Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 185
1.
J Immunol ; 212(11): 1766-1781, 2024 Jun 01.
Article En | MEDLINE | ID: mdl-38683120

Better understanding of the host responses to Mycobacterium tuberculosis infections is required to prevent tuberculosis and develop new therapeutic interventions. The host transcription factor BHLHE40 is essential for controlling M. tuberculosis infection, in part by repressing Il10 expression, where excess IL-10 contributes to the early susceptibility of Bhlhe40-/- mice to M. tuberculosis infection. Deletion of Bhlhe40 in lung macrophages and dendritic cells is sufficient to increase the susceptibility of mice to M. tuberculosis infection, but how BHLHE40 impacts macrophage and dendritic cell responses to M. tuberculosis is unknown. In this study, we report that BHLHE40 is required in myeloid cells exposed to GM-CSF, an abundant cytokine in the lung, to promote the expression of genes associated with a proinflammatory state and better control of M. tuberculosis infection. Loss of Bhlhe40 expression in murine bone marrow-derived myeloid cells cultured in the presence of GM-CSF results in lower levels of proinflammatory associated signaling molecules IL-1ß, IL-6, IL-12, TNF-α, inducible NO synthase, IL-2, KC, and RANTES, as well as higher levels of the anti-inflammatory-associated molecules MCP-1 and IL-10 following exposure to heat-killed M. tuberculosis. Deletion of Il10 in Bhlhe40-/- myeloid cells restored some, but not all, proinflammatory signals, demonstrating that BHLHE40 promotes proinflammatory responses via both IL-10-dependent and -independent mechanisms. In addition, we show that macrophages and neutrophils within the lungs of M. tuberculosis-infected Bhlhe40-/- mice exhibit defects in inducible NO synthase production compared with infected wild-type mice, supporting that BHLHE40 promotes proinflammatory responses in innate immune cells, which may contribute to the essential role for BHLHE40 during M. tuberculosis infection in vivo.


Basic Helix-Loop-Helix Transcription Factors , Interleukin-10 , Mice, Knockout , Myeloid Cells , Animals , Mice , Interleukin-10/immunology , Interleukin-10/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/immunology , Myeloid Cells/immunology , Mycobacterium tuberculosis/immunology , Macrophages/immunology , Homeodomain Proteins/genetics , Mice, Inbred C57BL , Granulocyte-Macrophage Colony-Stimulating Factor , Dendritic Cells/immunology , Lung/immunology , Tuberculosis/immunology , Cell Polarity , Cells, Cultured
2.
FEBS Lett ; 596(21): 2834-2850, 2022 11.
Article En | MEDLINE | ID: mdl-36053046

The hypoxia-inducible factors (HIF)-1α and HIF-2α are central regulators of transcriptional programmes in settings such as development and tumour expansion. HIF-2α moonlights as a cap-dependent translation factor. We provide new insights into how the interferon-stimulated gene 15 (ISG15), a ubiquitin-like modifier, and the HIFs regulate one another in hypoxia and interferon-induced cells. We show that upon ISGylation induction and HIF-α stabilization, both HIFs promote protein ISGylates through transcriptional and/or post-transcriptional pathways. We show the first evidence of HIF-2α modification by ISG15. ISGylation induces system-level alterations to the HIF transcriptional programme and increases the cytoplasmic/nuclear fraction and translation activity of HIF-2α. This work identifies ISG15 as a regulator of hypoxic mRNA translation, which has implications for immune processes and disease progression.


Basic Helix-Loop-Helix Transcription Factors , Hypoxia , Polyribosomes , Humans , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/immunology , Cell Hypoxia/genetics , Cell Hypoxia/immunology , Hypoxia/genetics , Hypoxia/immunology , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/immunology , Interferons/genetics , Interferons/immunology , Polyribosomes/genetics , Polyribosomes/immunology
3.
Front Immunol ; 13: 848577, 2022.
Article En | MEDLINE | ID: mdl-35990644

