29463744 PMCID: PMC5877958 DOI: 10.1073/pnas.1720169115] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Mixed chimerism has shown good potential to cure some autoimmune diseases and prevent tissue rejection. It is known that MHC-mismatched but not -matched mixed chimerism effectively tolerizes autoreactive T cells, even those noncross-reactive T cells that do not directly recognize donor-type antigen presenting cells [i.e., dendritic cells (DCs)]. How this is accomplished remains unknown. These studies have shown that tolerizing peripheral residual host-type noncross-reactive autoreactive T cells requires engraftment of donor-type DCs and involves a host-type DC-mediated increase in donor-type Treg cells, which associates with restoration of tolerogenic features of host-type plasmacytoid DCs and expansion of host-type Treg cells. This study suggests a previously unrecognized tolerance network among donor- and host-type DCs and Treg cells in MHC-mismatched mixed chimeras. Autoimmune type 1 diabetes (T1D) and other autoimmune diseases are associated with particular MHC haplotypes and expansion of autoreactive T cells. Induction of MHC-mismatched but not -matched mixed chimerism by hematopoietic cell transplantation effectively reverses autoimmunity in diabetic nonobese diabetic (NOD) mice, even those with established diabetes. As expected, MHC-mismatched mixed chimerism mediates deletion in the thymus of host-type autoreactive T cells that have T-cell receptor (TCR) recognizing (cross-reacting with) donor-type antigen presenting cells (APCs), which have come to reside in the thymus. However, how MHC-mismatched mixed chimerism tolerizes host autoreactive T cells that recognize only self-MHC–peptide complexes remains unknown. Here, using NOD.Rag1−/−.BDC2.5 or NOD.Rag1−/−.BDC12-4.1 mice that have only noncross-reactive transgenic autoreactive T cells, we show that induction of MHC-mismatched but not -matched mixed chimerism restores immune tolerance of peripheral noncross-reactive autoreactive T cells. MHC-mismatched mixed chimerism results in increased percentages of both donor- and host-type Foxp3+ Treg cells and up-regulated expression of programmed death-ligand 1 (PD-L1) by host-type plasmacytoid dendritic cells (pDCs). Furthermore, adoptive transfer experiments showed that engraftment of donor-type dendritic cells (DCs) and expansion of donor-type Treg cells are required for tolerizing the noncross-reactive autoreactive T cells in the periphery, which are in association with up-regulation of host-type DC expression of PD-L1 and increased percentage of host-type Treg cells. Thus, induction of MHC-mismatched mixed chimerism may establish a peripheral tolerogenic DC and Treg network that actively tolerizes autoreactive T cells, even those with no TCR recognition of the donor APCs.
Collapse
|
12
|
Gong Q, Wang C, Zhang W, Iqbal J, Hu Y, Greiner TC, Cornish A, Kim JH, Rabadan R, Abate F, Wang X, Inghirami GG, McKeithan TW, Chan WC. Assessment of T-cell receptor repertoire and clonal expansion in peripheral T-cell lymphoma using RNA-seq data. Sci Rep 2017; 7:11301. [PMID: 28900149 PMCID: PMC5595876 DOI: 10.1038/s41598-017-11310-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 08/22/2017] [Indexed: 12/11/2022] Open
Abstract
T-cell clonality of peripheral T-cell lymphoma (PTCL) is routinely evaluated with a PCR-based method using genomic DNA. However, there are limitations with this approach. The purpose of this study was to determine the utility of RNA-seq for assessing T-cell clonality and T-cell antigen receptor (TCR) repertoire of the neoplastic T-cells in 108 PTCL samples. TCR transcripts, including complementarity-determining region 3 (CDR3) sequences, were assessed. In normal T cells, the CDR3 sequences were extremely diverse, without any clonotype representing more than 2% of the overall TCR population. Dominant clones could be identified in 65 out of 76 PTCL cases (86%) with adequate TCR transcript expression. In monoclonal cases, the dominant clone varied between 11% and 99% of TCRβ transcripts. No unique Vα or Vβ usage was observed. Small T-cell clones were often observed in T- and NK-cell tumors in a percentage higher than observed in reactive conditions. γ chain expression was very low in tumors expressing TCRαβ, but its expression level was high and clonality was detected in a TCRγδ expressing tumor. NK cell lymphoma (NKCL) did not express significant levels of TCR Vβ or Vγ genes. RNA-seq is a useful tool for detecting and characterizing clonal TCR rearrangements in PTCL.
