Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
Add more filters










Publication year range
1.
JCI Insight ; 7(11)2022 06 08.
Article in English | MEDLINE | ID: mdl-35503415

ABSTRACT

Positive regulatory domain 1 (PRDM1) encodes B lymphocyte-induced maturation protein 1 (BLIMP1), also known as a master regulator of T cell homeostasis. We observed a negative relationship between Blimp-1 and IL-21 based on our previous data that Blimp-1 overexpression in T cells suppresses autoimmune diabetes while Blimp-1-deficient T cells contribute to colitis in NOD mice. Reanalysis of published data sets also revealed an inverse correlation between PRDM1 and IL21 in Crohn's disease. Here, we illustrate that Blimp-1 repressed IL-21 by reducing chromatin accessibility and evicting an IL-21 activator, c-Maf, from the Il21 promoter. Moreover, Blimp-1 overexpression-mediated reduction in permissive chromatin structures at the Il21 promoter could override IL-21-accelerated autoimmune diabetogenesis in small ubiquitin-like modifier-defective c-Maf-transgenic mice. An autoregulatory feedback loop to harness IL-21 expression was unveiled by the evidence that IL-21 addition induced time-dependent Blimp-1 expression and subsequently enriched its binding to the Il21 promoter to suppress IL-21 overproduction. Furthermore, intervention of this feedback loop by IL-21 blockade, with IL-21R.Fc administration or IL-21 receptor deletion, attenuated Blimp-1 deficiency-mediated colitis and reinforced the circuit between Blimp-1 and IL-21 in the regulation of autoimmunity. We highlight the translation of Blimp-1-based epigenetic and transcriptomic profiles applicable to a personalized medicine approach in autoimmune diseases.


Subject(s)
Autoimmune Diseases , Colitis , Positive Regulatory Domain I-Binding Factor 1 , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Chromatin/immunology , Colitis/genetics , Colitis/immunology , Epigenesis, Genetic , Homeostasis , Mice , Mice, Inbred NOD , Positive Regulatory Domain I-Binding Factor 1/genetics , Positive Regulatory Domain I-Binding Factor 1/immunology
2.
Eur J Immunol ; 52(7): 1095-1111, 2022 07.
Article in English | MEDLINE | ID: mdl-35389518

ABSTRACT

Tissue-resident memory T cells (Trm) are retained in peripheral tissues after infection for enhanced protection against secondary encounter with the same pathogen. We have previously shown that the transcription factor Hobit and its homolog Blimp-1 drive Trm development after viral infection, but how and when these transcription factors mediate Trm formation remains poorly understood. In particular, the major impact of Blimp-1 in regulating several aspects of effector T-cell differentiation impairs study of its specific role in Trm development. Here, we used the restricted expression of Hobit in the Trm lineage to develop mice with a conditional deletion of Blimp-1 in Trm, allowing us to specifically investigate the role of both transcription factors in Trm differentiation. We found that Hobit and Blimp-1 were required for the upregulation of CD69 and suppression of CCR7 and S1PR1 on virus-specific Trm precursors after LCMV infection, underlining a role in their retention within tissues. The early impact of Hobit and Blimp-1 favored Trm formation and prevented the development of circulating memory T cells. Thus, our findings highlight a role of Hobit and Blimp-1 at the branching point of circulating and resident memory lineages by suppressing tissue egress of Trm precursors early during infection.


Subject(s)
CD8-Positive T-Lymphocytes , Immunologic Memory , Lymphocytic Choriomeningitis , Lymphocytic choriomeningitis virus , Positive Regulatory Domain I-Binding Factor 1 , Transcription Factors , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/pathology , Lymphocytic choriomeningitis virus/immunology , Mice , Positive Regulatory Domain I-Binding Factor 1/immunology , Transcription Factors/genetics , Transcription Factors/immunology , Transcription Factors/metabolism
3.
J Exp Med ; 219(1)2022 01 03.
Article in English | MEDLINE | ID: mdl-34792530

ABSTRACT

During the immune response, CD4+ T cells differentiate into distinct effector subtypes, including follicular helper T (Tfh) cells that help B cells, and into memory cells. Tfh and memory cells are required for long-term immunity; both depend on the transcription factor Bcl6, raising the question whether they differentiate through similar mechanisms. Here, using single-cell RNA and ATAC sequencing, we show that virus-responding CD4+ T cells lacking both Bcl6 and Blimp1 can differentiate into cells with transcriptomic, chromatin accessibility, and functional attributes of memory cells but not of Tfh cells. Thus, Bcl6 promotes memory cell differentiation primarily through its repression of Blimp1. These findings demonstrate that distinct mechanisms underpin the differentiation of memory and Tfh CD4+ cells and define the Bcl6-Blimp1 axis as a potential target for promoting long-term memory T cell differentiation.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Memory T Cells/immunology , Positive Regulatory Domain I-Binding Factor 1/immunology , Proto-Oncogene Proteins c-bcl-6/immunology , T Follicular Helper Cells/immunology , Animals , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation/genetics , Cells, Cultured , Chromatin Immunoprecipitation Sequencing/methods , Gene Expression Profiling/methods , Memory T Cells/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Positive Regulatory Domain I-Binding Factor 1/genetics , Positive Regulatory Domain I-Binding Factor 1/metabolism , Proto-Oncogene Proteins c-bcl-6/genetics , Proto-Oncogene Proteins c-bcl-6/metabolism , RNA-Seq/methods , Single-Cell Analysis/methods , T Follicular Helper Cells/metabolism
4.
Nat Commun ; 12(1): 2710, 2021 05 11.
Article in English | MEDLINE | ID: mdl-33976194

