Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Allergy Clin Immunol ; 153(5): 1445-1455, 2024 May.
Article in English | MEDLINE | ID: mdl-38128835

ABSTRACT

BACKGROUND: Autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy (APECED, also called APS-1) is an inborn error of immunity with clear signs of B-cell autoimmunity such as neutralizing anti-IFN antibodies. In APECED, mutations in the AIRE gene impair thymic negative selection of T cells. The resulting T-cell alterations may then cause dysregulation of B-cell responses. However, no analysis of interactions of T and B cells in the germinal centers (GCs) in patients' secondary lymphatic tissues has been reported. OBJECTIVE: This study examined the relationship between B cells and follicular T helper cells (TfH) in peripheral blood and lymph node (LN) GCs in patients with APECED. METHODS: Immunophenotyping of peripheral blood B cells and TfH was performed for 24 patients with APECED. Highly multiplexed fluorescent immunohistochemical staining was performed on 7 LN biopsy samples from the patients to study spatial interactions of lymphocytes in the GCs at the single-cell level. RESULTS: The patients' peripheral B-cell phenotype revealed skewing toward a mature B-cell phenotype with marked loss of transitional and naive B cells. The frequency of circulating TfH cells was diminished in the patients, while in the LNs the TfH population was expanded. In LNs the overall frequency of Treg cells and interactions of Treg cells with nonfollicular T cells were reduced, suggesting that aberrant Treg cell function might fail to restrain TfH differentiation. CONCLUSIONS: GC reactions are disrupted in APECED as a result of defective T-cell control.


Subject(s)
B-Lymphocytes , Germinal Center , Lymph Nodes , Polyendocrinopathies, Autoimmune , T Follicular Helper Cells , Humans , Polyendocrinopathies, Autoimmune/immunology , Polyendocrinopathies, Autoimmune/genetics , Germinal Center/immunology , Female , Male , B-Lymphocytes/immunology , Lymph Nodes/immunology , Lymph Nodes/pathology , Adult , T Follicular Helper Cells/immunology , Adolescent , Child , Young Adult , Middle Aged , Immunophenotyping , AIRE Protein , T-Lymphocytes, Helper-Inducer/immunology
2.
Clin Immunol ; 255: 109733, 2023 10.
Article in English | MEDLINE | ID: mdl-37572949

ABSTRACT

High carrier prevalence of STAT3 SH2 domain somatic mutations was recently discovered in CD8+ T cells. We found these low-allele-fraction clones in 26% of donors, without difference between multiple sclerosis (MS) patients and controls. Here we tested whether anti-viral antibodies associate with the carriership of these mutant clones. We compared antibody responses against common viruses in mutation carriers vs. non-carriers. Plasma samples of 152 donors (92 MS patients, 60 controls) were analyzed for antibodies against cytomegalovirus (CMV), Epstein-Barr virus (EBV), human herpesvirus-6A and parvovirus B19. The mutation carrier status associated with EBV VCA IgG level (p = 0.005) and remained significant after logistic regression (p = 0.036). This association was contributed similarly by MS patients and controls. These results suggest that EBV contributes to the generation or growth of these clones. The pathogenic role of the STAT3 mutant clones in MS is presently unclear, but their detailed characterization warrants further study.


Subject(s)
Epstein-Barr Virus Infections , Multiple Sclerosis , Humans , Herpesvirus 4, Human/genetics , Epstein-Barr Virus Infections/complications , Capsid , src Homology Domains , Antigens, Viral , Antibodies, Viral , Immunoglobulin G , CD8-Positive T-Lymphocytes , STAT3 Transcription Factor/genetics
3.
Eur J Immunol ; 52(6): 882-894, 2022 06.
Article in English | MEDLINE | ID: mdl-35307831