Collapse
Affiliation(s)
- Qiang Gong
- Department of Pathology, City of Hope National Medical Center, Duarte, 91010, CA, United States
| | - Chao Wang
- Department of Pathology, City of Hope National Medical Center, Duarte, 91010, CA, United States.,Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, United States.,Department of Hematology, Shandong Provincial Hospital affiliated to Shandong University, Jinan, P.R. China
| | - Weiwei Zhang
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Javeed Iqbal
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Yang Hu
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Timothy C Greiner
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Adam Cornish
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Jo-Heon Kim
- Department of Pathology, City of Hope National Medical Center, Duarte, 91010, CA, United States.,Department of Pathology, Chonnam National University Medical School and Research Institute of Medical Sciences, Gwangju, South Korea
| | - Raul Rabadan
- Department of Biomedical Informatics, Columbia University, New York, NY, United States
| | - Francesco Abate
- Department of Biomedical Informatics, Columbia University, New York, NY, United States
| | - Xin Wang
- Department of Hematology, Shandong Provincial Hospital affiliated to Shandong University, Jinan, P.R. China
| | - Giorgio G Inghirami
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY, United States
| | - Timothy W McKeithan
- Department of Pathology, City of Hope National Medical Center, Duarte, 91010, CA, United States
| | - Wing C Chan
- Department of Pathology, City of Hope National Medical Center, Duarte, 91010, CA, United States.
| |
Collapse
|
13
|
Proinflammatory Dual Receptor T Cells in Chronic Graft-versus-Host Disease. Biol Blood Marrow Transplant 2017; 23:1852-1860. [PMID: 28750779 DOI: 10.1016/j.bbmt.2017.07.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 07/16/2017] [Indexed: 12/11/2022]
Abstract
Defective post-transplantation thymopoiesis is associated with chronic graft-versus-host disease (GVHD), a multiorgan pathology affecting up to 80% of patients after allogeneic hematopoietic stem cell transplantation (HSCT). Previous work demonstrated that the subset of T cells expressing 2 T cell receptors (TCRs) is predisposed to alloreactivity, driving selective and disproportionate activity in acute GVHD in both mouse models and HSCT patients. Here we investigate a potential role for this pathogenic T cell subset in chronic GVHD (cGVHD). HSCT patients with cGVHD demonstrated increased numbers of dual TCR cells in circulation. These dual receptor cells had an activated phenotype, indicating an active role in cGVHD. Notably, single-cell RNA sequencing identified the increased dual TCR cells in cGVHD as predominantly expressing Tbet, indicative of a proinflammatory phenotype. These results identify dual TCR cells as specific mediators of pathogenic inflammation underlying cGVHD and highlight Tbet-driven T cell function as a potential pathway for potential therapeutic targeting.
Collapse
|
14
|
Abstract
Alloimmune T cells are central mediators of rejection and graft-versus-host disease in both solid organ and hematopoietic stem cell transplantation. Unique among immune responses in terms of its strength and diversity, the T cell alloresponse reflects extensive genetic polymorphisms between allogeneic donors and recipients, most prominently within the major histocompatibility complex (MHC), which encodes human leukocyte antigens (HLAs) in humans. The repertoire of alloreactive T cell clones is distinct for every donor-recipient pair and includes potentially thousands of unique HLA/peptide specificities. The extraordinary magnitude of the primary alloresponse and diversity of the T cell population mediating it have presented technical challenges to its study in humans. High-throughput T cell receptor sequencing approaches have opened up new possibilities for tackling many fundamental questions about this important immunologic phenomenon.
Collapse
|
15
|
Abstract
Alloreactive T lymphocytes are the primary mediators of allograft rejection. The size and diversity of the HLA-alloreactive T cell repertoire has thus far precluded the ability to follow these T cells and thereby to understand their fate in human transplant recipients. This review summarizes the history, challenges, and recent advances in the study of alloreactive T cells. We highlight the historical development of assays to measure alloreactivity and discuss how high-throughput T cell receptor (TCR) sequencing-based assays can provide a new window into the fate of alloreactive T cells in human transplant recipients. A specific approach combining a classical in vitro assay, the mixed lymphocyte reaction, with deep T cell receptor sequencing is described as a tool to track the donor-reactive T cell repertoire for any specific HLA-mismatched donor-recipient pair. This assay can provide mechanistic insights and has potential as a noninvasive, highly specific biomarker for rejection and tolerance.
Collapse
|
16
|
The CD8 T-cell response during tolerance induction in liver transplantation. Clin Transl Immunology 2016; 5:e102. [PMID: 27867515 PMCID: PMC5099425 DOI: 10.1038/cti.2016.53] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 07/20/2016] [Accepted: 07/22/2016] [Indexed: 12/12/2022] Open
Abstract
Both experimental and clinical studies have shown that the liver possesses unique tolerogenic properties. Liver allografts can be spontaneously accepted across complete major histocompatibility mismatch in some animal models. In addition, some liver transplant patients can be successfully withdrawn from immunosuppressive medications, developing ‘operational tolerance'. Multiple mechanisms have been shown to be involved in inducing and maintaining alloimmune tolerance associated with liver transplantation. Here, we focus on CD8 T-cell tolerance in this setting. We first discuss how alloreactive cytotoxic T-cell responses are generated against allografts, before reviewing how the liver parenchyma, donor passenger leucocytes and the host immune system function together to attenuate alloreactive CD8 T-cell responses to promote the long-term survival of liver transplants.