ABSTRACT

Treg cells are critical regulators of immune homeostasis, and environment-driven Treg cell differentiation into effector (e)Treg cells is crucial for optimal functioning. However, human Treg cell programming in inflammation is unclear. Here, we combine transcriptional and epigenetic profiling to identify a human eTreg cell signature. Inflammation-derived functional Treg cells have a transcriptional profile characterized by upregulation of both a core Treg cell (FOXP3, CTLA4, TIGIT) and effector program (GITR, BLIMP-1, BATF). We identify a specific human eTreg cell signature that includes the vitamin D receptor (VDR) as a predicted regulator in eTreg cell differentiation. H3K27ac/H3K4me1 occupancy indicates an altered (super-)enhancer landscape, including enrichment of the VDR and BATF binding motifs. The Treg cell profile has striking overlap with tumor-infiltrating Treg cells. Our data demonstrate that human inflammation-derived Treg cells acquire a conserved and specific eTreg cell profile guided by epigenetic changes, and fine-tuned by environment-specific adaptations.


Subject(s)
Arthritis, Juvenile/genetics , Epigenesis, Genetic , Receptors, Calcitriol/genetics , T-Lymphocytes, Regulatory/immunology , Transcriptome , Adolescent , Arthritis, Juvenile/immunology , Arthritis, Juvenile/pathology , Base Sequence , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/immunology , CTLA-4 Antigen/genetics , CTLA-4 Antigen/immunology , Case-Control Studies , Cell Differentiation , Child , Child, Preschool , Female , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Gene Expression Profiling , Gene Regulatory Networks , Glucocorticoid-Induced TNFR-Related Protein/genetics , Glucocorticoid-Induced TNFR-Related Protein/immunology , Histones/genetics , Histones/immunology , Humans , Joints/immunology , Joints/pathology , Male , Metabolic Networks and Pathways/genetics , Metabolic Networks and Pathways/immunology , Positive Regulatory Domain I-Binding Factor 1/genetics , Positive Regulatory Domain I-Binding Factor 1/immunology , Primary Cell Culture , Receptors, Calcitriol/immunology , Receptors, Immunologic/genetics , Receptors, Immunologic/immunology , T-Lymphocytes, Regulatory/pathology , Young Adult
5.
Int Immunopharmacol ; 95: 107510, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33706054

ABSTRACT

Diabetic coronary heart disease (DM-CHD) poses a major threat to the world. The newly described T cell subset-Th9 cells and related cytokine interleukin (IL)-9 play important roles in the pathogenesis of diabetes and atherosclerosis. B lymphocyte-induced maturation protein 1 (Blimp-1) has been indicated to negatively regulate Th9 development in allergic asthma, but its role in DM-CHD remains unclear. Hence, this study was designed to investigate the role of Blimp-1 in DM-CHD and to elucidate whether the mechanism was associated with regulation of Th9 cell differentiation. Our results showed that serum Blimp-1 mRNA level was decreased whereas proportion of Th9 cells (IL-9+ CD4+ T cells) and serum level of Th9-related IL-9 were increased in DM-CHD patients. Furthermore, serum Blimp-1 mRNA level was negatively correlated with IL-9 level in DM-CHD patients. Importantly, administration of lentiviruses expressing Blimp-1 (LV-Blimp-1) significantly inhibited Th9 cell differentiation and alleviated the severity of atherosclerotic lesions in the aorta and coronary artery, dyslipidemia, inflammation, vascular endothelial dysfunction, and oxidative stress in DM-CHD model rats. Collectively, Blimp-1 exerts a protective effect in DM-CHD rats and the mechanism might involve inhibition of Th9 cell differentiation.


Subject(s)
Coronary Disease/immunology , Diabetes Mellitus, Type 2/immunology , Positive Regulatory Domain I-Binding Factor 1/immunology , T-Lymphocyte Subsets/immunology , Adult , Aged , Animals , Cell Differentiation , Female , Humans , Interleukin-9/immunology , Male , Middle Aged , Positive Regulatory Domain I-Binding Factor 1/genetics , Rats, Sprague-Dawley
6.
FEBS J ; 288(24): 7123-7142, 2021 12.
Article in English | MEDLINE | ID: mdl-33590946

ABSTRACT

The adaptive immune system has the enormous challenge to protect the host through the generation and differentiation of pathogen-specific short-lived effector T cells while in parallel developing long-lived memory cells to control future encounters with the same pathogen. A complex regulatory network is needed to preserve a population of naïve cells over lifetime that exhibit sufficient diversity of antigen receptors to respond to new antigens, while also sustaining immune memory. In parallel, cells need to maintain their proliferative potential and the plasticity to differentiate into different functional lineages. Initial signs of waning immune competence emerge after 50 years of age, with increasing clinical relevance in the 7th-10th decade of life. Morbidity and mortality from infections increase, as drastically exemplified by the current COVID-19 pandemic. Many vaccines, such as for the influenza virus, are poorly effective to generate protective immunity in older individuals. Age-associated changes occur at the level of the T-cell population as well as the functionality of its cellular constituents. The system highly relies on the self-renewal of naïve and memory T cells, which is robust but eventually fails. Genetic and epigenetic modifications contribute to functional differences in responsiveness and differentiation potential. To some extent, these changes arise from defective maintenance; to some, they represent successful, but not universally beneficial adaptations to the aging host. Interventions that can compensate for the age-related defects and improve immune responses in older adults are increasingly within reach.