ABSTRACT

Long-term T-cell memory is dependent on the maintenance of memory T cells in the lymphoid tissues, and at the surface interfaces that provide entry routes for pathogens. However, much of the current information on human T-cell memory is based on analyzing circulating T cells. Here, we have studied the distribution and age-related changes of memory T-cell subsets in samples from blood, mesenteric LNs, spleen, and ileum, obtained from donors ranging in age from 5 days to 67 years of age. Our data show that the main reservoir of polyclonal naive cells is found in the LNs, and the resting memory subsets capable of self-renewal are also prominent there. In contrast, nondividing but functionally active memory subsets dominate the spleen, and especially the ileum. In general, the replacement of naive cells with memory subsets continues throughout our period of observation, with no apparent plateau. In conclusion, the analysis of lymphoid and nonlymphoid tissues reveals a dynamic pattern of changes distinct to each tissue, and with substantial differences between CD4+ and CD8+ compartments.


Subject(s)
Lymphoid Tissue , T-Lymphocyte Subsets , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Cell Differentiation , Humans , Immunologic Memory , Lymphocyte Count , Spleen
4.
Immunity ; 39(6): 1143-57, 2013 Dec 12.
Article in English | MEDLINE | ID: mdl-24315997

ABSTRACT

Complement is viewed as a critical serum-operative component of innate immunity, with processing of its key component, C3, into activation fragments C3a and C3b confined to the extracellular space. We report here that C3 activation also occurred intracellularly. We found that the T cell-expressed protease cathepsin L (CTSL) processed C3 into biologically active C3a and C3b. Resting T cells contained stores of endosomal and lysosomal C3 and CTSL and substantial amounts of CTSL-generated C3a. While "tonic" intracellular C3a generation was required for homeostatic T cell survival, shuttling of this intracellular C3-activation-system to the cell surface upon T cell stimulation induced autocrine proinflammatory cytokine production. Furthermore, T cells from patients with autoimmune arthritis demonstrated hyperactive intracellular complement activation and interferon-γ production and CTSL inhibition corrected this deregulated phenotype. Importantly, intracellular C3a was observed in all examined cell populations, suggesting that intracellular complement activation might be of broad physiological significance.


Subject(s)
B-Lymphocyte Subsets/cytology , CD4-Positive T-Lymphocytes/immunology , Cathepsin L/metabolism , Cell Differentiation , Complement Activation/physiology , Complement C3/metabolism , Homeostasis/physiology , Adult , Arthritis, Rheumatoid/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Line , Cell Survival/immunology , Child , Complement C3/immunology , Complement C3a/metabolism , Complement C3b/metabolism , Gene Expression Regulation/immunology , Humans
5.
Clin Immunol ; 231: 108851, 2021 10.
Article in English | MEDLINE | ID: mdl-34508889

ABSTRACT

Almost all patients with autoimmune polyendocrine syndrome type 1 (APS-1) have neutralizing antibodies against type 1 interferons (IFN), important mediators of antiviral defense. Recently, neutralizing anti-IFN antibodies were shown to be a risk factor of severe COVID-19. Here we show in a cohort of 44 patients with APS-1 that higher titers of neutralizing anti-IFNα4 antibodies are associated with a higher and earlier incidence of VZV reactivation (herpes zoster). The patients also present with uncommonly severe clinical sequelae of herpetic infections. APS-1 patients had decreased humoral immune responses to varicella zoster virus, but cellular responses were comparable to healthy controls. These results suggest that blocking the type I interferon pathway in patients with APS-1 patients leads to a clinically significant immune deficiency, and susceptibility to herpesviruses should be taken into account when treating patients with APS-1.


Subject(s)
Herpesvirus 3, Human , Polyendocrinopathies, Autoimmune/complications , Varicella Zoster Virus Infection/complications , Adolescent , Adult , Aged , Child , Cohort Studies , Female , Humans , Immunity, Cellular , Interferon-alpha/immunology , Male , Middle Aged , Polyendocrinopathies, Autoimmune/immunology , Risk Factors , Varicella Zoster Virus Infection/pathology , Young Adult
7.
J Autoimmun ; 119: 102616, 2021 05.
Article in English | MEDLINE | ID: mdl-33652347