Collapse
|
17
|
Abstract
PURPOSE OF REVIEW T cells can mediate allograft rejection and graft-versus-host disease (GVHD), but are necessary for tolerance and protective immunity. Identifying T-cell populations differentially responsible for these effects has been a goal in transplant research. This review describes investigation of a small subset of T cells naturally predisposed toward alloreactivity, cells expressing two T-cell receptors (TCRs). RECENT FINDINGS Rare peripheral T cells express two αβTCRs. Their impact on T-cell development and function has been uncertain. Recent work demonstrates an important role for these cells in mouse models and human hematopoietic stem cell transplant patients with acute GVHD. Dual receptor T cells are preferentially activated and expanded in vitro and in vivo by allogeneic stimulation. Genetic elimination of dual TCR expression results in loss of approximately half of the alloreactive repertoire and impedes the earliest steps of GVHD. SUMMARY Identification of dual TCR T cells as predisposed to alloreactivity provides an opportunity to examine responses limiting transplantation. Continued investigation will reveal significant fundamental features of T-cell alloreactivity and important information about the earliest events determining allograft rejection and self-tolerance.
Collapse
|
18
|
Joachims ML, Leehan KM, Lawrence C, Pelikan RC, Moore JS, Pan Z, Rasmussen A, Radfar L, Lewis DM, Grundahl KM, Kelly JA, Wiley GB, Shugay M, Chudakov DM, Lessard CJ, Stone DU, Scofield RH, Montgomery CG, Sivils KL, Thompson LF, Farris AD. Single-cell analysis of glandular T cell receptors in Sjögren's syndrome. JCI Insight 2016; 1. [PMID: 27358913 DOI: 10.1172/jci.insight.85609] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
CD4+ T cells predominate in salivary gland (SG) inflammatory lesions in Sjögren's syndrome (SS). However, their antigen specificity, degree of clonal expansion, and relationship to clinical disease features remain unknown. We used multiplex reverse-transcriptase PCR to amplify paired T cell receptor α (TCRα) and β transcripts of single CD4+CD45RA- T cells from SG and peripheral blood (PB) of 10 individuals with primary SS, 9 of whom shared the HLA DR3/DQ2 risk haplotype. TCRα and β sequences were obtained from a median of 91 SG and 107 PB cells per subject. The degree of clonal expansion and frequency of cells expressing two productively rearranged α genes were increased in SG versus PB. Expanded clones from SG exhibited complementary-determining region 3 (CDR3) sequence similarity both within and among subjects, suggesting antigenic selection and shared antigen recognition. CDR3 similarities were shared among expanded clones from individuals discordant for canonical Ro and La autoantibodies, suggesting recognition of alternative SG antigen(s). The extent of SG clonal expansion correlated with reduced saliva production and increased SG fibrosis, linking expanded SG T cells with glandular dysfunction. Knowledge of paired TCRα and β sequences enables further work toward identification of target antigens and development of novel therapies.