The E protein transcription factors E2A and HEB are critical for many developmental processes, including T cell development. We have shown that the Tcf12 locus gives rise to two distinct HEB proteins, with alternative (HEBAlt) and canonical (HEBCan) N-terminal domains, which are co-expressed during early T cell development. While the functional domains of HEBCan have been well studied, the nature of the HEBAlt-specific (Alt) domain has been obscure. Here we provide compelling evidence that the Alt domain provides a site for the molecular integration of cytokine signaling and E protein activity. Our results indicate that phosphorylation of a unique YYY motif in the Alt domain increases HEBAlt activity by 10-fold, and that this increase is dependent on Janus kinase activity. To enable in vivo studies of HEBAlt in the T cell context, we generated ALT-Tg mice, which can be induced to express a HA-tagged HEBAlt coding cassette in the presence of Cre recombinases. Analysis of ALT-Tg mice on the Vav-iCre background revealed a minor change in the ratio of ISP cells to CD8+ SP cells, and a mild shift in the ratio of T cells to B cells in the spleen, but otherwise the thymus, spleen, and bone marrow lymphocyte subsets were comparable at steady state. However, kinetic analysis of T cell development in OP9-DL4 co-cultures revealed a delay in early T cell development and a partial block at the DN to DP transition when HEBAlt levels or activity were increased. We also observed that HEBCan and HEBAlt displayed significant differences in protein stability that were resolved in the thymocyte context. Finally, a proteomic screen identified STAT1 and Xpo1 as potential members of HEBAlt-containing complexes in thymocytes, consistent with JAK-induced activation of HEBAlt accompanied by translocation to the nucleus. Thus, our results show that the Alt domain confers access to multiple layers of post-translational control to HEBAlt that are not available to HEBCan, and thus may serve as a rheostat to tune E protein activity levels as cells move through different thymic signaling environments during T cell development.


Basic Helix-Loop-Helix Transcription Factors , Cell Differentiation , T-Lymphocytes , Animals , Basic Helix-Loop-Helix Transcription Factors/immunology , Cell Differentiation/immunology , Kinetics , Mice , Proteomics , T-Lymphocytes/immunology , Transcription Factors/immunology
4.
Front Immunol ; 13: 899413, 2022.
Article En | MEDLINE | ID: mdl-35757772

L. johnsonii N6.2 releases nano-sized vesicles (NVs) with distinct protein and lipid contents. We hypothesized that these NVs play a central role in the delivery of bioactive molecules that may act as mechanistic effectors in immune modulation. In this report, we observed that addition of NVs to the human pancreatic cell line ßlox5 reduced cytokine-induced apoptosis. Through RNAseq analyses, increased expression of CYP1A1, CYP1B1, AHRR, and TIPARP genes in the aryl hydrocarbon receptor (AHR) pathways were found to be significantly induced in presence of NVs. AHR nuclear translocation was confirmed by confocal microscopy. The role of NVs on beta cell function was further evaluated using primary human pancreatic islets. It was found that NVs significantly increased insulin secretion in presence of high glucose concentrations. These increases positively correlated with increased GLUT6 and SREBF1 mRNA and coincided with reduced oxidative stress markers. Furthermore, incubation of NVs with THP-1 macrophages promoted the M2 tolerogenic phenotype through STAT3 activation, expression of AHR-dependent genes and secretion of IL10. Altogether, our findings indicate that bacterial NVs have the potential to modulate glucose homeostasis in the host by directly affecting insulin secretion by islets and through the induction of a tolerogenic immune phenotype.


Basic Helix-Loop-Helix Transcription Factors , Interleukin-10 , Lactobacillus johnsonii , Receptors, Aryl Hydrocarbon , Apoptosis/immunology , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/immunology , Basic Helix-Loop-Helix Transcription Factors/metabolism , Glucose/metabolism , Humans , Interleukin-10/immunology , Interleukin-10/metabolism , Lactobacillus johnsonii/genetics , Lactobacillus johnsonii/immunology , Lactobacillus johnsonii/metabolism , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/immunology , Receptors, Aryl Hydrocarbon/metabolism
5.
Int Immunopharmacol ; 102: 107892, 2022 Jan.
Article En | MEDLINE | ID: mdl-34215553

Differentiated embryo-chondrocyte expressed gene 1 (DEC1) belongs to the family of basic helix-loop-helix (bHLH)-type transcription factors. DEC1 is expressed in various mammalian cells, but early studies focused on its roles outside the immune system. In recent years, relevant studies have found that DEC1 plays an important role in the immunotherapy of tumors, the functional regulation of the immune system, and the onset of autoimmune diseases. DEC1 promotes interferon (IFN)-γand granulocyte-macrophage colony-stimulating factor (GM-CSF) secretion through the production of CD4+ T cells, which promotes inflammatory defense responses and autoimmune diseases. Additionally, DEC1 can inhibit the expression of interleukin (IL)-10 to further strengthen the immune response. In this review, we summarized recent advances in our understanding of the roles of DEC1 in animal models and human cells, including regulating immune cell differentiation, controlling cytokine production, and maintaining the balance of pro- and anti-inflammatory signals.