Subject(s)
Aging/immunology , COVID-19/immunology , Memory T Cells/immunology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Regulatory/immunology , Adaptive Immunity , Aged , Aging/genetics , COVID-19/genetics , COVID-19/pathology , COVID-19/virology , Cell Differentiation , Cell Proliferation , Dual Specificity Phosphatase 6/genetics , Dual Specificity Phosphatase 6/immunology , Gene Expression Regulation , Humans , Memory T Cells/virology , MicroRNAs/genetics , MicroRNAs/immunology , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/immunology , Positive Regulatory Domain I-Binding Factor 1/genetics , Positive Regulatory Domain I-Binding Factor 1/immunology , SARS-CoV-2/immunology , SARS-CoV-2/pathogenicity , T-Lymphocytes, Cytotoxic/virology , T-Lymphocytes, Helper-Inducer/virology , T-Lymphocytes, Regulatory/virology
7.
J Neuroinflammation ; 18(1): 27, 2021 Jan 19.
Article in English | MEDLINE | ID: mdl-33468194

ABSTRACT

BACKGROUND: Follicular regulatory T (TFR) cells are essential for the regulation of germinal center (GC) response and humoral self-tolerance. Dysregulated follicular helper T (TFH) cell-GC-antibody (Ab) response secondary to dysfunctional TFR cells is the root of an array of autoimmune disorders. The contribution of TFR cells to the pathogenesis of multiple sclerosis (MS) and murine experimental autoimmune encephalomyelitis (EAE) remains largely unclear. METHODS: To determine the impact of dysregulated regulatory T cells (Tregs), TFR cells, and Ab responses on EAE, we compared the MOG-induced EAE in mice with a FoxP3-specific ablation of the transcription factor Blimp1 to control mice. In vitro co-culture assays were used to understand how Tregs and Ab regulate the activity of microglia and central nervous system (CNS)-infiltrating myeloid cells. RESULTS: Mice with a FoxP3-specific deletion of Blimp1 developed severe EAE and failed to recover compared to control mice, reflecting conversion of Tregs into interleukin (IL)-17A/granulocyte-macrophage colony-stimulating factor (GM-CSF)-producing effector T cells associated with increased TFH-Ab responses, more IgE deposition in the CNS, and inability to regulate CNS CD11b+ myeloid cells. Notably, serum IgE titers were positively correlated with EAE scores, and culture of CNS CD11b+ cells with sera from these EAE mice enhanced their activation, while transfer of Blimp1-deficient TFR cells promoted Ab production, activation of CNS CD11b+ cells, and EAE. CONCLUSIONS: Blimp1 is essential for the maintenance of TFR cells and Ab responses in EAE. Dysregulated TFR cells and Ab responses promote CNS autoimmunity.


Subject(s)
Antibody Formation/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Positive Regulatory Domain I-Binding Factor 1/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Autoimmunity/immunology , Cell Differentiation/immunology , Germinal Center , Mice , Mice, Inbred C57BL
8.
Front Immunol ; 12: 791100, 2021.
Article in English | MEDLINE | ID: mdl-35069572

ABSTRACT

CD4+CXCR5+Foxp3+ T-follicular regulatory (TFR) cells control the germinal center responses. Like T-follicular helper cells, they express high levels of Nuclear Factor of Activated T-cells c1, predominantly its short isoform NFATc1/αA. Ablation of NFATc1 in Tregs prevents upregulation of CXCR5 and migration of TFR cells into B-cell follicles. By contrast, constitutive active NFATc1/αA defines the surface density of CXCR5, whose level determines how deep a TFR migrates into the GC and how effectively it controls antibody production. As one type of effector Treg, TFR cells express B lymphocyte-induced maturation protein-1 (Blimp-1). Blimp-1 can directly repress Cxcr5 and NFATc1/αA is necessary to overcome this Blimp-1-mediated repression. Interestingly, Blimp-1 even reinforces the recruitment of NFATc1 to Cxcr5 by protein-protein interaction and by those means cooperates with NFATc1 for Cxcr5 transactivation. On the contrary, Blimp-1 is necessary to counterbalance NFATc1/αA and preserve the Treg identity. This is because although NFATc1/αA strengthens the follicular development of Tregs, it bears the inherent risk of causing an ex-Treg phenotype.


