Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2023 Jan 24.
Article in English | MEDLINE | ID: mdl-36711832

ABSTRACT

Foxp3+ regulatory T cells (Tregs) are capable suppressors of aberrant self-reactivity. However, TCR affinity and specificities that support Treg function, and how these compare to autoimmune T cells remain unresolved. In this study, we used antigen agnostic and epitope-focused analyses to compare TCR repertoires of regulatory and effector T cells that spontaneously infiltrate pancreatic islets of non-obese diabetic mice. We show that effector and regulatory T cell-derived TCRs possess similar wide-ranging reactivity for self-antigen. Treg-derived TCRs varied in their capacity to confer optimal protective function, and Treg suppressive capacity was in part determined by effector TCR affinity. Interestingly, when expressing the same TCR, Tregs showed higher Nur77-GFP expression than Teffs, suggesting Treg-intrinsic ability to compete for antigen. Our findings provide a new insight into TCR-dependent and independent mechanisms that regulate Treg function and indicate a TCR-intrinsic insufficiency in tissue-specific Tregs that may contribute to the pathogenesis of type 1 diabetes.

2.
Eur J Immunol ; 52(7): 1158-1170, 2022 07.
Article in English | MEDLINE | ID: mdl-35389516

ABSTRACT

The contribution of low-affinity T cells to autoimmunity in the context of polyclonal T-cell responses is understudied due to the limitations in their capture by tetrameric reagents and low level of activation in response to antigenic stimulation. As a result, low-affinity T cells are often disregarded as nonantigen-specific cells irrelevant to the immune response. Our study aimed to assess how the level of self-antigen reactivity shapes T-cell lineage and effector responses in the context of spontaneous tissue-specific autoimmunity observed in NOD mice. Using multicolor flow cytometry in combination with Nur77GFP reporter of TCR signaling, we identified a dormant population of T cells that infiltrated the pancreatic islets of prediabetic NOD mice, which exhibited reduced levels of self-tissue reactivity based on expression of CD5 and Nur77GFP . We showed that these CD5low T cells had a unique TCR repertoire and exhibited low activation and minimal effector function; however, induced rapid diabetes upon transfer. The CD4+ CD5low T-cell population displayed transcriptional signature of central memory T cells, consistent with the ability to acquire effector function post-transfer. Transcriptional profile of CD5low T cells was similar to T cells expressing a low-affinity TCR, indicating TCR affinity to be an important factor in shaping CD5low T-cell phenotype and function at the tissue site. Overall, our study suggests that autoimmune tissue can maintain a reservoir of undifferentiated central memory-like autoreactive T cells with pathogenic effector potential that might be an important source for effector T cells during long-term chronic autoimmunity.


Subject(s)
Diabetes Mellitus, Type 1 , Animals , CD4-Positive T-Lymphocytes , Mice , Mice, Inbred NOD , Mice, Transgenic , Receptors, Antigen, T-Cell/genetics
3.
J Immunol ; 203(2): 349-359, 2019 07 15.
Article in English | MEDLINE | ID: mdl-31182478

ABSTRACT

Loss of immune tolerance to self-antigens can promote chronic inflammation and disrupt the normal function of multiple organs, including the lungs. Degradation of elastin, a highly insoluble protein and a significant component of the lung structural matrix, generates proinflammatory molecules. Elastin fragments (EFs) have been detected in the serum of smokers with emphysema, and elastin-specific T cells have also been detected in the peripheral blood of smokers with emphysema. However, an animal model that could recapitulate T cell-specific autoimmune responses by initiating and sustaining inflammation in the lungs is lacking. In this study, we report an animal model of autoimmune emphysema mediated by the loss of tolerance to elastin. Mice immunized with a combination of human EFs plus rat EFs but not mouse EFs showed increased infiltration of innate and adaptive immune cells to the lungs and developed emphysema. We cloned and expanded mouse elastin-specific CD4+ T cells from the lung and spleen of immunized mice. Finally, we identified TCR sequences from the autoreactive T cell clones, suggesting possible pathogenic TCRs that can cause loss of immune tolerance against elastin. This new autoimmune model of emphysema provides a useful tool to examine the immunological factors that promote loss of immune tolerance to self.


