Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 650
Filter
1.
EMBO J ; 43(13): 2552-2581, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38637625

ABSTRACT

Natural killer (NK) cells are critical to the innate immune system, as they recognize antigens without prior sensitization, and contribute to the control and clearance of viral infections and cancer. However, a significant proportion of NK cells in mice and humans do not express classical inhibitory receptors during their education process and are rendered naturally "anergic", i.e., exhibiting reduced effector functions. The molecular events leading to NK cell anergy as well as their relation to those underlying NK cell exhaustion that arises from overstimulation in chronic conditions, remain unknown. Here, we characterize the "anergic" phenotype and demonstrate functional, transcriptional, and phenotypic similarities to the "exhausted" state in tumor-infiltrating NK cells. Furthermore, we identify zinc finger transcription factor Egr2 and diacylglycerol kinase DGKα as common negative regulators controlling NK cell dysfunction. Finally, experiments in a 3D organotypic spheroid culture model and an in vivo tumor model suggest that a nanoparticle-based delivery platform can reprogram these dysfunctional natural killer cell populations in their native microenvironment. This approach may become clinically relevant for the development of novel anti-tumor immunotherapeutic strategies.


Subject(s)
Killer Cells, Natural , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Animals , Mice , Humans , Early Growth Response Protein 2/metabolism , Early Growth Response Protein 2/genetics , Early Growth Response Protein 2/immunology , Clonal Anergy/immunology , Neoplasms/immunology , Neoplasms/therapy , Neoplasms/pathology , Mice, Inbred C57BL
2.
Cell Rep ; 38(3): 110259, 2022 01 18.
Article in English | MEDLINE | ID: mdl-35045301

ABSTRACT

CD21low age-associated or atypical memory B cells are autoantibody enriched and poised for plasma cell differentiation. These cells overaccumulate in chronic infections, autoimmune disease, and immunodeficiency, posing the question of what checkpoints normally oppose their accumulation. Here, we reveal a critical role for paralogous calcium-NFAT-regulated transcription factors EGR2 and EGR3 that are induced in self-reactive B cells. CD21low and B1 B cells lacking EGR2 and EGR3 accumulate and circulate in young mice in numbers 10- to 20-fold greater than normal and overexpress a large set of EGR2 ChIP-seq target genes, including known drivers of plasma cell differentiation. Most follicular B cells constitutively express Egr2 proportionally to surface IgM downregulation by self-antigens, and EGR2/3 deficiency abolishes this cardinal feature of B cell anergy. These results explain the cardinal features of B cell anergy, define a key transcriptional checkpoint repressing CD21low B cell formation, and inform how NFATC1 or EGR2 mutations promote B1 cell-derived chronic lymphocytic leukemias.


Subject(s)
B-Lymphocytes/immunology , Clonal Anergy/immunology , Early Growth Response Protein 2/immunology , Early Growth Response Protein 3/immunology , Animals , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , Autoimmunity/immunology , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , B-Lymphocytes/metabolism , Early Growth Response Protein 2/metabolism , Early Growth Response Protein 3/metabolism , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Male , Mice , Receptors, Complement 3d/immunology
3.
J Immunol ; 208(2): 371-383, 2022 01 15.
Article in English | MEDLINE | ID: mdl-34965965