Subject(s)
Cell Movement/immunology , Germinal Center/immunology , NFATC Transcription Factors/immunology , Positive Regulatory Domain I-Binding Factor 1/immunology , Animals , Cell Movement/genetics , Mice , Mice, Transgenic , NFATC Transcription Factors/genetics , Positive Regulatory Domain I-Binding Factor 1/genetics
9.
J Allergy Clin Immunol ; 147(4): 1281-1295.e5, 2021 04.
Article in English | MEDLINE | ID: mdl-32905799

ABSTRACT

BACKGROUND: Group 2 innate lymphoid cells (ILC2s) are the dominant innate lymphoid cell population in the lungs at steady state, and their release of type 2 cytokines is a central driver in responding eosinophil infiltration and increased airway hyperreactivity. Our laboratory has identified a unique subset of ILC2s in the lungs that actively produce IL-10 (ILC210s). OBJECTIVE: Our aim was to characterize the effector functions of ILC210s in the development and pathology of allergic asthma. METHODS: IL-4-stimulated ILC210s were isolated to evaluate cytokine secretion, transcription factor signaling, metabolic dependence, and effector functions in vitro. ILC210s were also adoptively transferred into Rag2-/-γc-/- mice, which were then challenged with IL-33 and assessed for airway hyperreactivity and lung inflammation. RESULTS: We have determined that the transcription factors cMaf and Blimp-1 regulate IL-10 expression in ILC210s. Strikingly, our results demonstrate that ILC210s can utilize both autocrine and paracrine signaling to suppress proinflammatory ILC2 effector functions in vitro. Further, this subset dampens airway hyperreactivity and significantly reduces lung inflammation in vivo. Interestingly, ILC210s demonstrated a metabolic dependency on the glycolytic pathway for IL-10 production, shifting from the fatty acid oxidation pathway conventionally utilized for proinflammatory effector functions. CONCLUSION: These findings provide an important and previously unrecognized role of ILC210s in diseases associated with ILC2s such as allergic lung inflammation and asthma. They also provide new insights into the metabolism dependency of proinflammatory and anti-inflammatory ILC2 phenotypes.


Subject(s)
Asthma/immunology , Bronchial Hyperreactivity/immunology , Interleukin-10/immunology , Lymphocytes/immunology , Positive Regulatory Domain I-Binding Factor 1/immunology , Proto-Oncogene Proteins c-maf/immunology , Animals , Bronchoalveolar Lavage Fluid/immunology , Female , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic
10.
J Exp Med ; 218(1)2021 01 04.
Article in English | MEDLINE | ID: mdl-32986812

ABSTRACT

Posttranslational modification with SUMO is known to regulate the activity of transcription factors, but how SUMOylation of individual proteins might influence immunity is largely unexplored. The NFAT transcription factors play an essential role in antigen receptor-mediated gene regulation. SUMOylation of NFATc1 represses IL-2 in vitro, but its role in T cell-mediated immune responses in vivo is unclear. To this end, we generated a novel transgenic mouse in which SUMO modification of NFATc1 is prevented. Avoidance of NFATc1 SUMOylation ameliorated experimental autoimmune encephalomyelitis as well as graft-versus-host disease. Elevated IL-2 production in T cells promoted T reg expansion and suppressed autoreactive or alloreactive immune responses. Mechanistically, increased IL-2 secretion counteracted IL-17 and IFN-γ expression through STAT5 and Blimp-1 induction. Then, Blimp-1 repressed IL-2 itself, as well as the induced, proliferation-associated survival factor Bcl2A1. Collectively, these data demonstrate that prevention of NFATc1 SUMOylation fine-tunes T cell responses toward lasting tolerance. Thus, targeting NFATc1 SUMOylation presents a novel and promising strategy to treat T cell-mediated inflammatory diseases.


Subject(s)
Autoimmunity , Encephalomyelitis, Autoimmune, Experimental/immunology , NFATC Transcription Factors/immunology , Sumoylation/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cytokines/genetics , Cytokines/immunology , Encephalomyelitis, Autoimmune, Experimental/genetics , Mice , Mice, Knockout , Minor Histocompatibility Antigens/genetics , Minor Histocompatibility Antigens/immunology , NFATC Transcription Factors/genetics , Positive Regulatory Domain I-Binding Factor 1/genetics , Positive Regulatory Domain I-Binding Factor 1/immunology , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/immunology , STAT5 Transcription Factor/genetics , STAT5 Transcription Factor/immunology , Sumoylation/genetics
11.
Biochem Biophys Res Commun ; 533(3): 282-288, 2020 12 10.
Article in English | MEDLINE | ID: mdl-32958255

ABSTRACT

Dendritic cells (DC) are the most important antigen-presenting cells, which guide T cell activation and function, and dysregulated DC function might be one of the crucial causes of inflammatory bowel disease (IBD). It has been well-known that microbiota and their metabolites play an essential role in regulating the biology and function of DC, thus contributing to the pathogenesis of IBD. However, the underlying mechanisms remain largely unknown. Amphiregulin (AREG), a molecule of the epidermal growth factor (EGF) family, is primarily described as an epithelial cell-derived cytokine and recognized as a critical regulator of cell proliferation and tissue repair. Here, we found that DC expression of AREG depended on butyrate (a microbiota-derived short chained fatty acid), which required the interaction between butyrate and G-protein-coupled receptor 43 (GPR43). Furthermore, we found that butyrate-GPR43 interaction failed to induce AREG expression in DC deficient in B lymphocyte induced maturation protein 1 (Blimp-1). Notably, DC-derived AREG was indispensable for the protection against experimental colitis in mice. Additionally, AREG expression was significantly decreased in DC from IBD patients. Our data provide novel evidences to interpret how AREG expression is regulated in DC, and shed new light on the mechanisms whereby microbiota regulate DC function.