ABSTRACT

The T-cell receptor (TCR) repertoire is generated in a semistochastic process of gene recombination and pairing of TCRα to TCRß chains with the estimated total TCR diversity of >108. Despite this high diversity, similar or identical TCR chains are found to recur in immune responses. Here, we analyzed the thymic generation of TCR sequences previously associated with recognition of self- and nonself-antigens, represented by sequences associated with autoimmune diabetes and HIV, respectively. Unexpectedly, in the CD4+ compartment TCRα chains associated with the recognition of self-antigens were generated in significantly higher numbers than TCRα chains associated with the recognition of nonself-antigens. The analysis of the circulating repertoire further showed that these chains are not lost in negative selection nor predominantly converted to the regulatory T-cell lineage. The high abundance of self-reactive TCRα chains in multiple individuals suggests that the human thymus has a predilection to generate self-reactive TCRα chains independently of the HLA-type and that the individual risk of autoimmunity may be modulated by the TCRß repertoire associated with these chains.


Subject(s)
Autoantigens/immunology , Autoimmunity , Receptors, Antigen, T-Cell, alpha-beta/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Thymus Gland/immunology , Thymus Gland/metabolism , Adult , Clonal Selection, Antigen-Mediated , Databases, Genetic , Diabetes Mellitus, Type 1/etiology , Diabetes Mellitus, Type 1/metabolism , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Female , Gene Rearrangement, T-Lymphocyte , Glutamate Decarboxylase/immunology , Humans , Insulin/immunology , Male , Receptors, Antigen, T-Cell, alpha-beta/genetics , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Young Adult
8.
Cell Immunol ; 347: 104026, 2020 01.
Article in English | MEDLINE | ID: mdl-31843201

ABSTRACT

The natural CD25+ FOXP3+ regulatory T cell (Treg) population is generated as a distinct lineage in the thymus, but the details of Treg development in humans remain unclear, and the timing of Treg commitment is also contested. Here we have analyzed the emergence of CD25+ cells at the CD4+CD8+ double positive (DP) stage in the human thymus. We show that these cells share T cell receptor repertoire with CD25+ CD4 single-positive thymocytes, believed to be committed Tregs. They already have a fully demethylated FOXP3 enhancer region and thus display stable expression of FOXP3 and the associated Treg phenotype. Transcriptome analysis also grouped the DP CD25+ and CD4 CD25+ thymocytes apart from the CD25- subsets. Together with earlier studies, our data are consistent with human Treg commitment already at the DP thymocyte stage. We suggest that the most important antigens and signals necessary for human Treg differentiation may be found in the thymic cortex.


Subject(s)
CD8-Positive T-Lymphocytes/cytology , Cell Differentiation/immunology , Forkhead Transcription Factors/metabolism , T-Lymphocytes, Regulatory/cytology , Thymocytes/cytology , Thymus Gland/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/genetics , Child, Preschool , Epigenesis, Genetic/genetics , Female , Gene Expression Profiling , Gene Expression Regulation/genetics , Humans , Infant , Infant, Newborn , Interleukin-2 Receptor alpha Subunit/metabolism , Lymphocyte Activation/immunology , Male , T-Lymphocytes, Regulatory/immunology , Thymocytes/immunology
9.
Scand J Immunol ; : e12681, 2018 Jun 15.
Article in English | MEDLINE | ID: mdl-29904934

ABSTRACT

Thymic commitment of human FOXP3+ regulatory T cells begins at the double positive (DP) CD4+ CD8+ stage. In the current study we show that interleukin-2 promotes the development of FOXP3+ thymocytes and enhances their survival at the DP phase. IL-2 increases the frequency of FOXP3+ cells and promotes the Treg phenotype after TCR-mediated positive selection at the most mature DP stage. However, it has no effect on FOXP3+ cells at the earlier maturation steps before positive selection. DP FOXP3+ are highly susceptible to cell death but IL-2 promotes their survival. The anti-apoptotic protein BCL-2 (B Cell Lymphoma 2) is also upregulated by IL-2 at the most mature DP stage. In addition to IL-2, we identify IL-15 to have a significant role in the upregulating FOXP3 and survival of Tregs at the DP phase. IL-7 also increases the expression of BCL-2 in the DP FOXP3+ thymocytes. Our results indicate that common gamma chain cytokines IL-2, IL-7 and IL-15 promote the development of regulatory T cells at the most mature DP stage after TCR-mediated positive selection through suppressing cell death. This article is protected by copyright. All rights reserved.