Collapse
Affiliation(s)
- Michelle L Joachims
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, Oklahoma, USA
| | - Kerry M Leehan
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, Oklahoma, USA; Department of Pathology, University of Oklahoma Health Sciences Center (OUHSC), Oklahoma City, Oklahoma, USA
| | - Christina Lawrence
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, Oklahoma, USA
| | - Richard C Pelikan
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, Oklahoma, USA
| | - Jacen S Moore
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, Oklahoma, USA
| | - Zijian Pan
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, Oklahoma, USA
| | - Astrid Rasmussen
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, Oklahoma, USA
| | - Lida Radfar
- Department of Oral Diagnosis and Radiology, College of Dentistry, OUHSC, Oklahoma City, Oklahoma, USA
| | - David M Lewis
- Department of Oral and Maxillofacial Pathology, College of Dentistry, OUHSC, Oklahoma City, Oklahoma, USA
| | - Kiely M Grundahl
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, Oklahoma, USA
| | - Jennifer A Kelly
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, Oklahoma, USA
| | - Graham B Wiley
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, Oklahoma, USA
| | - Mikhail Shugay
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia; Pirogov Russian National Research Medical University, Moscow, Russia; Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Dmitriy M Chudakov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia; Pirogov Russian National Research Medical University, Moscow, Russia; Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Christopher J Lessard
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, Oklahoma, USA; Department of Pathology, University of Oklahoma Health Sciences Center (OUHSC), Oklahoma City, Oklahoma, USA
| | - Donald U Stone
- Department of Ophthalmology, College of Medicine, OUHSC, Oklahoma City, Oklahoma, USA
| | - R Hal Scofield
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, Oklahoma, USA; Department of Pathology, University of Oklahoma Health Sciences Center (OUHSC), Oklahoma City, Oklahoma, USA; Section of Endocrinology and Diabetes, College of Medicine, OUHSC, Oklahoma City, Oklahoma, USA
| | - Courtney G Montgomery
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, Oklahoma, USA
| | - Kathy L Sivils
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, Oklahoma, USA; Department of Pathology, University of Oklahoma Health Sciences Center (OUHSC), Oklahoma City, Oklahoma, USA
| | - Linda F Thompson
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, Oklahoma, USA
| | - A Darise Farris
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, Oklahoma, USA; Department of Pathology, University of Oklahoma Health Sciences Center (OUHSC), Oklahoma City, Oklahoma, USA
| |
Collapse
|
19
|
Krummey SM, Martinez RJ, Andargachew R, Liu D, Wagener M, Kohlmeier JE, Evavold BD, Larsen CP, Ford ML. Low-Affinity Memory CD8+ T Cells Mediate Robust Heterologous Immunity. THE JOURNAL OF IMMUNOLOGY 2016; 196:2838-46. [PMID: 26864034 DOI: 10.4049/jimmunol.1500639] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 01/12/2016] [Indexed: 12/30/2022]
Abstract
Heterologous immunity is recognized as a significant barrier to transplant tolerance. Whereas it has been established that pathogen-elicited memory T cells can have high or low affinity for cross-reactive allogeneic peptide-MHC, the role of TCR affinity during heterologous immunity has not been explored. We established a model with which to investigate the impact of TCR-priming affinity on memory T cell populations following a graft rechallenge. In contrast to high-affinity priming, low-affinity priming elicited fully differentiated memory T cells with a CD45RB(hi) status. High CD45RB status enabled robust secondary responses in vivo, as demonstrated by faster graft rejection kinetics and greater proliferative responses. CD45RB blockade prolonged graft survival in low affinity-primed mice, but not in high affinity-primed mice. Mechanistically, low affinity-primed memory CD8(+) T cells produced more IL-2 and significantly upregulated IL-2Rα expression during rechallenge. We found that CD45RB(hi) status was also a stable marker of priming affinity within polyclonal CD8(+) T cell populations. Following high-affinity rechallenge, low affinity-primed CD45RB(hi) cells became CD45RB(lo), demonstrating that CD45RB status acts as an affinity-based differentiation switch on CD8(+) T cells. Thus, these data establish a novel mechanism by which CD45 isoforms tune low affinity-primed memory CD8(+) T cells to become potent secondary effectors following heterologous rechallenge. These findings have direct implications for allogeneic heterologous immunity by demonstrating that despite a lower precursor frequency, low-affinity priming is sufficient to generate memory cells that mediate potent secondary responses against a cross-reactive graft challenge.
Collapse
Affiliation(s)
| | - Ryan J Martinez
- Department of Microbiology and Immunology, Emory University, Atlanta, GA 30322
| | - Rakieb Andargachew
- Department of Microbiology and Immunology, Emory University, Atlanta, GA 30322
| | - Danya Liu
- Emory Transplant Center, Atlanta, GA 30322; and
| | | | - Jacob E Kohlmeier
- Department of Microbiology and Immunology, Emory University, Atlanta, GA 30322
| | - Brian D Evavold
- Department of Microbiology and Immunology, Emory University, Atlanta, GA 30322
| | | | | |
Collapse
|
20
|
MHC-mismatched mixed chimerism augments thymic regulatory T-cell production and prevents relapse of EAE in mice. Proc Natl Acad Sci U S A 2015; 112:15994-9. [PMID: 26647186 DOI: 10.1073/pnas.1521157112] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Multiple sclerosis (MS) is an autoimmune inflammatory disease of the central nervous system with demyelination, axon damage, and paralysis. Induction of mixed chimerism with allogeneic donors has been shown to not cause graft-versus-host disease (GVHD) in animal models and humans. We have reported that induction of MHC-mismatched mixed chimerism can cure autoimmunity in autoimmune NOD mice, but this approach has not yet been tested in animal models of MS, such as experimental autoimmune encephalomyelitis (EAE). Here, we report that MHC-mismatched mixed chimerism with C57BL/6 (H-2(b)) donor in SJL/J (H-2(s)) EAE recipients eliminates clinical symptoms and prevents relapse. This cure is demonstrated by not only disappearance of clinical signs but also reversal of autoimmunity; elimination of infiltrating T, B, and macrophage cells in the spinal cord; and regeneration of myelin sheath. The reversal of autoimmunity is associated with a marked reduction of autoreactivity of CD4(+) T cells and significant increase in the percentage of Foxp3(+) Treg among host-type CD4(+) T cells in the spleen and lymph nodes. The latter is associated with a marked reduction of the percentage of host-type CD4(+)CD8(+) thymocytes and an increase of Treg percentage among the CD4(+)CD8(+) and CD4(+)CD8(-) thymocytes. Thymectomy leads to loss of prevention of EAE relapse by induction of mixed chimerism, although there is a dramatic expansion of host-type Treg cells in the lymph nodes. These results indicate that induction of MHC-mismatched mixed chimerism can restore thymic negative selection of autoreactive CD4(+) T cells, augment production of Foxp3(+) Treg, and cure EAE.