Subject(s)
Amphiregulin/genetics , Butyrates/immunology , Colitis, Ulcerative/genetics , Crohn Disease/genetics , Dendritic Cells/immunology , Positive Regulatory Domain I-Binding Factor 1/genetics , Receptors, Cell Surface/genetics , Amphiregulin/immunology , Animals , B-Lymphocytes/immunology , B-Lymphocytes/microbiology , B-Lymphocytes/pathology , Butyrates/metabolism , Butyrates/pharmacology , Case-Control Studies , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/immunology , Colitis, Ulcerative/microbiology , Crohn Disease/immunology , Crohn Disease/microbiology , Crohn Disease/pathology , Dendritic Cells/microbiology , Dendritic Cells/pathology , Female , Gastrointestinal Microbiome/immunology , Gene Expression Regulation , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pancreatitis-Associated Proteins/deficiency , Pancreatitis-Associated Proteins/genetics , Pancreatitis-Associated Proteins/immunology , Positive Regulatory Domain I-Binding Factor 1/immunology , Receptors, Cell Surface/immunology , Receptors, G-Protein-Coupled/deficiency , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/immunology , Signal Transduction , Sodium Dodecyl Sulfate/administration & dosage , Zonula Occludens-1 Protein/genetics , Zonula Occludens-1 Protein/immunology
12.
Front Immunol ; 11: 1085, 2020.
Article in English | MEDLINE | ID: mdl-32655552

ABSTRACT

In many infectious diseases, the immune response operates as a double-edged sword. While required for protective immunity, infection-induced inflammation can be detrimental if it is not properly controlled, causing collateral body damage and potentially leading to death. It is in this context that the potent anti-inflammatory cytokine interleukin-10 (IL-10) is required to dampen the pro-inflammatory immune response that hallmarks trypanosomosis. Effective control of this infection requires not just the action of antibodies specific for the parasite's variable surface glycoprotein (VSG) coat antigens, but also a pro-inflammatory immune response mediated mainly by IFNγ, TNF, and NO. However, strict control of inflammation is mandatory, as IL-10-deficient mice succumb from an unrestrained cytokine storm within 10 days of a Trypanosome brucei infection. The relevant cellular source of IL-10 and the associated molecular mechanisms implicated in its trypanosomosis associated production are poorly understood. Using an IL-10 reporter mouse strain (Vert-X), we demonstrate here that NK cells, CD8+ T cells and CD4+ T cells as well as B cells and plasma cells constitute potential cellular sources of IL-10 within the spleen and liver during acute infection. The IL-10 wave follows peak pro-inflammatory cytokine production, which accompanied the control of peak parasitemia. Similar results were observed following conventional experimental needle infection and physiological infections via T. brucei-infected tsetse flies. Our results show that conditional T cell-specific ablation of the IL-10 regulating Prdm1 gene (encoding for the Blimp-1 transcription factor), leads to an uncontrolled trypanosome-induced pro-inflammatory syndrome like the one observed in infected IL-10-deficient mice. This result indicates that the biological role of IL-10-derived from non-T cells, including NK cells, is of minor importance when considering host survival. The cytokine IL-27 that is also considered to be an IL-10 regulator, did not affect IL-10 production during infection. Together, these data suggest that T. brucei activates a Blimp-1-dependent IL-10 regulatory pathway in T cells that acts as a critical anti-inflammatory rheostat, mandatory for host survival during the acute phase of parasitemia.


Subject(s)
Cytokine Release Syndrome/prevention & control , Interleukin-10/biosynthesis , Positive Regulatory Domain I-Binding Factor 1/immunology , T-Lymphocytes/immunology , Trypanosoma brucei brucei , Trypanosomiasis, African/immunology , Animals , Cytokine Release Syndrome/etiology , Cytokine Release Syndrome/immunology , Disease Models, Animal , Female , Inflammation/etiology , Inflammation/immunology , Inflammation/prevention & control , Insect Vectors/parasitology , Interleukin-10/deficiency , Interleukin-10/genetics , Interleukins/antagonists & inhibitors , Interleukins/deficiency , Interleukins/immunology , Liver/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Positive Regulatory Domain I-Binding Factor 1/deficiency , Positive Regulatory Domain I-Binding Factor 1/genetics , Spleen/immunology , Trypanosomiasis, African/complications , Trypanosomiasis, African/parasitology , Tsetse Flies/parasitology
13.
J Leukoc Biol ; 108(6): 1765-1776, 2020 12.
Article in English | MEDLINE | ID: mdl-32421907

ABSTRACT

Extracellular vesicles (EVs) are small, double-membrane vesicles derived from erythrocytes, leukocytes, platelets, and cells of multiple tissues under physiologic or pathologic conditions. The role of EVs in stored RBC units is of great interest with respect to transfusion-related immunomodulation. The current study focuses on the quantity of EVs isolated from stored RBC units and their action on B cell-mediated immune responses. The in vitro experiment demonstrated that EVs exhibited a negative role in B cell survival, plasmacytic differentiation, and class switch recombination under LPS stimulation. Furthermore, LPS-induced antibody production was significantly decreased after EVs injection in vivo. Biochemical analysis revealed that EVs hampered the expression of Blimp-1 and IRF4 and the activation of NF-κB pathway in LPS-primed B cells. Overall, these data imply a vital role for EVs isolated from RBC units in B cell-mediated immune responses.