ABSTRACT

Monoallelic AgR gene expression underlies specific adaptive immune responses. AgR allelic exclusion is achieved by sequential initiation of V(D)J recombination between alleles and resultant protein from one allele signaling to prevent recombination of the other. The ATM kinase, a regulator of the DNA double-strand break (DSB) response, helps enforce allelic exclusion through undetermined mechanisms. ATM promotes repair of RAG1/RAG2 (RAG) endonuclease-induced DSBs and transduces signals from RAG DSBs during Igk gene rearrangement on one allele to transiently inhibit RAG1 protein expression, Igk accessibility, and RAG cleavage of the other allele. Yet, the relative contributions of ATM functions in DSB repair versus signaling to enforce AgR allelic exclusion remain undetermined. In this study, we demonstrate that inactivation in mouse pre-B cells of the NF-κB essential modulator (Nemo) protein, an effector of ATM signaling, diminishes RAG DSB-triggered repression of Rag1/Rag2 transcription and Igk accessibility but does not result in aberrant repair of RAG DSBs like ATM inactivation. We show that Nemo deficiency increases simultaneous biallelic Igk cleavage in pre-B cells and raises the frequency of B cells expressing Igκ proteins from both alleles. In contrast, the incidence of biallelic Igκ expression is not elevated by inactivation of the SpiC transcriptional repressor, which is induced by RAG DSBs in an ATM-dependent manner and suppresses Igk accessibility. Thus, we conclude that Nemo-dependent, ATM-mediated DNA damage signals enforce Igκ allelic exclusion by orchestrating transient repression of RAG expression and feedback inhibition of additional Igk rearrangements in response to RAG cleavage on one Igk allele.


Subject(s)
DNA Breaks, Double-Stranded , DNA Repair/genetics , Immunoglobulins/immunology , Intracellular Signaling Peptides and Proteins/metabolism , Loss of Heterozygosity/genetics , Animals , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Cells, Cultured , Clonal Anergy/genetics , Clonal Anergy/immunology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Immunoglobulin Variable Region/genetics , Immunoglobulin Variable Region/immunology , Immunoglobulins/genetics , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , V(D)J Recombination/genetics
4.
J Immunol ; 208(2): 293-302, 2022 01 15.
Article in English | MEDLINE | ID: mdl-34930782

ABSTRACT

A considerable proportion of peripheral B cells is autoreactive, and it is unclear how the activation of such potentially harmful cells is regulated. In this study, we show that the different activation thresholds or IgM and IgD BCRs adjust B cell activation to the diverse requirements during development. We rely on the autoreactive 3-83 model BCR to generate and analyze mice expressing exclusively autoreactive IgD BCRs on two different backgrounds that determine two stages of autoreactivity, depending on the presence or absence of the cognate Ag. By comparing these models with IgM-expressing control mice, we found that, compared with IgM, IgD has a higher activation threshold in vivo, as it requires autoantigen to enable normal B cell development, including allelic exclusion. Our data indicate that IgM provides the high sensitivity required during early developmental stages to trigger editing of any autoreactive specificities, including those enabling weak interaction with autoantigen. In contrast, IgD has the unique ability to neglect weakly interacting autoantigens while retaining reactivity to higher-affinity Ag. This IgD function enables mature B cells to ignore autoantigens while remaining able to efficiently respond to foreign threats.


Subject(s)
Autoantigens/immunology , B-Lymphocytes/immunology , Clonal Anergy/immunology , Immunoglobulin D/immunology , Receptors, Antigen, B-Cell/immunology , Animals , Antibody Specificity/immunology , Cell Line , Gene Knock-In Techniques , Immunoglobulin Heavy Chains/genetics , Immunoglobulin M/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
6.
Front Immunol ; 12: 680279, 2021.
Article in English | MEDLINE | ID: mdl-34335582

ABSTRACT

Cysteine cathepsins are primarily involved in the degradation and recycling of proteins in endo-lysosomal compartments but are also gaining recognition as pivotal proteolytic contributors to various immune functions. Through their extracellular proteolytic activities within the hematopoietic stem cell niche, they are involved in progenitor cell mobilization and differentiation. Cysteine cathepsins, such as cathepsins L and S contribute to antigen-induced adaptive immunity through major histocompatibility complex class II antigen presentation whereas cathepsin X regulates T-cell migration. By regulating toll-like receptor signaling and cytokine secretion cysteine cathepsins activate innate immune cells and affect their functional differentiation. Cathepsins C and H are expressed in cytotoxic T lymphocytes and natural killer cells and are involved in processing of pro-granzymes into proteolytically active forms. Cytoplasmic activities of cathepsins B and L contribute to the maintenance of homeostasis of the adaptive immune response by regulating cell death of T and B lymphocytes. The expression pattern, localization, and activity of cysteine cathepsins is tightly connected to their function in immune cells. Furthermore, cysteine cathepsins together with their endogenous inhibitors, serve as mediators in the interplay between cancer and immune cells that results in immune cell anergy. The aim of the present article is to review the mechanisms of dysregulation of cysteine cathepsins and their inhibitors in relation to immune dysfunction to address new possibilities for regulation of their function.