Collapse
|
21
|
Zhang L, Chu J, Yu J, Wei W. Cellular and molecular mechanisms in graft-versus-host disease. J Leukoc Biol 2015; 99:279-87. [PMID: 26643713 DOI: 10.1189/jlb.4ru0615-254rr] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 10/19/2015] [Indexed: 12/18/2022] Open
Abstract
Graft-versus-host disease is a complication in patients undergoing hematopoietic stem cell transplantation. Graft-versus-host disease includes acute graft-versus-host disease and chronic graft-versus-host disease. Host APCs (e.g., dendritic cells and macrophages), effector T cells (e.g., Th1, Th17, and abnormal Th17:regulatory T cell ratio), B cells, and NK cells are implicated in graft-versus-host disease physiopathology. Proinflammation cytokines (e.g., IL-17, IL-1β, and TNF-α) are increased in graft-versus-host disease . Costimulatory molecules play an important role in inducing graft-versus-host disease . Pattern-recognition receptors, such as TLRs and nucleotide-binding oligomerization domain-like receptors, are critically involved in the pathogenesis of graft-versus-host disease . Complement system C3 mediates Th1/Th17 polarization in human T cell activation and skin graft-versus-host disease. Accumulation of CD26 T cells in graft-versus-host disease target organs was found. As a therapeutic target, soluble CD83 molecules or antibodies have been demonstrated to have therapeutic effects against graft-versus-host disease, and signaling molecules promote the inflammatory and immune process of graft-versus-host disease . These immune cells and molecules could be the predictors of graft-versus-host disease development and the drug targets of the treatments for graft-versus-host disease. This article focuses on major advances on cellular and molecular mechanisms in graft-versus-host disease.
Collapse
Affiliation(s)
- Lingling Zhang
- *Institute of Clinical Pharmacology and Key Laboratory of Antiinflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei, China; and Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Jianhong Chu
- *Institute of Clinical Pharmacology and Key Laboratory of Antiinflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei, China; and Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Jianhua Yu
- *Institute of Clinical Pharmacology and Key Laboratory of Antiinflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei, China; and Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Wei Wei
- *Institute of Clinical Pharmacology and Key Laboratory of Antiinflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei, China; and Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
22
|
McCarthy DP, Bryant J, Galvin JP, Miller SD, Luo X. Tempering allorecognition to induce transplant tolerance with chemically modified apoptotic donor cells. Am J Transplant 2015; 15:1475-83. [PMID: 25807873 PMCID: PMC4439351 DOI: 10.1111/ajt.13237] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Revised: 01/12/2015] [Accepted: 01/23/2015] [Indexed: 01/25/2023]
Abstract
The development of organ transplantation as a therapy for end-stage organ failure is among the most significant achievements of 20th century medicine, but chronic rejection remains a barrier to achieving long-term success. Current therapeutic regimens consist of immunosuppressive drugs that are efficient at delaying rejection but are associated with significant risks such as opportunistic infections, toxicity, and malignancy. Thus, the induction of specific immune tolerance to transplant antigens is the coveted aim of researchers. The use of 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide (ECDI)-treated, autoantigen-coupled syngeneic leukocytes has been developed as a specific immunotherapy in preclinical models of autoimmunity and is currently in a phase II clinical trial for the treatment of multiple sclerosis. In this review, we discuss the use of allogeneic ECDI-treated apoptotic donor leukocytes (allo-ECDI-SP) as a strategy for inducing antigen-specific tolerance in allogeneic transplantation. Allo-ECDI-SP therapy induces long-term systemic immune tolerance to transplant antigens by subverting alloimmune recognition and exploiting apoptotic cell uptake pathways to recapitulate innate mechanisms of peripheral tolerance. Lastly, we discuss potential indications and challenges for transitioning allo-ECDI-SP therapy into clinical practice.