Subject(s)
Blood Preservation , Cell Differentiation/immunology , Erythrocytes/immunology , Extracellular Vesicles/immunology , Plasma Cells/immunology , Animals , Cell Differentiation/drug effects , Interferon Regulatory Factors/immunology , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred NOD , Mice, SCID , Positive Regulatory Domain I-Binding Factor 1/immunology
14.
Immunity ; 52(5): 808-824.e7, 2020 05 19.
Article in English | MEDLINE | ID: mdl-32433949

ABSTRACT

Tissue-resident memory CD8+ T cells (Trm) provide host protection through continuous surveillance of non-lymphoid tissues. Using single-cell RNA-sequencing (scRNA-seq) and genetic reporter mice, we identified discrete lineages of intestinal antigen-specific CD8+ T cells, including a Blimp1hiId3lo tissue-resident effector cell population most prominent in the early phase of acute viral and bacterial infections and a molecularly distinct Blimp1loId3hi tissue-resident memory population that subsequently accumulated at later infection time points. These Trm populations exhibited distinct cytokine production, secondary memory potential, and transcriptional programs including differential roles for transcriptional regulators Blimp1, T-bet, Id2, and Id3 in supporting and maintaining intestinal Trm. Extending our analysis to malignant tissue, we also identified discrete populations of effector-like and memory-like CD8+ T cell populations with tissue-resident gene-expression signatures that shared features of terminally exhausted and progenitor-exhausted T cells, respectively. Our findings provide insight into the development and functional heterogeneity of Trm cells, which has implications for enhancing vaccination and immunotherapy approaches.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Neoplasms/therapy , Animals , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/virology , Cells, Cultured , Immunotherapy/methods , Inhibitor of Differentiation Protein 2/genetics , Inhibitor of Differentiation Protein 2/immunology , Inhibitor of Differentiation Protein 2/metabolism , Inhibitor of Differentiation Proteins/genetics , Inhibitor of Differentiation Proteins/immunology , Inhibitor of Differentiation Proteins/metabolism , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/physiology , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neoplasms/immunology , Positive Regulatory Domain I-Binding Factor 1/genetics , Positive Regulatory Domain I-Binding Factor 1/immunology , Positive Regulatory Domain I-Binding Factor 1/metabolism
15.
Biochem Biophys Res Commun ; 527(4): 902-908, 2020 07 05.
Article in English | MEDLINE | ID: mdl-32430179

ABSTRACT

Inflammatory bowel disease (IBD) is a complex inflammatory disorder of the digestive tract with dysregulated innate and adaptive immune responses. Dendritic cells (DC), the most important antigen presenting cells, act as bridges connecting the adaptive and innate immune systems, and play a crucial role in the regulation of local homeostasis in the gut and are also essential mediators in the initiation and development of intestinal inflammation. Our recent study found that sauchinone (SAU) was able to ameliorate experimental colitis in mice by restraining Th17 cell differentiation and their pathogenicity. Here, we found that SAU significantly inhibited LPS-induced DC activation. Moreover, SAU suppressed the ability of LPS-primed DC to induce Th1/Th17 cell differentiation, but SAU-treated DC up-regulated their ability to initiate Foxp3+ Treg cell generation. Of note, we found that genetical ablation of Blimp-1 in DC markedly abrogated the SAU suppression of pro-inflammatory cytokine or promote immunomodulatory molecule production by DC. Blimp-1 deficiency boosted the ability of DC to polarize naïve CD4+ T cells into Th1/Th17 cell lineages. SAU failed to alleviated DSS-induced colitis in mice with Blimp-1-deficient DC. Our results shed new lights on the mechanisms of how SAU regulates DC biology and intestinal inflammation.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Benzopyrans/therapeutic use , Colitis/drug therapy , Dendritic Cells/drug effects , Dioxoles/therapeutic use , Inflammation/drug therapy , Positive Regulatory Domain I-Binding Factor 1/immunology , Animals , Anti-Inflammatory Agents/pharmacology , Benzopyrans/pharmacology , Colitis/chemically induced , Colitis/immunology , Dendritic Cells/immunology , Dextran Sulfate , Dioxoles/pharmacology , Inflammation/immunology , Male , Mice, Inbred C57BL , Th17 Cells/drug effects , Th17 Cells/immunology
16.
Eur J Immunol ; 50(6): 809-821, 2020 06.
Article in English | MEDLINE | ID: mdl-32022930

ABSTRACT

Terminal differentiation of B cells into antibody-secreting cells is the foundation of humoral immune response. B-1 cells, which are different from B-2 cells, preferentially differentiate into plasma cells. CMTM7 is a MARVEL-domain-containing membrane protein predominantly expressed in B cells that plays an important role in B-1a cell development. The present study assessed CMTM7 function in response to antigen stimulation. Following immunization with T cell-dependent and T cell-independent antigens, Cmtm7-deficient mice exhibited decreased IgM but normal IgG responses in vivo. In vitro stimulation with LPSs induced Cmtm7-/- B-1 cell activation, whereas proliferation was marginally reduced. Notably, Cmtm7 deficiency markedly suppressed plasma cell differentiation in response to TLR agonists, accompanied by a decrease in IgM and IL-10 production. At the molecular level, loss of Cmtm7 repressed the downregulation of Pax5 and the upregulation of Xbp1, Irf4, and Prdm1. Furthermore, p38 phosphorylation was inhibited in Cmtm7-/- B-1 cells. Experiments using a p38 inhibitor revealed that p38 activation was essential for the terminal differentiation of B-1 cells, suggesting that Cmtm7 contributes to B-1 cell differentiation by maintaining p38 activation. Overall, the data reveal the crucial functions of CMTM7 in TLR-induced terminal differentiation and p38 activation in B-1 cells.