Subject(s)
Autoimmunity/immunology , Elastin/immunology , Lung/immunology , Pulmonary Emphysema/immunology , Adaptive Immunity/immunology , Animals , Cell Line , Disease Models, Animal , Female , HEK293 Cells , Humans , Immune Tolerance/immunology , Immunity, Innate/immunology , Inflammation/immunology , Mice , Mice, Inbred C57BL , Pulmonary Disease, Chronic Obstructive/immunology , Smoking/immunology
4.
Immunology ; 156(4): 305-318, 2019 04.
Article in English | MEDLINE | ID: mdl-30560993

ABSTRACT

The immune system is essential for maintaining a delicate balance between eliminating pathogens and maintaining tolerance to self-tissues to avoid autoimmunity. An enormous and complex community of gut microbiota provides essential health benefits to the host, particularly by regulating immune homeostasis. Many of the metabolites derived from commensals can impact host health by directly regulating the immune system. Many autoimmune diseases arise from an imbalance between pathogenic effector T cells and regulatory T (Treg) cells. Recent interest has emerged in understanding how cross-talk between gut microbiota and the host immune system promotes autoimmune development by controlling the differentiation and plasticity of T helper and Treg cells. At the molecular level, our recent study, along with others, demonstrates that asymptomatic colonization by commensal bacteria in the gut is capable of triggering autoimmune disease by molecular mimicking self-antigen and skewing the expression of dual T-cell receptors on T cells. Dysbiosis, an imbalance of the gut microbiota, is involved in autoimmune development in both mice and humans. Although it is well known that dysbiosis can impact diseases occurring within the gut, growing literature suggests that dysbiosis also causes the development of gut-distal/non-gut autoimmunity. In this review, we discuss recent advances in understanding the potential molecular mechanisms whereby gut microbiota induces autoimmunity, and the evidence that the gut microbiota triggers gut-distal autoimmune diseases.


Subject(s)
Autoimmunity/immunology , Gastrointestinal Microbiome/immunology , T-Lymphocytes/immunology , Animals , Dysbiosis/immunology , Humans
5.
JCI Insight ; 3(2)2018 01 25.
Article in English | MEDLINE | ID: mdl-29367462

ABSTRACT

T cell receptor (TCR) affinity is a critical factor of Treg lineage commitment, but whether self-reactivity is a determining factor in peripheral Treg function remains unknown. Here, we report that a high degree of self-reactivity is crucial for tissue-specific Treg function in autoimmunity. Based on high expression of CD5, we identified a subset of self-reactive Tregs expressing elevated levels of T-bet, GITR, CTLA-4, and ICOS, which imparted significant protection from autoimmune diabetes. We observed that T-bet expression in Tregs, necessary for control of Th1 autoimmunity, could be induced in an IFNγ-independent fashion and, unlike in conventional T cells (Tconv), was strongly correlated with the strength of TCR signaling. The level of CD5 similarly identified human Tregs with an increased functional profile, suggesting that CD5hi Tregs may constitute an efficacious subpopulation appropriate for use in adoptive Treg therapies for treatment of inflammatory conditions. Overall, this work establishes an instrumental role of high TCR self-reactivity in driving Treg function.