10.
J Immunol ; 196(7): 2955-64, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26903483

ABSTRACT

Autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy (APECED) is a monogenic autoimmune disease caused by mutations in the AIRE gene. Although mainly an endocrine disease, a substantial fraction of patients have gastrointestinal manifestations. In this study, we have examined the role of anticommensal responses and their regulation. APECED patients had increased levels of Abs against Saccharomyces cerevisiae (p < 0.0001) and against several species of commensal gut bacteria, but not against species predominantly associated with other locations. The anticommensal Ab levels did not correlate with gastrointestinal autoantibodies, neutralizing anti-IL-17 or -IL-22 Abs, or gastrointestinal symptoms, although scarcity of the available clinical data suggests that further study is required. However, the anti-S. cerevisiae Ab levels showed a significant inverse correlation with FOXP3 expression levels in regulatory T cells (Treg), previously shown to be dysfunctional in APECED. The correlation was strongest in the activated CD45RO(+) population (ρ = -0.706; p < 0.01). APECED patients also had decreased numbers of FOXP3(+) cells in gut biopsies. These results show that APECED patients develop early and sustained responses to gut microbial Ags in a pattern reminiscent of Crohn's disease. This abnormal immune recognition of gut commensals is linked to a systemic Treg defect, which is also reflected as a local decrease of gut-associated Treg. To our knowledge, these data are the first to show dysregulated responses to non-self commensal Ags in APECED and indicate that AIRE contributes to the regulation of gut homeostasis, at least indirectly. The data also raise the possibility of persistent microbial stimulation as a contributing factor in the pathogenesis of APECED.


Subject(s)
Autoantibodies/immunology , Gastrointestinal Microbiome/immunology , Polyendocrinopathies, Autoimmune/immunology , T-Lymphocytes, Regulatory/immunology , Adult , Aged , Antibodies, Fungal/immunology , Autoimmunity , Case-Control Studies , Cytokines/immunology , Cytokines/metabolism , Duodenum/immunology , Duodenum/metabolism , Duodenum/microbiology , Duodenum/pathology , Female , Humans , Immunoglobulin G/immunology , Inflammatory Bowel Diseases/immunology , Male , Middle Aged , Polyendocrinopathies, Autoimmune/genetics , Polyendocrinopathies, Autoimmune/metabolism , T-Lymphocytes, Regulatory/metabolism , Young Adult
11.
Eur J Immunol ; 45(3): 915-21, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25446578

ABSTRACT

In addition to its effector functions, complement is an important regulator of adaptive immune responses. Murine studies suggest that complement modulates helper T-cell differentiation, and Th1 responses in particular are impaired in the absence of functional complement. Here, we have studied humoral responses to toxoid vaccines in eight patients with C3 deficiency, representing more than 25% of all the known patients worldwide. Serum cytokine levels were also studied. The patients developed normal Ig responses to tetanus and diphtheria toxoids, but IgE levels were low. The pattern of antigen-specific IgG subclasses was abnormal, with increased Th1-related IgG3 responses, low IgG2, and almost completely undetectable IgG4. The patients also had increased amounts of Th1-related cytokines IL-12p70 and IL-21, and these showed a positive correlation with IgG3 levels. Our results confirm that complement modulates Th differentiation, but reveal a more nuanced outcome than previously reported. Since IgG4 has been linked to tolerogenic responses, the data also suggest that in the absence of functional complement at least some aspects of systemic tolerance are impaired.