Basic Helix-Loop-Helix Transcription Factors/immunology , Homeodomain Proteins/immunology , Animals , Autoimmune Diseases/immunology , Humans , Immunomodulation , Neoplasms/immunology , Neoplasms/therapy
6.
Front Immunol ; 12: 738435, 2021.
Article En | MEDLINE | ID: mdl-34603319

Background: Although mRNA vaccines have been efficient for combating a variety of tumors, their effectiveness against glioma remains unclear. There is growing evidence that immunophenotyping can reflect the comprehensive immune status and microenvironment of the tumor, which correlates closely with treatment response and vaccination potency. The purpose of this research was to screen for effective antigens in glioma that could be used for developing mRNA vaccines and to further differentiate the immune subtypes of glioma to create an selection criteria for suitable patients for vaccination. Methods: Gene expression profiles and clinical data of 698 glioma samples were extracted from The Cancer Genome Atlas, and RNA_seq data of 1018 glioma samples was gathered from Chinese Glioma Genome Atlas. Gene Expression Profiling Interactive Analysis was used to determine differential expression genes and prognostic markers, cBioPortal software was used to verify gene alterations, and Tumor Immune Estimation Resource was used to investigate the relationships among genes and immune infiltrating cells. Consistency clustering was applied for consistent matrix construction and data aggregation, Gene oncology enrichment was performed for functional annotation, and a graph learning-based dimensionality reduction method was applied to describe the subtypes of immunity. Results: Four overexpressed and mutated tumor antigens associated with poor prognosis and infiltration of antigen presenting cells were identified in glioma, including TP53, IDH1, C3, and TCF12. Besides, four immune subtypes of glioma (IS1-IS4) and 10 immune gene modules were identified consistently in the TCGA data. The immune subtypes had diverse molecular, cellular, and clinical features. IS1 and IS4 displayed an immune-activating phenotype and were associated with worse survival than the other two subtypes, while IS2 and IS3 had lower levels of tumor immune infiltration. Immunogenic cell death regulators and immune checkpoints were also diversely expressed in the four immune subtypes. Conclusion: TP53, IDH1, C3, and TCF12 are effective antigens for the development of anti-glioma mRNA vaccines. We found four stable and repeatable immune subtypes of human glioma, the classification of the immune subtypes of glioma may play a crucial role in the predicting mRNA vaccine outcome.


Antigens, Neoplasm/genetics , Brain Neoplasms/drug therapy , Cancer Vaccines/therapeutic use , Glioma/drug therapy , Vaccine Development , mRNA Vaccines/therapeutic use , Antigens, Neoplasm/immunology , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/immunology , Brain Neoplasms/genetics , Brain Neoplasms/immunology , Cancer Vaccines/immunology , Clinical Decision-Making , Complement C3/genetics , Complement C3/immunology , Databases, Genetic , Gene Expression Profiling , Glioma/genetics , Glioma/immunology , Humans , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/metabolism , Mutation , Precision Medicine , Transcriptome , Tumor Microenvironment , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/immunology , mRNA Vaccines/immunology
7.
Front Immunol ; 12: 697663, 2021.
Article En | MEDLINE | ID: mdl-34249001

The prevalence of chronic inflammatory diseases including inflammatory bowel disease (IBD), autoimmunity and cancer have increased in recent years. Herbal-based compounds such as flavonoids have been demonstrated to contribute to the modulation of these diseases although understanding their mechanism of action remains limited. Flavonoids are able to interact with cellular immune components in a distinct way and influence immune responses at a molecular level. In this mini review, we highlight recent progress in our understanding of the modulation of immune responses by the aryl hydrocarbon receptor (AhR), a ligand-dependent transcription factor whose activity can be regulated by diverse molecules including flavonoids. We focus on the role of AhR in integrating signals from flavonoids to modulate inflammatory responses using in vitro and experimental animal models. We also summarize the limitations of these studies. Medicinal herbs have been widely used to treat inflammatory disorders and may offer a valuable therapeutic strategy to treat aberrant inflammatory responses by modulation of the AhR pathway.