Subject(s)
Autoimmunity , Diabetes Mellitus, Type 1/immunology , Receptors, Antigen, T-Cell/metabolism , T-Box Domain Proteins/metabolism , T-Lymphocytes, Regulatory/immunology , Adult , Animals , CD5 Antigens , Cells, Cultured , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/therapy , Disease Models, Animal , Female , Healthy Volunteers , Humans , Immunotherapy, Adoptive/methods , Islets of Langerhans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Primary Cell Culture , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , T-Box Domain Proteins/immunology , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/transplantation , Young Adult
6.
J Immunol ; 200(3): 909-914, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29282307

ABSTRACT

Regulatory T cells (Tregs) use a distinct TCR repertoire and are more self-reactive compared with conventional T cells. However, the extent to which TCR affinity regulates the function of self-reactive Tregs is largely unknown. In this study, we used a two-TCR model to assess the role of TCR affinity in Treg function during autoimmunity. We observed that high- and low-affinity Tregs were recruited to the pancreas and contributed to protection from autoimmune diabetes. Interestingly, high-affinity cells preferentially upregulated the TCR-dependent Treg functional mediators IL-10, TIGIT, GITR, and CTLA4, whereas low-affinity cells displayed increased transcripts for Areg and Ebi3, suggesting distinct functional profiles. The results of this study suggest mechanistically distinct and potentially nonredundant roles for high- and low-affinity Tregs in controlling autoimmunity.


Subject(s)
Autoimmunity/immunology , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/prevention & control , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes, Regulatory/immunology , Amphiregulin/biosynthesis , Animals , CTLA-4 Antigen/biosynthesis , Cell Adhesion/immunology , Glucocorticoid-Induced TNFR-Related Protein/biosynthesis , Interleukin-10/biosynthesis , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Minor Histocompatibility Antigens/biosynthesis , Pancreas/cytology , Pancreas/immunology , Receptors, Cytokine/biosynthesis , Receptors, Immunologic/biosynthesis
7.
J Vis Exp ; (127)2017 09 10.
Article in English | MEDLINE | ID: mdl-28930975

ABSTRACT

Although, several methods for sequencing of paired T cell receptor (TCR) alpha and beta chains from single T cells have been developed, none so far have been conducive to downstream in vivo functional analysis of TCR heterodimers. We have developed an improved protocol based on a two-step multiplex-nested PCR, which results in a PCR product that spans entire variable regions of a human TCR alpha and beta chains. By identifying unique restriction sites and incorporating them into the PCR primers, we have made the PCR product compatible with direct sub-cloning into the template retroviral vector. The resulting retroviral construct encodes a chimeric human/mouse TCR with a mouse intracellular domain, which is functional in mouse cells or in in vivo mouse models. Overall, the protocol described here combines human single cell paired TCR alpha and beta chain identification with streamlined generation of retroviral vectors readily adaptable for in vitro and in vivo TCR expression. The video and the accompanying material are designed to give a highly detailed description of the single cell PCR, so that the critical steps can be followed and potential pitfalls avoided. Additionally, we provide a detailed description of the cloning steps necessary to generate the expression vector. Once mastered, the whole procedure from single cell sorting to TCR expression could be performed in a short two-week period.


Subject(s)
Genetic Vectors/genetics , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/isolation & purification , Animals , Humans , Mice
8.
J Immunol ; 199(7): 2270-2278, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28835461

ABSTRACT

Type 1 diabetes is a T cell-mediated autoimmune disease that is characterized by Ag-specific targeting and destruction of insulin-producing ß cells. Although multiple studies have characterized the pathogenic potential of ß cell-specific T cells, we have limited mechanistic insight into self-reactive autoimmune T cell development and their escape from negative selection in the thymus. In this study, we demonstrate that ectopic expression of insulin epitope B:9-23 (InsB9-23) by thymic APCs is insufficient to induce deletion of high- or low-affinity InsB9-23-reactive CD4+ T cells; however, we observe an increase in the proportion and number of thymic and peripheral Foxp3+ regulatory T cells. In contrast, the MHC stable insulin mimetope (InsB9-23 R22E) efficiently deletes insulin-specific T cells and prevents escape of high-affinity thymocytes. Collectively, these results suggest that Ag dose and peptide-MHC complex stability can lead to multiple fates of insulin-reactive CD4+ T cell development and autoimmune disease outcome.