Subject(s)
Cell Differentiation/immunology , Complement C3/deficiency , Immune Tolerance , Immunity, Humoral/immunology , Immunologic Deficiency Syndromes/immunology , Th1 Cells/immunology , Child , Child, Preschool , Complement C3/immunology , Female , Hereditary Complement Deficiency Diseases , Humans , Immunity, Humoral/drug effects , Immunoglobulin E/blood , Immunoglobulin E/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Immunologic Deficiency Syndromes/blood , Immunologic Deficiency Syndromes/pathology , Interleukin-12/blood , Interleukin-12/immunology , Interleukins/blood , Interleukins/immunology , Male , Tetanus Toxoid/administration & dosage , Th1 Cells/metabolism , Th1 Cells/pathology , Young Adult
12.
Cell Immunol ; 295(2): 99-104, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25880100

ABSTRACT

Autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy (APECED) is caused by mutations in the AIRE gene. Murine studies suggest that AIRE controls thymic expression of tissue-restricted antigens, its absence allowing nonselected autoreactive cells to escape. We tested this in humans using the TCRß CDR3 length repertoire as a surrogate of thymic selection, as it shortens during the process. Analysis of healthy thymuses showed an altogether 1.9 base pair shortening, starting at the CD4(+)CD8(+)CD3(low) stage and continuing until the CD4(+) subset, likely encompassing both the positive and negative selection. Comparison of five APECED patients with eight healthy controls showed a skewed repertoire with oligoclonal expansions in the patients' CD4(+) and CD8(+) populations. The average CDR3 length, however, was normal and unaffected by the skewing. This was also true of the hypothesized autoreactive CD8(+)CD45RA(+) population. We failed to detect a subset with an abnormally long CDR3 repertoire, as would be predicted by a failure in selection.


Subject(s)
Complementarity Determining Regions/immunology , Polyendocrinopathies, Autoimmune/immunology , Receptors, Antigen, T-Cell, alpha-beta/immunology , Adult , Complementarity Determining Regions/genetics , Female , Flow Cytometry , Humans , Leukocytes, Mononuclear/immunology , Male , Middle Aged , Polyendocrinopathies, Autoimmune/genetics , RNA/chemistry , RNA/genetics , Receptors, Antigen, T-Cell, alpha-beta/genetics , Reverse Transcriptase Polymerase Chain Reaction , Selection, Genetic
13.
Eur J Immunol ; 43(8): 2185-93, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23670527

ABSTRACT

The T-cell repertoire depends on intrathymic genetic rearrangement events in the T-cell receptor (TCR) locus, followed by positive and negative selection. The repertoire thus generated is highly diverse, but recent data indicate that the recombination of gene segments is less stochastic than previously suggested. Very little is known of the junctional complementarity determining region 3 (CDR3), which is to a large degree not germline encoded. We have analyzed the development of the human TCR ß CDR3 repertoire, from the nonselected CD4(+) CD8(+) CD3(-) cells up to the fully selected CD4(+) CD8(-) thymocytes. In addition to spectratyping, a fraction of the CDR3 repertoire was sequenced and a structural in silico analysis of the CDR3 loop characteristics performed. Our data show that the thymic TCR repertoire is extremely diverse, and the effect of the selection events can be detected as a measurable loss of polyclonality in the CDR3 loop. However, the main physicochemical features of the CDR3 loop were found already at the nonselected repertoire and showed no progressive changes during the selection. Thus, the main structural characteristics of the CDR3 loop were already determined by the recombination process and not significantly affected by the extensive thymocyte death associated with selection in the thymus.


Subject(s)
CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Complementarity Determining Regions/genetics , Receptors, Antigen, T-Cell, alpha-beta/genetics , Thymus Gland/embryology , Amino Acid Sequence , Base Sequence , Cells, Cultured , Child, Preschool , Female , Gene Rearrangement, beta-Chain T-Cell Antigen Receptor , Humans , Infant , Infant, Newborn , Male , Sequence Analysis, DNA
14.
J Allergy Clin Immunol ; 131(4): 1167-75, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23098807