Basic Helix-Loop-Helix Transcription Factors/immunology , Immunomodulation , Phytotherapy , Plants, Medicinal/chemistry , Receptors, Aryl Hydrocarbon/immunology , Animals , Autoimmune Diseases/drug therapy , Autoimmune Diseases/immunology , Flavonoids/chemistry , Flavonoids/immunology , Flavonoids/therapeutic use , Humans , Inflammation/drug therapy , Inflammation/immunology , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/immunology , Ligands , Signal Transduction/drug effects , Signal Transduction/immunology
8.
Pathol Res Pract ; 224: 153511, 2021 Aug.
Article En | MEDLINE | ID: mdl-34214845

Ulcerative colitis (UC) is a chronic inflammatory intestinal disease. Genetic susceptibility, gut microbiota and mucosal immune dysfunction play important roles in the pathogenesis and development of UC. We investigate the effect of Mist1 in model of colitis and its underlying mechanism. The expressions of Mist1 in patients with colitis tissue were up-regulated. Meanwhile, Mist1 mRNA and protein expressions in DSS-induced colitis mice model were also induced and Mist1 mRNA and protein expressions of LPS induced THP-1 cell were also up-regulated. we found Mist1 human protein promoted inflammation in DSS-induced colitis mice by NLRP3. So, we up-regulated Mist1 expression and over-expression of Mist1 promoted IL-1ß and NLRP3 protein expression levels in vitro model. However, down-regulation of Mist1 suppressed IL-1ß and NLRP3 protein expression levels in vitro model. Next, SNAI1 is a shooting point of Mist1 in the effects of Mist1 in colitis. The inhibition of SNAI1 reduced the effects of Mist1 on NLRP3 inflammasome in vitro model. Activation of SNAI1 induced the effects of Mist1 on NLRP3 inflammasome in vitro model. Lastly, anti-SNAI1 human protein lowered the effects of Mist1 human protein on NLRP3 inflammasome in DSS-induced colitis mice. We demonstrated that Mist1 promoted inflammation in colitis model via NLRP3 inflammasome by SNAI1, whereas the absence of these macrophages led to a significant improvement in colitis treatment.


Basic Helix-Loop-Helix Transcription Factors/metabolism , Inflammation/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Snail Family Transcription Factors/metabolism , Adenosine Triphosphatases/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/immunology , Colitis/pathology , Humans , Inflammasomes/drug effects , Inflammasomes/immunology , Inflammasomes/metabolism , Mice
9.
Front Immunol ; 12: 564948, 2021.
Article En | MEDLINE | ID: mdl-34290693

Background: Previous studies have reported the potential of aryl hydrocarbon receptor (AhR) in cancer immunotherapy. However, the mechanisms underpinning its therapeutic value have yet to be comprehensively investigated. Thus, this research aimed to explore the underlying association between AhR and cancer immunotherapy in 33 human cancers. Methods: The gene expression data and clinical characteristics of 33 cancers were retrieved from The Cancer Genome Atlas database. The immunotherapeutic cohorts included GSE67501 and GSE78220 as well as IMvigor210, which were obtained from the Gene Expression Omnibus database and included in a previously published study respectively. Clinical parameters, including patient age, gender, survival, and tumor stage were analyzed to assess the prognostic value of AhR. The activity of AhR was generated by single sample gene set enrichment analysis and used to evaluate the difference between the AhR transcriptome and protein expression level. To better understand the role of AhR in cancer immunotherapy, the correlation between AhR and tumor microenvironment, as well as its relation to immune processes/elements, such as immune cell infiltration, immune inhibitors and stimulators, and the major histocompatibility complex were analyzed. The relevant underlying pathways associated with AhR signaling in cancer were also explored. Furthermore, the correlation between AhR and two immunotherapeutic biomarkers (tumor mutational burden and microsatellite instability) was investigated. Finally, the relationship between AhR and immunotherapeutic response was explored using three independent immunotherapeutic cohorts. Results: Although AhR was not closely associated with age (5/33), gender (3/33), or tumor stage (3/21) in any of the studied human cancers, it exhibited potential prognostic value for predicting patient survival. Consistency has been observed between AhR activity and expression in some cancers (7/33). Generally, AhR presented a robust correlation with immune cell infiltration, immune modulators, and immunotherapeutic markers. Moreover, high AhR expression was significantly related to immune-relevant pathways. However, no significant correlation was observed between AhR and the immunotherapeutic response. Conclusions: This research investigated the immunotherapeutic value of AhR in 33 human cancers, providing evidence regarding the function of AhR and its role in clinical treatment. However, considering that a bioinformatics approach was adopted, the current results are preliminary and require further validation.