Subject(s)
Cell Differentiation/immunology , Cysteine Proteases/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Immunomodulation , Animals , Cell Differentiation/genetics , Clonal Anergy/immunology , Cysteine Proteases/chemistry , Cysteine Proteases/genetics , Cysteine Proteinase Inhibitors/pharmacology , Humans , Immune Tolerance , Immunomodulation/drug effects , Immunosenescence/drug effects , Multigene Family , Organogenesis/genetics , Organogenesis/immunology , Structure-Activity Relationship , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
7.
Cells ; 10(6)2021 06 17.
Article in English | MEDLINE | ID: mdl-34204485

ABSTRACT

The ability of T cells to identify foreign antigens and mount an efficient immune response while limiting activation upon recognition of self and self-associated peptides is critical. Multiple tolerance mechanisms work in concert to prevent the generation and activation of self-reactive T cells. T cell tolerance is tightly regulated, as defects in these processes can lead to devastating disease; a wide variety of autoimmune diseases and, more recently, adverse immune-related events associated with checkpoint blockade immunotherapy have been linked to a breakdown in T cell tolerance. The quantity and quality of antigen receptor signaling depend on a variety of parameters that include T cell receptor affinity and avidity for peptide. Autoreactive T cell fate choices (e.g., deletion, anergy, regulatory T cell development) are highly dependent on the strength of T cell receptor interactions with self-peptide. However, less is known about how differences in the strength of T cell receptor signaling during differentiation influences the 'function' and persistence of anergic and regulatory T cell populations. Here, we review the literature on this subject and discuss the clinical implications of how T cell receptor signal strength influences the 'quality' of anergic and regulatory T cell populations.


Subject(s)
Clonal Anergy/immunology , Self Tolerance/immunology , T-Lymphocytes/immunology , Animals , Cell Differentiation/immunology , Humans
8.
J Exp Med ; 218(7)2021 07 05.
Article in English | MEDLINE | ID: mdl-33974042

ABSTRACT

T cell anergy is an important peripheral tolerance mechanism. We studied how T cell anergy is established using an anergy model in which the Zap70 hypermorphic mutant W131A is coexpressed with the OTII TCR transgene (W131AOTII). Anergy was established in the periphery, not in the thymus. Contrary to enriched tolerance gene signatures and impaired TCR signaling in mature peripheral CD4 T cells, CD4SP thymocytes exhibited normal TCR signaling in W131AOTII mice. Importantly, the maintenance of T cell anergy in W131AOTII mice required antigen presentation via MHC-II. We investigated the functional importance of the inhibitory receptor PD-1 and the E3 ubiquitin ligases Cbl-b and Grail in this model. Deletion of each did not affect expression of phenotypic markers of anergic T cells or T reg numbers. However, deletion of Cbl-b, but not Grail or PD-1, in W131AOTII mice restored T cell responsiveness and signaling. Thus, Cbl-b plays an essential role in the establishment and/or maintenance of unresponsiveness in T cell anergy.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , CD4-Positive T-Lymphocytes/immunology , Proto-Oncogene Proteins c-cbl/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Clonal Anergy/immunology , Immune Tolerance/immunology , Lymphocyte Activation/immunology , Male , Mice , Mice, Transgenic , Peripheral Tolerance/immunology , Programmed Cell Death 1 Receptor/immunology , Signal Transduction/immunology , Ubiquitin-Protein Ligases/immunology , ZAP-70 Protein-Tyrosine Kinase/immunology
9.
Nat Commun ; 12(1): 2782, 2021 05 13.
Article in English | MEDLINE | ID: mdl-33986293