ABSTRACT

BACKGROUND: Cells of the innate immune system regulate both adaptive immune responses and the maintenance of tolerance, especially in the gut. However, relatively little is known about the effects of complement on lymphocyte homeostasis. OBJECTIVE: This study explored complement C3 deficiency in mice and human subjects for its effect on intestinal tolerance. METHODS: C3-deficient mice and control C57BL/6 mice were fed ovalbumin (OVA) by means of gavage, and subsequent response to immunization with OVA in Freund's adjuvant was monitored. Serum antibodies against commensal microbes were measured, and the activation status of peripheral blood lymphocytes bearing mucosal homing markers was determined from 2 rare cases of C3-deficient patients. RESULTS: We show in C3-deficient mice and human patients that intestinal tolerance fails in the absence of functional complement. In contrast to wild-type control animals, in which oral tolerance was induced, intragastric administration of OVA did not result in a significantly decreased response to subsequent subcutaneous OVA challenge in C3-deficient mice. In the jejunum of C3-deficient mice the cytokine ratio between IL-10 and IFN-γ or IL-17 levels was decreased, indicating a shift in favor of proinflammatory cytokines. In 2 C3-deficient children the frequency of gut-homing T cells expressing activation markers was increased, and the patients had increased serum IgG levels against gut commensal microbes. The data also suggest that the impaired oral tolerance was at least partly caused by the absence of signaling through C3-binding complement regulators in T cells. CONCLUSIONS: Taken together, our results identify complement as an important and nonredundant regulator of intestinal tolerance.


Subject(s)
Antigens, Bacterial/immunology , Antigens, Fungal/immunology , Complement C3/deficiency , Immune Tolerance , Immunity, Mucosal , Jejunum/immunology , Administration, Oral , Adult , Animals , Case-Control Studies , Child, Preschool , Complement C3/genetics , Complement C3/immunology , Freund's Adjuvant/immunology , Humans , Immunoglobulin G/immunology , Injections, Subcutaneous , Interferon-gamma/immunology , Interleukin-10/immunology , Interleukin-17/immunology , Jejunum/microbiology , Jejunum/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Ovalbumin/immunology , T-Lymphocytes/immunology , T-Lymphocytes/microbiology , T-Lymphocytes/pathology , Young Adult
15.
J Immunol ; 187(4): 2023-30, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21753149

ABSTRACT

Autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy (APECED) is a monogenic autoimmune disease that is caused by mutations in the AIRE gene. Murine studies have linked AIRE to thymocyte selection and peripheral deletional tolerance, but the pathogenesis of the human disease remains unclear. In this study, we show that APECED patients have elevated IL-7 levels and a drastically decreased expression of IL-7R on CD8(+) T cells. This is associated with increased proliferation and a decreased expression of the negative TCR regulator CD5 in the CD45RO(-) subset. The CD45RO(-) cells also display oligoclonal expansions, decreased expression of the lymph node homing factors CCR7 and CD62L, and increased expression of perforin, consistent with the accumulation of highly differentiated effector cells. The CD45RO(-)CCR7(+)CD8(+) population of cells with markers characteristic of naive phenotype is also skewed, as shown by decreased expression of CD5 and increased expression of perforin. The putative CD31(+) recent thymic emigrant population is likewise affected. These data are consistent with IL-7 dysregulation inducing a decreased threshold of TCR signaling and self-antigen-driven proliferation, probably in synergy with the failed thymic selection. The resultant loss of CD8(+) T cell homeostasis is likely to play a significant role in the pathogenesis of APECED. Our findings may also hold lessons for other diseases in which the IL-7-IL-7R pathway has emerged as a risk factor.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Gene Expression Regulation/immunology , Homeostasis/immunology , Interleukin-7/immunology , Polyendocrinopathies, Autoimmune/immunology , Transcription Factors/immunology , Adult , Animals , Antigens, Differentiation/blood , Antigens, Differentiation/genetics , Antigens, Differentiation/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , Child , Child, Preschool , Female , Gene Expression Regulation/genetics , Homeostasis/genetics , Humans , Interleukin-7/biosynthesis , Interleukin-7/genetics , Male , Mice , Middle Aged , Polyendocrinopathies, Autoimmune/blood , Polyendocrinopathies, Autoimmune/genetics , Polyendocrinopathies, Autoimmune/pathology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Receptors, Interleukin-7/blood , Receptors, Interleukin-7/genetics , Receptors, Interleukin-7/immunology , Thymus Gland/immunology , Thymus Gland/metabolism , Thymus Gland/pathology , Transcription Factors/genetics , Transcription Factors/metabolism , AIRE Protein
16.
Cancer Med ; 12(12): 13486-13496, 2023 06.
Article in English | MEDLINE | ID: mdl-37114587