Subject(s)
B-Lymphocyte Subsets/immunology , Cell Differentiation/immunology , Chemokines/immunology , MAP Kinase Signaling System/immunology , MARVEL Domain-Containing Proteins/immunology , Plasma Cells/immunology , Toll-Like Receptors/immunology , p38 Mitogen-Activated Protein Kinases/immunology , Animals , B-Lymphocyte Subsets/cytology , Cell Differentiation/genetics , Chemokines/genetics , Enzyme Activation/genetics , Enzyme Activation/immunology , Immunoglobulin M/genetics , Immunoglobulin M/immunology , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/immunology , Interleukin-10/genetics , Interleukin-10/immunology , MAP Kinase Signaling System/genetics , MARVEL Domain-Containing Proteins/genetics , Mice , Mice, Knockout , Plasma Cells/cytology , Positive Regulatory Domain I-Binding Factor 1/genetics , Positive Regulatory Domain I-Binding Factor 1/immunology , Toll-Like Receptors/genetics , p38 Mitogen-Activated Protein Kinases/genetics
17.
Nat Commun ; 11(1): 60, 2020 01 02.
Article in English | MEDLINE | ID: mdl-31896754

ABSTRACT

Short-chain fatty acids (SCFAs) butyrate and propionate are metabolites from dietary fiber's fermentation by gut microbiota that can affect differentiation or functions of T cells, macrophages and dendritic cells. We show here that at low doses these SCFAs directly impact B cell intrinsic functions to moderately enhance class-switch DNA recombination (CSR), while decreasing at higher doses over a broad physiological range, AID and Blimp1 expression, CSR, somatic hypermutation and plasma cell differentiation. In human and mouse B cells, butyrate and propionate decrease B cell Aicda and Prdm1 by upregulating select miRNAs that target Aicda and Prdm1 mRNA-3'UTRs through inhibition of histone deacetylation (HDAC) of those miRNA host genes. By acting as HDAC inhibitors, not as energy substrates or through GPR-engagement signaling in these B cell-intrinsic processes, these SCFAs impair intestinal and systemic T-dependent and T-independent antibody responses. Their epigenetic impact on B cells extends to inhibition of autoantibody production and autoimmunity in mouse lupus models.


Subject(s)
Antibodies/genetics , Epigenesis, Genetic/drug effects , Fatty Acids, Volatile/pharmacology , Gastrointestinal Microbiome/immunology , Animals , Antibodies/immunology , Antibodies/metabolism , Autoantibodies/genetics , Autoantibodies/immunology , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Butyrates/pharmacology , Cytidine Deaminase/antagonists & inhibitors , Cytidine Deaminase/genetics , Cytidine Deaminase/immunology , Cytidine Deaminase/metabolism , Dietary Fiber , Fatty Acids, Volatile/isolation & purification , Fatty Acids, Volatile/pharmacokinetics , Female , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/genetics , Histone Deacetylase Inhibitors/immunology , Histone Deacetylase Inhibitors/pharmacology , Humans , Lupus Erythematosus, Systemic/drug therapy , Lupus Erythematosus, Systemic/immunology , Mice, Inbred C57BL , Mice, Mutant Strains , Positive Regulatory Domain I-Binding Factor 1/antagonists & inhibitors , Positive Regulatory Domain I-Binding Factor 1/genetics , Positive Regulatory Domain I-Binding Factor 1/immunology , Positive Regulatory Domain I-Binding Factor 1/metabolism , Propionates/pharmacology , Tissue Distribution
18.
Front Immunol ; 11: 599647, 2020.
Article in English | MEDLINE | ID: mdl-33469456

ABSTRACT

Germinal centers (GC) are sites for extensive B cell proliferation and homeostasis is maintained by programmed cell death. The complement regulatory protein Decay Accelerating Factor (DAF) blocks complement deposition on host cells and therefore also phagocytosis of cells. Here, we show that B cells downregulate DAF upon BCR engagement and that T cell-dependent stimuli preferentially led to activation of DAFlo B cells. Consistent with this, a majority of light and dark zone GC B cells were DAFlo and susceptible to complement-dependent phagocytosis, as compared with DAFhi GC B cells. We could also show that the DAFhi GC B cell subset had increased expression of the plasma cell marker Blimp-1. DAF expression was also modulated during B cell hematopoiesis in the human bone marrow. Collectively, our results reveal a novel role of DAF to pre-prime activated human B cells for phagocytosis prior to apoptosis.