Immunotherapy , Neoplasms/genetics , Neoplasms/therapy , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/immunology , Basic Helix-Loop-Helix Transcription Factors/immunology , Basic Helix-Loop-Helix Transcription Factors/metabolism , Biomarkers, Tumor/genetics , Computational Biology , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Microsatellite Instability , Neoplasms/classification , Neoplasms/pathology , Receptors, Aryl Hydrocarbon/metabolism , Transcriptome , Tumor Microenvironment/immunology
10.
Front Immunol ; 12: 645168, 2021.
Article En | MEDLINE | ID: mdl-34093534

Accumulating evidence indicates that nutrition can modulate the immune system through metabolites, either produced by host digestion or by microbiota metabolism. In this review, we focus on dietary metabolites that are agonists of the Aryl hydrocarbon Receptor (AhR). AhR is a ligand-activated transcription factor, initially characterized for its interaction with xenobiotic pollutants. Numerous studies have shown that AhR also recognizes indoles and tryptophan catabolites originating from dietary compounds and commensal bacteria. Here, we review recent work employing diet manipulation to address the impact of nutritional AhR agonists on immune responses, both locally in the intestine and at distant sites. In particular, we examine the physiological role of these metabolites in immune cell development and functions (including T lymphocytes, innate-like lymphoid cells, and mononuclear phagocytes) and their effect in inflammatory disorders.


Basic Helix-Loop-Helix Transcription Factors/immunology , Food , Intestines/immunology , Phagocytes/immunology , Receptors, Aryl Hydrocarbon/immunology , T-Lymphocytes/immunology , Humans , Inflammation/immunology , Ligands
11.
Front Immunol ; 12: 643260, 2021.
Article En | MEDLINE | ID: mdl-33936062

We have previously demonstrated that benzo(a)pyrene (BaP) co-exposure with dermatophagoides group 1 allergen (Der f 1) can potentiate Der f 1-induced airway inflammation. The underlying mechanism, however, remains undetermined. Here we investigated the molecular mechanisms underlying the potentiation of BaP exposure on Der f 1-induced airway inflammation in asthma. We found that BaP co-exposure potentiated Der f 1-induced TGFß1 secretion and signaling activation in human bronchial epithelial cells (HBECs) and the airways of asthma mouse model. Moreover, BaP exposure alone or co-exposure with Der f 1-induced aryl hydrocarbon receptor (AhR) activity was determined by using an AhR-dioxin-responsive element reporter plasmid. The BaP and Der f 1 co-exposure-induced TGFß1 expression and signaling activation were attenuated by either AhR antagonist CH223191 or AhR knockdown in HBECs. Furthermore, AhR knockdown led to the reduction of BaP and Der f 1 co-exposure-induced active RhoA. Inhibition of RhoA signaling with fasudil, a RhoA/ROCK inhibitor, suppressed BaP and Der f 1 co-exposure-induced TGFß1 expression and signaling activation. This was further confirmed in HBECs expressing constitutively active RhoA (RhoA-L63) or dominant-negative RhoA (RhoA-N19). Luciferase reporter assays showed prominently increased promoter activities for the AhR binding sites in the promoter region of RhoA. Inhibition of RhoA suppressed BaP and Der f 1 co-exposure-induced airway hyper-responsiveness, Th2-associated airway inflammation, and TGFß1 signaling activation in asthma. Our studies reveal a previously unidentified functional axis of AhR-RhoA in regulating TGFß1 expression and signaling activation, representing a potential therapeutic target for allergic asthma.


Antigens, Dermatophagoides/toxicity , Arthropod Proteins/toxicity , Asthma , Basic Helix-Loop-Helix Transcription Factors/immunology , Benzo(a)pyrene/toxicity , Cysteine Endopeptidases/toxicity , Receptors, Aryl Hydrocarbon/immunology , Signal Transduction , Transforming Growth Factor beta1/immunology , rhoA GTP-Binding Protein/immunology , Animals , Asthma/chemically induced , Asthma/immunology , Asthma/pathology , Female , Male , Mice , Signal Transduction/drug effects , Signal Transduction/immunology
12.
J Clin Invest ; 131(7)2021 04 01.
Article En | MEDLINE | ID: mdl-33792560

Adoptive T cell therapies (ACTs) hold great promise in cancer treatment, but low overall response rates in patients with solid tumors underscore remaining challenges in realizing the potential of this cellular immunotherapy approach. Promoting CD8+ T cell adaptation to tissue residency represents an underutilized but promising strategy to improve tumor-infiltrating lymphocyte (TIL) function. Here, we report that deletion of the HIF negative regulator von Hippel-Lindau (VHL) in CD8+ T cells induced HIF-1α/HIF-2α-dependent differentiation of tissue-resident memory-like (Trm-like) TILs in mouse models of malignancy. VHL-deficient TILs accumulated in tumors and exhibited a core Trm signature despite an exhaustion-associated phenotype, which led to retained polyfunctionality and response to αPD-1 immunotherapy, resulting in tumor eradication and protective tissue-resident memory. VHL deficiency similarly facilitated enhanced accumulation of chimeric antigen receptor (CAR) T cells with a Trm-like phenotype in tumors. Thus, HIF activity in CD8+ TILs promotes accumulation and antitumor activity, providing a new strategy to enhance the efficacy of ACTs.