Collapse
Affiliation(s)
- D. P. McCarthy
- Department of Microbiology-Immunology and Interdepartmental Immunobiology Center, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - J. Bryant
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Evanston, IL
| | - J. P. Galvin
- Department of Microbiology-Immunology and Interdepartmental Immunobiology Center, Feinberg School of Medicine, Northwestern University, Chicago, IL
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Evanston, IL
| | - S. D. Miller
- Department of Microbiology-Immunology and Interdepartmental Immunobiology Center, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - X. Luo
- Department of Microbiology-Immunology and Interdepartmental Immunobiology Center, Feinberg School of Medicine, Northwestern University, Chicago, IL
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Evanston, IL
- Comprehensive Transplant Center, Feinberg School of Medicine, Northwestern University, Evanston, IL
| |
Collapse
|
23
|
Fast LD. Harnessing alloreactivity to achieve anti-leukemic responses. Leuk Suppl 2014; 3:S11. [PMID: 27175261 DOI: 10.1038/leusup.2014.6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
- L D Fast
- Division of Hematology/Oncology, Rhode Island Hospital and Warren Alpert School of Medicine at Brown University, Providence, RI , USA
| |
Collapse
|
24
|
Racine JJ, Zhang M, Wang M, Morales W, Shen C, Zeng D. MHC-mismatched mixed chimerism mediates thymic deletion of cross-reactive autoreactive T cells and prevents insulitis in nonobese diabetic mice. THE JOURNAL OF IMMUNOLOGY 2014; 194:407-17. [PMID: 25429069 DOI: 10.4049/jimmunol.1401584] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Type 1 diabetic NOD mice have defects in both thymic negative selection and peripheral regulation of autoreactive T cells, and induction of mixed chimerism can effectively reverse these defects. Our recent studies suggest that MHC-mismatched mixed chimerism mediates negative selection of autoreactive thymocytes in wild-type NOD and TCR-transgenic NOD.Rag1(+/+).BDC2.5 mice. However, it remains unknown how mismatched I-A(b) MHC class II can mediate deletion of autoreactive T cells positively selected by I-A(g7). In the present study, we directly tested the hypothesis that mismatched MHC class II in mixed chimeras mediates deletion of cross-reactive autoreactive thymocytes. We first identify that transgenic BDC2.5 T cells from NOD.Rag1(+/+).BDC2.5 but not NOD.Rag1(-/-).BDC2.5 mice possess cross-reactive TCRs with endogenous TCRα-chains; MHC-mismatched H-2(b) but not matched H-2(g7) mixed chimerism mediates thymic deletion of the cross-reactive transgenic T cells in NOD.Rag1(+/+).BDC2.5 mice. Second, by transplanting T cell-depleted (TCD) bone marrow (BM) cells from NOD.Rag1(+/+).BDC2.5 or NOD.Rag1(-/-).BDC2.5 mice into lethally irradiated MHC-mismatched H-2(b) C57BL/6 or MHC-matched congenic B6.H-2(g7) recipients, we demonstrate that NOD.Rag1(+/+).BDC2.5 BM-derived cross-reactive transgenic T cells, but not NOD.Rag1(-/-).BDC2.5 BM-derived non-cross-reactive transgenic T cells, can be positively selected in MHC-mismatched H-2(b) thymus. Third, by cotransplanting NOD.Rag1(+/+).BDC2.5 TCD BM cells with BM cells from MHC-mismatched T cell-deficient C57BL/6 mice into lethally irradiated MHC-matched B6.H-2(g7) recipients, we demonstrate that thymic deletion of the cross-reactive transgenic T cells is dependent on MHC-mismatched donor BM-derived APCs but not on donor BM-derived T cells. Taken together, our studies indicate that MHC-mismatched mixed chimerism can mediate thymic deletion of cross-reactive autoreactive T cells that express more than one TCR.