ABSTRACT

Chronic stimulation of CD8+ T cells triggers exhaustion, a distinct differentiation state with diminished effector function. Exhausted cells exist in multiple differentiation states, from stem-like progenitors that are the key mediators of the response to checkpoint blockade, through to terminally exhausted cells. Due to its clinical relevance, there is substantial interest in defining the pathways that control differentiation and maintenance of these subsets. Here, we show that chronic antigen induces the anergy-associated transcription factor EGR2 selectively within progenitor exhausted cells in both chronic LCMV and tumours. EGR2 enables terminal exhaustion and stabilizes the exhausted transcriptional state by both direct EGR2-dependent control of key exhaustion-associated genes, and indirect maintenance of the exhausted epigenetic state. We show that EGR2 is a regulator of exhaustion that epigenetically and transcriptionally maintains the differentiation competency of progenitor exhausted cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Clonal Anergy/immunology , Early Growth Response Protein 2/metabolism , Lymphopoiesis/physiology , Animals , Antigens/immunology , CD4-Positive T-Lymphocytes/immunology , Early Growth Response Protein 2/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Knockout
10.
Front Immunol ; 12: 600829, 2021.
Article in English | MEDLINE | ID: mdl-33717081

ABSTRACT

Understanding the mechanisms behind T cell dysfunctions during chronic diseases is critical in developing effective immunotherapies. As demonstrated by several animal models and human studies, T cell dysfunctions are induced during chronic diseases, spanning from infections to cancer. Although factors governing the onset and the extent of the functional impairment of T cells can differ during infections and cancer, most dysfunctional phenotypes share common phenotypic traits in their immune receptor and biophysical landscape. Through the latest developments in biophysical techniques applied to explore cell membrane and receptor-ligand dynamics, we are able to dissect and gain further insights into the driving mechanisms behind T cell dysfunctions. These insights may prove useful in developing immunotherapies aimed at reinvigorating our immune system to fight off infections and malignancies more effectively. The recent success with checkpoint inhibitors in treating cancer opens new avenues to develop more effective, targeted immunotherapies. Here, we highlight the studies focused on the transformation of the biophysical landscape during infections and cancer, and how T cell biomechanics shaped the immunopathology associated with chronic diseases.


Subject(s)
Biomechanical Phenomena , Disease Susceptibility , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Biomarkers , Cellular Microenvironment/immunology , Cellular Senescence/immunology , Chronic Disease , Clonal Anergy/immunology , Host-Pathogen Interactions/immunology , Humans , Immunological Synapses/immunology , Immunological Synapses/metabolism , Lymphocyte Count , Mechanotransduction, Cellular , Phenotype
11.
Mol Med Rep ; 23(4)2021 04.
Article in English | MEDLINE | ID: mdl-33649856

ABSTRACT

Semaphorin 3A (Sema3A), a member of the Sema family of proteins, appears to serve an important role in sepsis and sepsis­induced immunosuppression and has been regarded as a crucial regulator involved in cellular immune response. However, the role of Sema3A in CD4+ T cell anergy during sepsis remains to be elucidated. In the present study, the cecal ligation and perforation model and lipopolysaccharide (LPS) were used to simulate sepsis and the role of Sema3A in sepsis­induced CD4+ T cell anergy was investigated in vivo and in vitro. In vivo, the serum concentration of Sema3A was enhanced and exacerbated sepsis­induced T cell immunosuppression and multiple organ dysfunction syndromes (MODS). Administration of (­)­epigallocatechin­3­gallate, an inhibitor of Sema3A, markedly improved sepsis­induced T cell immunosuppression and MODS. In vitro, both lymphoid and myeloid lineages secreted high concentration of Sema3A in LPS­induced sepsis, especially in the lymphoid lineage. Inhibition of Sema3A alleviated T cell anergy. The NF­κB signaling pathway was involved in Sema3A­mediated autocrine loop aggravating T cell immune dysfunction during LPS­induced sepsis. Inhibiting Sema3A exerted significant improvement of sepsis­induced immunosuppression and MODS, which was associated with improvement of CD4+ T cells anergy via regulation of the NF­κB signaling pathway.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Immune Tolerance/immunology , Semaphorin-3A/immunology , Sepsis/immunology , Animals , Antioxidants/administration & dosage , CD4-Positive T-Lymphocytes/metabolism , Catechin/administration & dosage , Catechin/analogs & derivatives , Cells, Cultured , Clonal Anergy/immunology , Humans , Lipopolysaccharides/immunology , Male , Mice, Inbred C57BL , Multiple Organ Failure/immunology , Multiple Organ Failure/metabolism , Multiple Organ Failure/prevention & control , NF-kappa B/immunology , NF-kappa B/metabolism , Semaphorin-3A/antagonists & inhibitors , Semaphorin-3A/metabolism , Sepsis/metabolism , Signal Transduction/drug effects , Signal Transduction/immunology
12.
Cancer Res ; 81(7): 1788-1801, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33483371