Basic Helix-Loop-Helix Transcription Factors/immunology , CD8-Positive T-Lymphocytes/immunology , Hypoxia-Inducible Factor 1, alpha Subunit/immunology , Immunity, Cellular , Immunologic Memory , Lymphocytes, Tumor-Infiltrating/immunology , Neoplasms, Experimental/immunology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , CD8-Positive T-Lymphocytes/pathology , Cell Line, Tumor , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Lymphocytes, Tumor-Infiltrating/pathology , Mice , Mice, Knockout , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Von Hippel-Lindau Tumor Suppressor Protein/immunology
13.
Proc Natl Acad Sci U S A ; 118(16)2021 04 20.
Article En | MEDLINE | ID: mdl-33859041

During an acute viral infection, CD8 T cells encounter a myriad of antigenic and inflammatory signals of variable strength, which sets off individual T cells on their own differentiation trajectories. However, the developmental path for each of these cells will ultimately lead to one of only two potential outcomes after clearance of the infection-death or survival and development into memory CD8 T cells. How this cell fate decision is made remains incompletely understood. In this study, we explore the transcriptional changes during effector and memory CD8 T cell differentiation at the single-cell level. Using single-cell, transcriptome-derived gene regulatory network analysis, we identified two main groups of regulons that govern this differentiation process. These regulons function in concert with changes in the enhancer landscape to confer the establishment of the regulatory modules underlying the cell fate decision of CD8 T cells. Furthermore, we found that memory precursor effector cells maintain chromatin accessibility at enhancers for key memory-related genes and that these enhancers are highly enriched for E2A binding sites. Finally, we show that E2A directly regulates accessibility of enhancers of many memory-related genes and that its overexpression increases the frequency of memory precursor effector cells and accelerates memory cell formation while decreasing the frequency of short-lived effector cells. Overall, our results suggest that effector and memory CD8 T cell differentiation is largely regulated by two transcriptional circuits, with E2A serving as an important epigenetic regulator of the memory circuit.


Basic Helix-Loop-Helix Transcription Factors/metabolism , CD8-Positive T-Lymphocytes/metabolism , Immunologic Memory/immunology , Animals , Basic Helix-Loop-Helix Transcription Factors/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/physiology , Cell Differentiation/genetics , Chromatin/metabolism , Epigenesis, Genetic/genetics , Epigenomics/methods , Female , Gene Expression/genetics , Gene Expression Profiling/methods , Gene Expression Regulation/genetics , Gene Regulatory Networks/genetics , Hematopoiesis , Humans , Immunologic Memory/genetics , Lymphocyte Activation , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Sequence Analysis, RNA , Single-Cell Analysis
14.
J Allergy Clin Immunol ; 148(2): 633-638, 2021 08.
Article En | MEDLINE | ID: mdl-33819507

BACKGROUND: IL-31 is a major pruritogen associated with atopic dermatitis (AD). Although a specific antibody for IL-31 receptor has been shown to alleviate pruritus in patients with AD, therapeutic approaches to inhibition of IL-31 production remain unexploited. IL-31 production by TH cells critically depends on the transcription factor EPAS1, which mediates IL31 promoter activation in collaboration with SP1. OBJECTIVE: We aimed at developing small-molecule inhibitors that selectively block IL-31 production by TH cells. METHODS: We generated the reporter cell line that inducibly expressed EPAS1 in the presence of doxycycline to mediate Il31 promoter activation, and we screened 9600 chemical compounds. The selected compounds were further examined by using TH cells from a spontaneous mouse model of AD and TH cells from patients with AD. RESULTS: We have identified 4-(2-(4-isopropylbenzylidene)hydrazineyl)benzoic acid (IPHBA) as an inhibitor of IL31 induction. Although IPHBA did not affect nonspecific T-cell proliferation, IPHBA inhibited antigen-induced IL-31 production by TH cells from both an AD mouse model and patients with AD without affecting other cytokine production and hypoxic responses. In line with this, itch responses induced by adoptive transfer of IL-31-producing TH cells were attenuated when mice were orally treated with IPHBA. Mechanistically, IPHBA inhibited the association between EPAS1 and SP1, resulting in defective recruitment of both transcription factors to the specific sites of the IL31 promoter. We also determined the structure-activity relationship of IPHBA by synthesizing and analyzing 201 analogous compounds. CONCLUSION: IPHBA could be a potential drug leading to inhibition of EPAS1-driven IL-31 production.


Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Dermatitis, Atopic/immunology , Doxycycline/pharmacology , Gene Expression Regulation/drug effects , Interleukins/immunology , Signal Transduction/drug effects , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/immunology , Dermatitis, Atopic/genetics , Dermatitis, Atopic/pathology , Gene Expression Regulation/immunology , Interleukins/genetics , Mice , Mice, Knockout , Promoter Regions, Genetic , Signal Transduction/genetics , Signal Transduction/immunology , T-Lymphocytes, Helper-Inducer
16.
Biomed Environ Sci ; 34(3): 192-202, 2021 Mar 20.
Article En | MEDLINE | ID: mdl-33766215

OBJECTIVE: To investigate involvement of the aryl hydrocarbon receptor (AhR) in the immunomodulatory effects of cadmium (Cd). METHODS: The effect of Cd on AhR activation ( CYP1A1 and CYP1B1 mRNA expression) was examined in lung leukocytes of Cd-exposed rats (5 and 50 mg/L, 30 d orally) and by in vitro leukocyte exposure. The involvement of AhR signaling in the effects of Cd on the interleukin (IL)-1ß, IL-6, and tumor necrosis factor (TNF) lung leukocyte response was investigated in vitro using the receptor antagonist CH-223191. RESULTS: Cd increased CYP1B1 ( in vivo and in vitro) and CYP1A1 ( in vitro) mRNA, indicating AhR involvement in the action of Cd. In response to Cd, lung leukocytes increased IL-6 and decreased TNF at the gene expression and protein levels, but decreased IL-1ß production due to reduced NLRP3. The AhR antagonist CH-223191 abrogated the observed effects of Cd on the cytokine response. The absence of AhR reactivity and cytokine response to Cd of leukocytes from the lungs of a rat strain that is less sensitive to Cd toxicity coincided with a high AhR repressor mRNA level. CONCLUSION: AhR signaling is involved in the lung leukocyte proinflammatory cytokine response to Cd. The relevance of the AhR to the cytokine response to Cd provides new insight into the mechanisms of Cd immunotoxicity.


Basic Helix-Loop-Helix Transcription Factors/genetics , Cadmium/toxicity , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1B1/genetics , Cytokines/immunology , Environmental Pollutants/toxicity , Receptors, Aryl Hydrocarbon/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/immunology , Cytochrome P-450 CYP1A1/immunology , Cytochrome P-450 CYP1B1/immunology , Male , Rats , Receptors, Aryl Hydrocarbon/immunology
17.
Int J Mol Sci ; 22(4)2021 Feb 12.
Article En | MEDLINE | ID: mdl-33673338

Identifying historical trajectories is a useful exercise in research, as it helps clarify important, perhaps even "paradigmatic", shifts in thinking and moving forward in science. In this review, the development of research regarding the role of the transcription factor "aryl hydrocarbon receptor" (AHR) as a mediator of the toxicity of environmental pollution towards a link between the environment and a healthy adaptive response of the immune system and the skin is discussed. From this fascinating development, the opportunities for targeting the AHR in the therapy of many diseases become clear.


Basic Helix-Loop-Helix Transcription Factors/immunology , Receptors, Aryl Hydrocarbon/immunology , Skin/immunology , Thymus Gland/immunology , Animals , Humans
18.
Cancer Immunol Res ; 9(4): 401-414, 2021 04.
Article En | MEDLINE | ID: mdl-33602720

Adoptive transfer of antitumor cytotoxic T cells is an emerging form of cancer immunotherapy. A key challenge to expanding the utility of adoptive cell therapies is how to enhance the survival and function of the transferred T cells. Immune-cell survival requires adaptation to different microenvironments and particularly to the hypoxic milieu of solid tumors. The hypoxia-inducible factor (HIF) transcription factors are an essential aspect of this adaptation. In this study, we undertook experiments to define structural determinants of HIF that potentiate antitumor efficacy in cytotoxic T cells. We first created retroviral vectors to deliver ectopic expression of HIF1α and HIF2α in mouse CD8+ T cells, together or individually and with or without sensitivity to the oxygen-dependent HIFα inhibitors Von Hippel-Lindau and factor-inhibiting HIF (FIH). HIF2α, but not HIF1α, drove broad transcriptional changes in CD8+ T cells, resulting in increased cytotoxic differentiation and cytolytic function against tumor targets. A specific mutation replacing the hydroxyl group-acceptor site for FIH in HIF2α gave rise to the most effective antitumor T cells after adoptive transfer in vivo In addition, codelivering an FIH-insensitive form of HIF2α with an anti-CD19 chimeric antigen receptor greatly enhanced cytolytic function of human CD8+ T cells against lymphoma cells both in vitro and in a xenograft adoptive transfer model. These experiments point to a means to increase the antitumor efficacy of therapeutic CD8+ T cells via ectopic expression of the HIF transcription factor.See related Spotlight on p. 364.