Collapse
Affiliation(s)
- Jeremy J Racine
- Irell and Manella Graduate School of Biological Sciences, City of Hope, Duarte, CA 91010; Department of Diabetes Research, Beckman Research Institute, City of Hope, Duarte, CA 91010; Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope, Duarte, CA 91010; and
| | - Mingfeng Zhang
- Department of Diabetes Research, Beckman Research Institute, City of Hope, Duarte, CA 91010; Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope, Duarte, CA 91010; and
| | - Miao Wang
- Department of Diabetes Research, Beckman Research Institute, City of Hope, Duarte, CA 91010; Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope, Duarte, CA 91010; and
| | - William Morales
- Eugene and Ruth Roberts Summer Student Academy, Beckman Research Institute, City of Hope, Duarte, CA 91010
| | - Christine Shen
- Eugene and Ruth Roberts Summer Student Academy, Beckman Research Institute, City of Hope, Duarte, CA 91010
| | - Defu Zeng
- Irell and Manella Graduate School of Biological Sciences, City of Hope, Duarte, CA 91010; Department of Diabetes Research, Beckman Research Institute, City of Hope, Duarte, CA 91010; Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope, Duarte, CA 91010; and
| |
Collapse
|
25
|
Jama BP, Morris GP. Generation of human alloantigen-specific T cells from peripheral blood. J Vis Exp 2014:e52257. [PMID: 25490283 DOI: 10.3791/52257] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The study of human T lymphocyte biology often involves examination of responses to activating ligands. T cells recognize and respond to processed peptide antigens presented by MHC (human ortholog HLA) molecules through the T cell receptor (TCR) in a highly sensitive and specific manner. While the primary function of T cells is to mediate protective immune responses to foreign antigens presented by self-MHC, T cells respond robustly to antigenic differences in allogeneic tissues. T cell responses to alloantigens can be described as either direct or indirect alloreactivity. In alloreactivity, the T cell responds through highly specific recognition of both the presented peptide and the MHC molecule. The robust oligoclonal response of T cells to allogeneic stimulation reflects the large number of potentially stimulatory alloantigens present in allogeneic tissues. While the breadth of alloreactive T cell responses is an important factor in initiating and mediating the pathology associated with biologically-relevant alloreactive responses such as graft versus host disease and allograft rejection, it can preclude analysis of T cell responses to allogeneic ligands. To this end, this protocol describes a method for generating alloreactive T cells from naive human peripheral blood leukocytes (PBL) that respond to known peptide-MHC (pMHC) alloantigens. The protocol applies pMHC multimer labeling, magnetic bead enrichment and flow cytometry to single cell in vitro culture methods for the generation of alloantigen-specific T cell clones. This enables studies of the biochemistry and function of T cells responding to allogeneic stimulation.
Collapse
Affiliation(s)
- Burhan P Jama
- Department of Pathology, University of California, San Diego
| | - Gerald P Morris
- Department of Pathology, University of California, San Diego;
| |
Collapse
|
26
|
Ni PP, Solomon B, Hsieh CS, Allen PM, Morris GP. The ability to rearrange dual TCRs enhances positive selection, leading to increased Allo- and Autoreactive T cell repertoires. THE JOURNAL OF IMMUNOLOGY 2014; 193:1778-86. [PMID: 25015825 DOI: 10.4049/jimmunol.1400532] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Thymic selection is designed to ensure TCR reactivity to foreign Ags presented by self-MHC while minimizing reactivity to self-Ags. We hypothesized that the repertoire of T cells with unwanted specificities such as alloreactivity or autoreactivity are a consequence of simultaneous rearrangement of both TCRα loci. We hypothesized that this process helps maximize production of thymocytes capable of successfully completing thymic selection, but results in secondary TCRs that escape stringent selection. In T cells expressing two TCRs, one TCR can mediate positive selection and mask secondary TCR from negative selection. Examination of mice heterozygous for TRAC (TCRα(+/-)), capable of only one functional TCRα rearrangement, demonstrated a defect in generating mature T cells attributable to decreased positive selection. Elimination of secondary TCRs did not broadly alter the peripheral T cell compartment, though deep sequencing of TCRα repertoires of dual TCR T cells and TCRα(+/-) T cells demonstrated unique TCRs in the presence of secondary rearrangements. The functional impact of secondary TCRs on the naive peripheral repertoire was evidenced by reduced frequencies of T cells responding to autoantigen and alloantigen peptide-MHC tetramers in TCRα(+/-) mice. T cell populations with secondary TCRs had significantly increased ability to respond to altered peptide ligands related to their allogeneic ligand as compared with TCRα(+/-) cells, suggesting increased breadth in peptide recognition may be a mechanism for their reactivity. Our results imply that the role of secondary TCRs in forming the T cell repertoire is perhaps more significant than what has been assumed.