Subject(s)
Autoantigens/genetics , Autoimmunity , CD4-Positive T-Lymphocytes/physiology , Ectopic Gene Expression , Insulin/genetics , Peptide Fragments/genetics , T-Lymphocytes, Regulatory/immunology , Animals , Autoantigens/immunology , Autoimmune Diseases/immunology , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation , Diabetes Mellitus, Type 1/immunology , Insulin/immunology , Insulin-Secreting Cells/immunology , Mice , Peptide Fragments/immunology , Thymocytes/immunology
9.
J Immunol Methods ; 439: 29-36, 2016 12.
Article in English | MEDLINE | ID: mdl-27589924

ABSTRACT

Single-cell paired TCR identification is a powerful tool, but has been limited in its previous incompatibility with further functional analysis. The current protocol describes a method to clone and functionally evaluate in vivo TCRs derived from single antigen-responsive human T cells and monoclonal T cell lines. We have improved upon current PCR-based TCR sequencing protocols by developing primers that allow amplification of human TCRα and TCRß variable regions, while incorporating specific restriction cut sites for direct subcloning into the template retroviral vector. This streamlined approach for generating human:mouse chimeric TCR vectors allows for rapid TCR expression in humanized-retrogenic (hu-Rg) mice through retroviral mediated stem cell gene transfer. Using widely available techniques and equipment, this method is easily adaptable by most laboratories. This is the first TCR identification protocol that is efficiently combined with subsequent in vivo TCR expression.


Subject(s)
Cloning, Molecular/methods , Diabetes Mellitus, Type 1/genetics , Gene Transfer Techniques , Genetic Vectors , Receptors, Antigen, T-Cell, alpha-beta/genetics , Retroviridae/genetics , Stem Cell Transplantation , Stem Cells/immunology , T-Lymphocytes/immunology , Animals , Cell Separation/methods , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/metabolism , Genotype , HEK293 Cells , HLA Antigens/genetics , HLA Antigens/immunology , Humans , Mice, Inbred NOD , Mice, Transgenic , Multiplex Polymerase Chain Reaction , Phenotype , Receptors, Antigen, T-Cell, alpha-beta/immunology , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/metabolism , T-Lymphocytes/metabolism , Time Factors , Transduction, Genetic , Transfection
10.
J Vis Exp ; (113)2016 07 11.
Article in English | MEDLINE | ID: mdl-27500835

ABSTRACT

T cell receptor (TCR) signaling is essential in the development and differentiation of T cells in the thymus and periphery, respectively. The vast array of TCRs proves studying a specific antigenic response difficult. Therefore, TCR transgenic mice were made to study positive and negative selection in the thymus as well as peripheral T cell activation, proliferation and tolerance. However, relatively few TCR transgenic mice have been generated specific to any given antigen. Thus, studies involving TCRs of varying affinities for the same antigenic peptide have been lacking. The generation of a new TCR transgenic line can take six or more months. Additionally, any specific backcrosses can take an additional six months. In order to allow faster generation and screening of multiple TCRs, a protocol for retroviral transduction of bone marrow was established with stoichiometric expression of the TCRα and TCRß chains and the generation of retrogenic mice. Each retrogenic mouse is essentially a founder, virtually negating a founder effect, while the length of time to generate a TCR retrogenic is cut from six months to approximately six weeks. Here we present a rapid and flexible alternative to TCR transgenic mice that can be expressed on any chosen background with any particular TCR.


Subject(s)
Mice, Transgenic , Receptors, Antigen, T-Cell/genetics , Retroviridae , Stem Cells/cytology , Transduction, Genetic/methods , Animals , Bone Marrow , Mice , T-Lymphocytes , Thymus Gland
SELECTION OF CITATIONS
SEARCH DETAIL
...