Basic Helix-Loop-Helix Transcription Factors/immunology , CD8-Positive T-Lymphocytes/immunology , Hypoxia-Inducible Factor 1, alpha Subunit/immunology , Hypoxia/immunology , Immunotherapy, Adoptive , Animals , Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Cell Line, Tumor , Female , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Male , Mice , Mice, Inbred C57BL , Transcription Factors , Tumor Microenvironment
19.
Curr Opin Immunol ; 70: 33-39, 2021 06.
Article En | MEDLINE | ID: mdl-33607496

Dendritic cells (DCs) are potent antigen-presenting cells (APCs), which sample the exogenous and endogenous cues to control adaptive immunity, balancing effector and regulatory components of the immune response. Multiple subsets of DCs, such as plasmacytoid and conventional DCs, have been defined based on specific phenotypic markers, functions and regulatory transcriptional programs. Tolerogenic DCs (tolDCs) have been functionally defined based on their ability to expand the regulatory T-cell compartment and suppress immune responses. However, it is still unclear whether tolDCs represent a homogeneous population, a specific DC subset and/or a heterogeneous collection of DC activation/maturation states. The ligand-activated transcription factor aryl hydrocarbon receptor (AHR) has been shown to control transcriptional programs associated to tolDCs. In this review, we discuss the role of AHR in the control of tolDCs, and also AHR-targeted approaches for the therapeutic induction of tolDCs in autoimmune diseases and allergy.


Basic Helix-Loop-Helix Transcription Factors/immunology , Dendritic Cells/immunology , Receptors, Aryl Hydrocarbon/immunology , Autoimmune Diseases/immunology , Humans , Hypersensitivity/immunology , Signal Transduction/immunology
20.
Nat Commun ; 12(1): 1042, 2021 02 15.
Article En | MEDLINE | ID: mdl-33589625

Necrotizing enterocolitis (NEC) is a disease of premature infants characterized by acute intestinal necrosis. Current dogma suggests that NEC develops in response to post-natal dietary and bacterial factors, and so a potential role for in utero factors in NEC remains unexplored. We now show that during pregnancy, administration of a diet rich in the aryl hydrocarbon receptor (AHR) ligand indole-3-carbinole (I3C), or of breast milk, activates AHR and prevents NEC in newborn mice by reducing Toll-like receptor 4 (TLR4) signaling in the newborn gut. Protection from NEC requires activation of AHR in the intestinal epithelium which is reduced in mouse and human NEC, and is independent of leukocyte activation. Finally, we identify an AHR ligand ("A18") that limits TLR4 signaling in mouse and human intestine, and prevents NEC in mice when administered during pregnancy. In summary, AHR signaling is critical in NEC development, and maternally-delivered, AHR-based therapies may alleviate NEC.


Basic Helix-Loop-Helix Transcription Factors/genetics , Enterocolitis, Necrotizing/genetics , Indoles/administration & dosage , Milk, Human/physiology , Receptors, Aryl Hydrocarbon/genetics , Toll-Like Receptor 4/genetics , Animals , Animals, Newborn , Basic Helix-Loop-Helix Transcription Factors/agonists , Basic Helix-Loop-Helix Transcription Factors/immunology , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1A1/immunology , Diet/methods , Disease Models, Animal , Enterocolitis, Necrotizing/immunology , Enterocolitis, Necrotizing/pathology , Enterocolitis, Necrotizing/prevention & control , Female , Gene Expression Regulation , Humans , Infant, Newborn , Infant, Premature , Intestinal Mucosa/drug effects , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Ligands , Maternal Exposure , Mice , Pregnancy , Receptors, Aryl Hydrocarbon/agonists , Receptors, Aryl Hydrocarbon/immunology , Signal Transduction , Swine , Toll-Like Receptor 4/immunology
...