ABSTRACT

CD1d-restricted invariant natural killer T cells (iNKT cells) mediate strong antitumor immunity when stimulated by glycolipid agonists. However, attempts to develop effective iNKT cell agonists for clinical applications have been thwarted by potential problems with dose-limiting toxicity and by activation-induced iNKT cell anergy, which limits the efficacy of repeated administration. To overcome these issues, we developed a unique bispecific T-cell engager (BiTE) based on covalent conjugates of soluble CD1d with photoreactive analogues of the glycolipid α-galactosylceramide. Here we characterize the in vivo activities of iNKT cell-specific BiTEs and assess their efficacy for cancer immunotherapy in mouse models using transplantable colorectal cancer or melanoma tumor lines engineered to express human Her2 as a tumor-associated antigen. Systemic administration of conjugated BiTEs stimulated multiple iNKT cell effector functions including cytokine release, secondary activation of NK cells, and induction of dendritic cell maturation and also initiated epitope spreading for tumor-specific CD8+ cytolytic T-cell responses. The antitumor effects of iNKT-cell activation with conjugated BiTEs were further enhanced by simultaneous checkpoint blockade with antibodies to CTLA-4, providing a potential approach for combination immunotherapy. Multiple injections of covalently stabilized iNKT cell-specific BiTEs activated iNKT cells without causing iNKT cell anergy or exhaustion, thus enabling repeated administration for effective and nontoxic cancer immunotherapy regimens. SIGNIFICANCE: Covalently stabilized conjugates that engage the antigen receptors of iNKT cells and target a tumor antigen activate potent antitumor immunity without induction of anergy or depletion of the responding iNKT cells.


Subject(s)
Antigens, CD1d/pharmacology , Clonal Anergy/drug effects , Galactosylceramides/pharmacology , Immunotherapy/methods , Natural Killer T-Cells/drug effects , Animals , Antigens, CD1d/chemistry , Antigens, CD1d/immunology , Clonal Anergy/immunology , Female , Galactosylceramides/chemistry , Humans , Immunoconjugates/pharmacology , Lymphocyte Activation/drug effects , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Melanoma, Experimental/therapy , Mice , Mice, Inbred C57BL , Mice, Knockout , Natural Killer T-Cells/immunology , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Skin Neoplasms/therapy , Tumor Cells, Cultured
14.
Nat Rev Immunol ; 21(4): 257-267, 2021 04.
Article in English | MEDLINE | ID: mdl-33077935

ABSTRACT

Following their exit from the thymus, T cells are endowed with potent effector functions but must spare host tissue from harm. The fate of these cells is dictated by a series of checkpoints that regulate the quality and magnitude of T cell-mediated immunity, known as tolerance checkpoints. In this Perspective, we discuss the mediators and networks that control the six main peripheral tolerance checkpoints throughout the life of a T cell: quiescence, ignorance, anergy, exhaustion, senescence and death. At the naive T cell stage, two intrinsic checkpoints that actively maintain tolerance are quiescence and ignorance. In the presence of co-stimulation-deficient T cell activation, anergy is a dominant hallmark that mandates T cell unresponsiveness. When T cells are successfully stimulated and reach the effector stage, exhaustion and senescence can limit excessive inflammation and prevent immunopathology. At every stage of the T cell's journey, cell death exists as a checkpoint to limit clonal expansion and to terminate unrestrained responses. Here, we compare and contrast the T cell tolerance checkpoints and discuss their specific roles, with the aim of providing an integrated view of T cell peripheral tolerance and fate regulation.