Subject(s)
B-Lymphocytes/immunology , CD55 Antigens/immunology , Gene Expression Regulation/immunology , Germinal Center/immunology , Lymphocyte Activation , Phagocytosis , B-Lymphocytes/cytology , Germinal Center/cytology , Humans , Positive Regulatory Domain I-Binding Factor 1/immunology
19.
Cell Rep ; 29(7): 1848-1861.e6, 2019 11 12.
Article in English | MEDLINE | ID: mdl-31722202

ABSTRACT

Follicular regulatory T (TFR) cells are a specialized suppressive subset that controls the germinal center (GC) response and maintains humoral self-tolerance. The mechanisms that maintain TFR lineage identity and suppressive activity remain largely unknown. Here, we show that expression of Blimp1 by FoxP3+ TFR cells is essential for TFR lineage stability, entry into the GC, and expression of regulatory activity. Deletion of Blimp1 in TFR cells reduced FoxP3 and CTLA-4 expression and increased pro-inflammatory cytokines and spontaneous production of autoantibodies, including elevated IgE. Maintenance of TFR stability reflected Blimp1-dependent repression of the IL-23R-STAT3 axis and activation of the CD25-STAT5 pathway, while silenced IL-23R-STAT3 or increased STAT5 activation rescued the Blimp1-deficient TFR phenotype. Blimp1-dependent control of CXCR5/CCR7 expression also regulated TFR homing into the GC. These findings uncover a Blimp1-dependent TFR checkpoint that enforces suppressive activity and acts as a gatekeeper of GC entry.


Subject(s)
Gene Expression Regulation/immunology , Germinal Center/immunology , Positive Regulatory Domain I-Binding Factor 1/immunology , Signal Transduction/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cell Line , Germinal Center/cytology , Humans , Interleukin-2 Receptor alpha Subunit/genetics , Interleukin-2 Receptor alpha Subunit/immunology , Mice , Mice, Transgenic , Positive Regulatory Domain I-Binding Factor 1/genetics , Receptors, CCR7/genetics , Receptors, CCR7/immunology , Receptors, CXCR5/genetics , Receptors, CXCR5/immunology , Receptors, Interleukin/genetics , Receptors, Interleukin/immunology , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/immunology , STAT5 Transcription Factor/genetics , STAT5 Transcription Factor/immunology , Signal Transduction/genetics , T-Lymphocytes, Regulatory/cytology
20.
Front Immunol ; 10: 1909, 2019.
Article in English | MEDLINE | ID: mdl-31474988

ABSTRACT

Regulatory B cells (Bregs) are a B cell subset that plays a suppressive role in immune responses. The CD19+CD1dhiCD5+ Bregs that can execute regulatory functions via secreting IL-10 are defined as B10 cells. Bregs suppress autoimmune and inflammatory diseases, whereas they exacerbate infectious diseases caused by bacteria, viruses, or parasites. Notably, the molecular mechanisms regulating the development and functions of Bregs are still largely unknown. Furthermore, the biological impact of Bregs in fungal infection has not yet been demonstrated. Here, we compared the gene expression profiles of IL-10-producing and -non-producing mouse splenic B cells stimulated with lipopolysaccharide (LPS) or anti-CD40 antibody. Blimp-1, a transcription factor known to be critical for plasma cell differentiation, was found to be enriched in the IL-10-producing B cells. The frequency of Blimp-1+ B10 cells was increased in LPS-treated mice and in isolated B10 cells that were stimulated with LPS. Surprisingly, B cell-specific Blimp-1 knockout (Cko) mice, generated by CD19 promoter driven Cre recombinase-dependent deletion of Prdm1 (gene encoding Blimp-1), showed higher frequencies of B10 cells both in the steady state and following injection with LPS, as compared with control littermates. However, B10 cells lacking Blimp-1 failed to efficiently suppress the proliferation of naïve CD4+ T cells primed with anti-CD3 and anti-CD28 antibodies. B10 cells can be stimulated for further differentiation into plasmablasts, and a subset of plasmablasts express IL-10. We found that B10 cells from Cko mice failed to generate both IL-10-non-producing and IL-10-producing plasmablasts. Mechanistically, we found that Blimp-1 can directly suppress Il-10, whereas, in the presence of activated STAT3, Blimp-1 works together with activated STAT3 to upregulate Il-10. Moreover, we also found that B10 cells improve the clearance of Candida albicans infection but worsen the infection mortality. Notably, a lack of Blimp-1 in B10 cells did not change these effects of adoptively transferred B10 cells on fungal infections. Together, our data show that Blimp-1 regulates the generation, differentiation, and IL-10 production of Bregs.


Subject(s)
Antigens, CD19/immunology , B-Lymphocytes, Regulatory/immunology , Interleukin-10/immunology , Positive Regulatory Domain I-Binding Factor 1/immunology , Animals , Antigens, CD19/genetics , Antigens, CD19/metabolism , B-Lymphocytes, Regulatory/metabolism , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cells, Cultured , Gene Expression Profiling/methods , Humans , Interleukin-10/genetics , Interleukin-10/metabolism , Lipopolysaccharides/immunology , Lipopolysaccharides/pharmacology , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Positive Regulatory Domain I-Binding Factor 1/genetics , Positive Regulatory Domain I-Binding Factor 1/metabolism , Spleen/cytology , Spleen/drug effects , Spleen/immunology , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...