Collapse
Affiliation(s)
- Peggy P Ni
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Benjamin Solomon
- Division of Rheumatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110; and
| | - Chyi-Song Hsieh
- Division of Rheumatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110; and
| | - Paul M Allen
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Gerald P Morris
- Department of Pathology, University of California, San Diego, La Jolla, CA 92093
| |
Collapse
|
27
|
Wang M, Racine J, Zhang M, Wu T, Deng R, Johnston H, Shen C, Siswanto K, Zeng D. MHC-mismatched chimerism is required for induction of transplantation tolerance in autoimmune nonobese diabetic recipients. THE JOURNAL OF IMMUNOLOGY 2014; 193:2005-15. [PMID: 25000982 DOI: 10.4049/jimmunol.1401137] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
In nonautoimmune recipients, induction of mixed and complete chimerism with hematopoietic progenitor cells from MHC (HLA)-matched or -mismatched donors are effective approaches for induction of organ transplantation immune tolerance in both animal models and patients. But it is still unclear whether this is the case in autoimmune recipients. With the autoimmune diabetic NOD mouse model, we report that, although mixed and complete MHC-mismatched chimerism provide immune tolerance to donor-type islet and skin transplants, neither mixed nor complete MHC-matched chimerism does. The MHC-mismatched chimerism not only tolerizes the de novo developed, but also the residual pre-existing host-type T cells in a mismatched MHC class II-dependent manner. In the MHC-mismatched chimeras, the residual host-type peripheral T cells appear to be anergic with upregulation of PD-1 and downregulation of IL-7Rα. Conversely, in the MHC-matched chimeras, the residual host-type peripheral T cells manifest both alloreactivity and autoreactivity; they not only mediate insulitis and sialitis in the recipient, but also reject allogeneic donor-type islet and skin grafts. Interestingly, transgenic autoreactive BDC2.5 T cells from Rag1(+/+), but not from Rag1(-/-), NOD mice show alloreactivity and mediate both insulitis and rejection of allografts. Taken together, MHC-mismatched, but not MHC-matched, chimerism can effectively provide transplantation immune tolerance in autoimmune recipients.
Collapse
Affiliation(s)
- Miao Wang
- Department of Diabetes Research, Beckman Research Institute of City of Hope, Duarte, CA 91010; Department of Hematopoietic Cell Transplantation, Beckman Research Institute of City of Hope, Duarte, CA 91010
| | - Jeremy Racine
- Department of Diabetes Research, Beckman Research Institute of City of Hope, Duarte, CA 91010; Department of Hematopoietic Cell Transplantation, Beckman Research Institute of City of Hope, Duarte, CA 91010; Irell & Manella Graduate School of Biological Sciences of City of Hope, Duarte, CA 91010
| | - Mingfeng Zhang
- Department of Diabetes Research, Beckman Research Institute of City of Hope, Duarte, CA 91010; Irell & Manella Graduate School of Biological Sciences of City of Hope, Duarte, CA 91010
| | - Tao Wu
- Department of Diabetes Research, Beckman Research Institute of City of Hope, Duarte, CA 91010; Department of Hematology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China; and
| | - Ruishu Deng
- Department of Diabetes Research, Beckman Research Institute of City of Hope, Duarte, CA 91010
| | - Heather Johnston
- Department of Diabetes Research, Beckman Research Institute of City of Hope, Duarte, CA 91010; Irell & Manella Graduate School of Biological Sciences of City of Hope, Duarte, CA 91010
| | - Christine Shen
- Eugene and Ruth Roberts Summer Student Academy of City of Hope, Duarte, CA 91010
| | - Kathleen Siswanto
- Eugene and Ruth Roberts Summer Student Academy of City of Hope, Duarte, CA 91010
| | - Defu Zeng
- Department of Diabetes Research, Beckman Research Institute of City of Hope, Duarte, CA 91010; Department of Hematopoietic Cell Transplantation, Beckman Research Institute of City of Hope, Duarte, CA 91010; Irell & Manella Graduate School of Biological Sciences of City of Hope, Duarte, CA 91010;
| |
Collapse
|
28
|
Abstract
Over the past 5 years, many novel approaches to early diagnosis, prevention, and treatment of acute graft-versus-host disease (aGVHD) have been translated from the bench to the bedside. In this review, we highlight recent discoveries in the context of current aGVHD care. The most significant innovations that have already reached the clinic are prophylaxis strategies based upon a refinement of our understanding of key sensors, effectors, suppressors of the immune alloreactive response, and the resultant tissue damage from the aGVHD inflammatory cascade. In the near future, aGVHD prevention and treatment will likely involve multiple modalities, including small molecules regulating immunologic checkpoints, enhancement of suppressor cytokines and cellular subsets, modulation of the microbiota, graft manipulation, and other donor-based prophylaxis strategies. Despite long-term efforts, major challenges in treatment of established aGVHD still remain. Resolution of inflammation and facilitation of rapid immune reconstitution in those with only a limited response to corticosteroids is a research arena that remains rife with opportunity and urgent clinical need.
Collapse
|
29
|
Abstract
T lymphocytes that express dual TCRs were found more frequently in patients with acute graft-versus-host disease (GVHD) as compared with patients who also underwent allogeneic stem cell transplantation but did not develop acute GVHD (Morris et al., this issue).
Collapse
Affiliation(s)
- Benjamin G Vincent
- Department of Medicine and the Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7295, USA
| | | |
Collapse
|
30
|
Cellular Immunotherapy: Using Alloreactivity to Induce Anti-Leukemic Responses without Prolonged Persistence of Donor Cells. Med Sci (Basel) 2013. [DOI: 10.3390/medsci1010037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|