Subject(s)
Apoptosis/immunology , Cellular Senescence/immunology , Clonal Anergy/immunology , Immunologic Memory/immunology , Peripheral Tolerance/immunology , T-Lymphocytes/immunology , Cell Death/immunology , Humans
15.
Transplant Proc ; 53(1): 474-480, 2021.
Article in English | MEDLINE | ID: mdl-32919804

ABSTRACT

BACKGROUND: New strategies to inhibit acute rejection are needed for further applications of composite tissue allotransplantation. The nuclear receptor subfamily 4 group A member 1 (NR4A1) is considered a key controller of maintaining tolerance homeostasis. However, the effect of NR4A1 in suppressing rejection responses after allotransplantation remains unknown. METHODS: Brown Norway rat groin flaps were transplanted into Lewis rat recipients. The recipients were administrated cytosporone B, an NR4A1 activator. NR4A1 expression and graft survival time were assessed. T helper type 1 and regulatory T cell populations in the second lymphoid organ were detected by flow cytometry. Furthermore, a retrovirus containing NR4A1 was constructed and transfected to T cells in vitro. After stimulation, interleukin 2 and interferon gamma secretions were detected in the T cells. RESULTS: Administration of cytosporone B activated NR4A1 expression in allotransplant recipients and was associated with prolonged survival time of the vascularized free flap allograft. T helper type 1 cells in the recipient secondary lymphoid organs were decreased, whereas the population of regulatory T cells did not change. Interleukin 2 and interferon gamma were suppressed in vitro in the T cells overexpressing NR4A1. CONCLUSIONS: We demonstrated that the overexpressed NR4A1 is associated with suppressed graft rejection response. The suppression effect may attribute to induction of T-cell anergy and blockade of key immunologic cytokines.


Subject(s)
Clonal Anergy/immunology , Graft Survival/immunology , Lymphocyte Activation/immunology , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , T-Lymphocytes/immunology , Animals , Benzopyrans/pharmacology , Free Tissue Flaps/immunology , Free Tissue Flaps/transplantation , Graft Rejection/immunology , Lymphocyte Activation/drug effects , Rats , Rats, Inbred BN , Rats, Inbred Lew , Transplantation, Homologous , Vascularized Composite Allotransplantation
16.
Front Immunol ; 11: 567342, 2020.
Article in English | MEDLINE | ID: mdl-33363531

ABSTRACT

Sarcoidosis is a systemic inflammatory disease characterized by infiltration of immune cells into granulomas. Previous gene expression studies using heterogeneous cell mixtures lack insight into cell-type-specific immune dysregulation. We performed the first single-cell RNA-sequencing study of sarcoidosis in peripheral immune cells in 48 patients and controls. Following unbiased clustering, differentially expressed genes were identified for 18 cell types and bioinformatically assessed for function and pathway enrichment. Our results reveal persistent activation of circulating classical monocytes with subsequent upregulation of trafficking molecules. Specifically, classical monocytes upregulated distinct markers of activation including adhesion molecules, pattern recognition receptors, and chemokine receptors, as well as enrichment of immunoregulatory pathways HMGB1, mTOR, and ephrin receptor signaling. Predictive modeling implicated TGFß and mTOR signaling as drivers of persistent monocyte activation. Additionally, sarcoidosis T cell subsets displayed patterns of dysregulation. CD4 naïve T cells were enriched for markers of apoptosis and Th17/Treg differentiation, while effector T cells showed enrichment of anergy-related pathways. Differentially expressed genes in regulatory T cells suggested dysfunctional p53, cell death, and TNFR2 signaling. Using more sensitive technology and more precise units of measure, we identify cell-type specific, novel inflammatory and regulatory pathways. Based on our findings, we suggest a novel model involving four convergent arms of dysregulation: persistent hyperactivation of innate and adaptive immunity via classical monocytes and CD4 naïve T cells, regulatory T cell dysfunction, and effector T cell anergy. We further our understanding of the immunopathology of sarcoidosis and point to novel therapeutic targets.