ABSTRACT

BACKGROUND: The number of mutations in cancer cells is an important predictor of a positive response to cancer immunotherapy. It has been suggested that the neoantigens produced by these mutations are more immunogenic than nonmutated tumor antigens, which are likely to be protected by immunological tolerance. However, the mechanisms of tolerance as regards tumor antigens are incompletely understood. METHODS: Here, we have analyzed the impact of thymic negative selection on shared T-cell receptor (TCR) repertoire associated with the recognition of either mutated or nonmutated tumor antigens by comparing previously known TCR-antigen-pairs to TCR repertoires of 21 immunologically healthy individuals. RESULTS: Our results show that TCRα chains associated with either type of tumor antigens are readily generated in the thymus, at a frequency similar to TCRα chains associated with nonself. In the peripheral repertoire, the relative clone size of nonself-associated chains is higher than that of the tumor antigens, but importantly, there is no difference between TCRα chains associated with mutated or nonmutated tumor antigens. CONCLUSION: This suggests that the tolerance mechanisms protecting nonmutated tumor antigens are non-deletional and therefore potentially reversible. As unmutated antigens are, unlike mutations, shared by a large number of patients, they may offer advantages in designing immunological approaches to cancer treatment.


Subject(s)
Antigens, Neoplasm , Immune Tolerance , Neoplasms , Receptors, Antigen, T-Cell, alpha-beta , Thymus Gland , Thymus Gland/immunology , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/therapy , Humans , Immune Tolerance/genetics , Mutation , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/immunology
17.
Pathog Immun ; 8(2): 74-87, 2023.
Article in English | MEDLINE | ID: mdl-38347963

ABSTRACT

Background: Lymphopenia is common in COVID-19. This has raised concerns that COVID-19 could affect the immune system akin to measles infection, which causes immune amnesia and a reduction in protective antibodies. Methods: We recruited COVID-19 patients (n = 59) in Helsinki, Finland, and collected plasma samples on 2 to 3 occasions during and after infection. We measured IgG antibodies to diphtheria toxin, tetanus toxoid, and pertussis toxin, along with total IgG, SARS-CoV-2 spike protein IgG, and neutralizing antibodies. We also surveyed the participants for up to 17 months for long-term impaired olfaction as a proxy for prolonged post-acute COVID-19 symptoms. Results: No significant differences were found in the unrelated vaccine responses while the serological response against COVID-19 was appropriate. During the acute phase of the disease, the SARSCoV-2 IgG levels were lower in outpatients when compared to inpatients. SARS-CoV-2 serology kinetics matched expectations. In the acute phase, anti-tetanus and anti-diphtheria IgG levels were lower in patients with prolonged impaired olfaction during follow up than in those without. Conclusions: We could not detect significant decline in overall humoral immunity during or after COVID-19 infection. In severe COVID-19, there appears to be a temporary decline in total IgG levels.

18.
Scand J Immunol ; 74(1): 71-9, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21352256

ABSTRACT

Transcriptional regulator autoimmune regulator (AIRE) controls thymic negative selection but it is also expressed in secondary lymphoid organs. The relative contribution of AIRE's central and peripheral function to the maintenance of tolerance is unclear. We transferred mature lymphocytes from Aire(-/-) or wild-type donors to Aire(+/+) lymphopenic recipients, which allowed us to gauge the autoreactivity inherent in the cells originating in an Aire(-/-) thymus. In the ensuing lymphopenia-induced proliferation (LIP), the recipients of cells from Aire(-/-) showed definite T cell hyperproliferation and developed autoantibodies at a higher frequency than the recipients of wild-type cells. However, neither of the recipient groups developed clinical symptoms, and pathological tissue infiltrates were also absent. The recipients of Aire(-/-) cells showed hyperproliferation and increased accumulation of regulatory T cells (Tregs), especially in tissues susceptible to inflammation triggered by LIP. These data are consistent with the view that T cells developing in the absence of Aire are autoreactive. However, overt autoimmunity was prevented, most likely by the suppressive function of Treg cells in the Aire-sufficient recipients. Our results support the importance of the peripheral AIRE expression in the maintenance of immunological tolerance.


Subject(s)
Autoantibodies/immunology , Autoimmunity/genetics , Immune Tolerance/genetics , Lymphopenia/immunology , T-Lymphocytes, Regulatory/immunology , Transcription Factors/physiology , Adoptive Transfer , Animals , Autoantibodies/analysis , Cell Proliferation , Mice , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes, Regulatory/transplantation , Thymus Gland/immunology , Transcription Factors/genetics , AIRE Protein
19.
Vaccine ; 39(13): 1818-1825, 2021 03 26.
Article in English | MEDLINE | ID: mdl-33678453

ABSTRACT

Persistence of immune memory in humans is a crucial yet poorly understood aspect of immunology. Here we have studied the effect of Puumala hantavirus infection on unrelated, pre-existing immune memory by studying T cell- and antibody responses against toxoid vaccine antigens of diphtheria, tetanus and pertussis in a cohort of 45 patients. We found that tetanus- and pertussis -specific IgG concentrations elevate during acute Puumala virus infection. Increase in vaccine IgG was associated with proliferation of heterologous T cells. Interestingly, increases in tetanus-specific IgG persisted a year after the infection while pertussis-specific IgG declined rapidly; a difference in IgG kinetics resembling the difference seen after vaccination against tetanus and pertussis. These results suggest that persistence of immune memory is facilitated by heterologous boosting of old memory during memory formation against newly encountered antigens. They also show that different toxoid antigens may be treated differently. Our study gives new insight into how immune memory formation may alter pre-existing immune memory, and also shows that heterologous immunity may have an impact on vaccination outcomes.


Subject(s)
Diphtheria-Tetanus-acellular Pertussis Vaccines , Diphtheria , Tetanus , Whooping Cough , Antibodies, Bacterial , Diphtheria-Tetanus-Pertussis Vaccine , Humans , Tetanus Toxoid , Toxoids
20.
Data Brief ; 35: 106751, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33553521

ABSTRACT

T cell receptor (TCR) is a heterodimer consisting of TCRα and TCRß chains that are generated by somatic recombination of multiple gene segments. Nascent TCR repertoire undergoes thymic selections where non-functional and potentially autoreactive receptors are removed. During the last years, the development of high-throughput sequencing technology has allowed a large scale assessment of TCR repertoire and multiple analysis tools are now also available. In our recent manuscript, Human thymic T cell repertoire is imprinted with strong convergence to shared sequences[1], we show highly overlapping thymic TCR repertoires in unrelated individuals. In the current Data in Brief article, we provide a more detailed characterization of the basic features of these thymic and related peripheral blood TCR repertoires. The thymus samples were collected from eight infants undergoing corrective cardiac surgery, two of whom were monozygous twins [2]. In parallel with the surgery, a small aliquot of peripheral blood was drawn from four of the donors. Genomic DNA was extracted from mechanically released thymocytes and circulating leukocytes. The sequencing of TCRα and TCRß repertoires was performed at ImmunoSEQ platform (Adaptive Biotechnologies). The obtained repertoire data were analysed applying relevant features from immunoSEQ® 3.0 Analyzer (Adaptive Biotechnologies) and a freely available VDJTools software package for programming language R [3]. The current data analysis displays the basic features of the sequenced repertoires including observed TCR diversity, various descriptive TCR diversity measures, and V and J gene usage. In addition, multiple methods to calculate repertoire overlap between two individuals are applied. The raw sequence data provide a large database of reference TCRs in healthy individuals at an early developmental stage. The data can be exploited to improve existing computational models on TCR repertoire behaviour as well as in the generation of new models.

SELECTION OF CITATIONS
SEARCH DETAIL