Subject(s)
Gene Expression Profiling , Monocytes/immunology , Monocytes/metabolism , Sarcoidosis/etiology , Single-Cell Analysis , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Transcriptome , Apoptosis/genetics , Apoptosis/immunology , Biomarkers , Case-Control Studies , Cell Movement/genetics , Cell Movement/immunology , Clonal Anergy/genetics , Clonal Anergy/immunology , Disease Progression , Female , Humans , Immunophenotyping , Male , Models, Biological , Organ Specificity , Receptors, Antigen, T-Cell/metabolism , Sarcoidosis/diagnosis , Sarcoidosis/metabolism , Sarcoidosis/therapy , Signal Transduction
17.
Int J Mycobacteriol ; 9(3): 268-273, 2020.
Article in English | MEDLINE | ID: mdl-32862159

ABSTRACT

Background: Murine leprosy is a chronic granulomatous disease caused by Mycobacterium lepraemurium (MLM) in mice and rats. The disease evolves with the development of cellular anergy that impedes the production of interferon gamma (IFNγ), tumor necrosis factor-alpha (TNFα), and nitric oxide (NO) required to kill the microorganism. In this study we investigated whether histone deacetylase inhibitors (HDACi) (valproic acid and sodium butyrate [NaB]) and the immunomodulator transfer factor in dialyzable leukocyte extracts (DLE) can prevent anergy in murine leprosy. Methods: Five groups of six Balb/c mice were intraperitoneally inoculated with 2 × 107 MLM. Thirty-days post inoculation, treatment was started; one group received no treatment, one was treated with rifampicin-clofazimine (R-C), one with sodium valproate (VPA), one with NaB, and one with DLE. The animals were monitored for the evidence of disease for 96 days. After euthanasia, their spleens were removed and processed for histologic, bacteriologic, and cytokine studies. Results: R-C completely controlled the ongoing disease. DLE and NaB significantly reduced the development of lesions, including granuloma size and the number of bacilli; VPA was less effective. DLE, NaB, and VPA reverted the anergic condition in diverse grades and allowed the expression of IFNγ, TNFα, and inducible NO synthase, also in diverse grades. Conclusion: Anergy in leprosy and murine leprosy allows disease progression. In this study, anergy was prevented, in significant degree, by DLE (an immunomodulator) and NaB (HDACi). VPA was less effective. These results suggest potential beneficial effects of DLE and NaB in the ancillary treatment of leprosy.


Subject(s)
Butyric Acid/administration & dosage , Cell Extracts/pharmacology , Clonal Anergy/immunology , Histone Deacetylase Inhibitors/administration & dosage , Leprosy/immunology , Valproic Acid/administration & dosage , Animals , Cell Extracts/immunology , Dialysis , Female , Leukocytes/chemistry , Leukocytes/immunology , Mice , Mice, Inbred BALB C , Mycobacterium lepraemurium/drug effects , Mycobacterium lepraemurium/immunology
18.
RMD Open ; 6(2)2020 07.
Article in English | MEDLINE | ID: mdl-32675278

ABSTRACT

Systemic lupus erythematosus (SLE) is characterised by numerous abnormalities in B lineage cells, including increased CD27++ plasmablasts/plasma cells, atypical CD27-IgD- B cells with increased CD95, spleen tyrosine kinase (Syk)++, CXCR5- and CXCR5+ subsets and anergic CD11c+Tbet+ age-associated B cells. Most findings, together with preclinical lupus models, support the concept of B cell hyperactivity in SLE. However, it remains largely unknown whether these specific B cell subsets have pathogenic consequences and whether they provide relevant therapeutic targets. Recent findings indicate a global distortion of B cell functional capability, in which the entire repertoire of naïve and memory B cells in SLE exhibits an anergic or postactivated (APA) functional phenotype. The APA status of SLE B cells has some similarities to the functional derangement of lupus T cells. APA B cells are characterised by reduced global cytokine production, diminished B cell receptor (BCR) signalling with decreased Syk and Bruton's tyrosine kinase phosphorylation related to repeated in vivo BCR stimulation as well as hyporesponsiveness to toll-like receptor 9 engagement, but intact CD40 signalling. This APA status was related to constitutive co-localisation of CD22 linked to phosphatase SHP-1 and increased overall protein phosphatase activities. Notably, CD40 co-stimulation could revert this APA status and restore BCR signalling, downregulate protein tyrosine phosphatase transcription and promote B cell proliferation and differentiation. The APA status and their potential rescue by bystander help conveyed through CD40 stimulation not only provides insights into possible mechanisms of escape of autoreactive clones from negative selection but also into novel ways to target B cells therapeutically.


Subject(s)
B-Lymphocytes/immunology , Clonal Anergy/immunology , Lupus Erythematosus, Systemic/etiology , Lymphocyte Activation/immunology , Animals , B-Lymphocytes/metabolism , Biomarkers , CD40 Antigens/metabolism , Cell Differentiation/immunology , Disease Susceptibility , Gene Expression Profiling , Gene Expression Regulation , Humans , Lupus Erythematosus, Systemic/metabolism , Lupus Erythematosus, Systemic/therapy , Receptors, Antigen, B-Cell/metabolism , Signal Transduction , Toll-Like Receptor 9/metabolism
19.
Front Immunol ; 11: 1350, 2020.
Article in English | MEDLINE | ID: mdl-32714330

ABSTRACT

The main role of the human immune system is to eliminate cells presenting foreign antigens and abnormal patterns, while maintaining self-tolerance. However, when facing highly variable pathogens or antigens very similar to self-antigens, this system can fail in completely eliminating the anomalies, leading to the establishment of chronic pathologies. Prototypical examples of immune system defeat are cancer and Human Immunodeficiency Virus-1 (HIV-1) infection. In both conditions, the immune system is persistently exposed to antigens leading to systemic inflammation, lack of generation of long-term memory and exhaustion of effector cells. This triggers a negative feedback loop where effector cells are unable to resolve the pathology and cannot be replaced due to the lack of a pool of undifferentiated, self-renewing memory T cells. In addition, in an attempt to reduce tissue damage due to chronic inflammation, antigen presenting cells and myeloid components of the immune system activate systemic regulatory and tolerogenic programs. Beside these homologies shared between cancer and HIV-1 infection, the immune system can be shaped differently depending on the type and distribution of the eliciting antigens with ultimate consequences at the phenotypic and functional level of immune exhaustion. T cell differentiation, functionality, cytotoxic potential and proliferation reserve, immune-cell polarization, upregulation of negative regulators (immune checkpoint molecules) are indeed directly linked to the quantitative and qualitative differences in priming and recalling conditions. Better understanding of distinct mechanisms and functional consequences underlying disease-specific immune cell dysfunction will contribute to further improve and personalize immunotherapy. In the present review, we describe relevant players of immune cell exhaustion in cancer and HIV-1 infection, and enumerate the best-defined hallmarks of T cell dysfunction. Moreover, we highlight shared and divergent aspects of T cell exhaustion and T cell activation to the best of current knowledge.


Subject(s)
Clonal Anergy/immunology , HIV Infections/immunology , Neoplasms/immunology , T-Lymphocytes/immunology , Humans
20.
Eur J Immunol ; 50(11): 1838-1840, 2020 11.
Article in English | MEDLINE | ID: mdl-32627182

ABSTRACT

Recent studies have demonstrated that laboratory mice lack a robust repertoire of memory T cell. Administration of an anti-CD3ε activating antibody (clone 145-2C11) induces persistent CD4 and CD8 T cell memory in both lymphatic and solid organs while maintaining T cell responses and without increased anergy or altering innate immunity.


Subject(s)
Antibodies, Monoclonal/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Lymphocyte Activation/immunology , Animals , CD3 Complex/immunology , Clonal Anergy/immunology , Mice , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL
...