Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 852
1.
JCI Insight ; 8(24)2023 Dec 22.
Article En | MEDLINE | ID: mdl-37917177

Control of visceral leishmaniasis (VL) depends on proinflammatory Th1 cells that activate infected tissue macrophages to kill resident intracellular parasites. However, proinflammatory cytokines produced by Th1 cells can damage tissues and require tight regulation. Th1 cell IL-10 production is an important cell-autologous mechanism to prevent such damage. However, IL-10-producing Th1 (type 1 regulatory; Tr1) cells can also delay control of parasites and the generation of immunity following drug treatment or vaccination. To identify molecules to target in order to alter the balance between Th1 and Tr1 cells for improved antiparasitic immunity, we compared the molecular and phenotypic profiles of Th1 and Tr1 cells in experimental VL caused by Leishmania donovani infection of C57BL/6J mice. We also identified a shared Tr1 cell protozoan signature by comparing the transcriptional profiles of Tr1 cells from mice with experimental VL and malaria. We identified LAG3 as an important coinhibitory receptor in patients with VL and experimental VL, and we reveal tissue-specific heterogeneity of coinhibitory receptor expression by Tr1 cells. We also discovered a role for the transcription factor Pbx1 in suppressing CD4+ T cell cytokine production. This work provides insights into the development and function of CD4+ T cells during protozoan parasitic infections and identifies key immunoregulatory molecules.


Interleukin-10 , Protozoan Infections , Th1 Cells , Th1 Cells/immunology , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-10/metabolism , T-Lymphocytes, Regulatory/immunology , Mice, Inbred C57BL , Leishmania donovani , Leishmaniasis, Visceral/immunology , Pre-B-Cell Leukemia Transcription Factor 1/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/parasitology , Protozoan Infections/immunology , Humans , Animals , Mice , Lymphocyte Activation Gene 3 Protein/antagonists & inhibitors , Interferon-gamma/metabolism , Protein Binding , Promoter Regions, Genetic/immunology , Disease Models, Animal
2.
Mol Immunol ; 141: 265-272, 2022 01.
Article En | MEDLINE | ID: mdl-34902807

Targeting the immune checkpoint to inhibit tumor immune escape, which is one of the fundamental causes of cancer, has become an important strategy for cancer treatment. The molecular mechanism of tumor immune escape involved in the process of spontaneous hepatocellular carcinoma after specifically knocking out NFE2L1, the core regulator of redox homeostasis, in the mouse liver is still unclear. Transcriptome data showed that the immunostimulatory TNFSF9/41BBL was significantly reduced in NFE2L1 knockdown hepatocarcinoma HepG2 cells, and this suggests that 41BBL may be an oxidative stress-responsive immune checkpoint. The results of the promoter activity experiment showed that NFE2L1 can promote 41BBL gene transcription activation through the ARE element in the promoter region. In addition, cell biology experiments have found that overexpression of 41BBL can inhibit cell proliferation and promote senescence. Importantly, reactive oxygen species in cells significantly increased after overexpression of 41BBL, whereas NFE2L1 was inhibited, indicating that 41BBL has the effect of feedback regulating oxidative stress in cells. In conclusion, in this study, the transcriptional activation effect of NFE2L1 on 41BBL and the feedback inhibition relationship of 41BBL on NFE2L1 was clarified. The NFE2L1/41BBL axis might be an important pathway that mediates the crosstalk between oxidative stress and the tumor immune response.


4-1BB Ligand/immunology , Antioxidants/metabolism , Carcinoma, Hepatocellular/immunology , Liver Neoplasms/immunology , NF-E2-Related Factor 1/immunology , Oxidative Stress/immunology , Animals , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Cell Proliferation/physiology , Feedback , Gene Expression Regulation/immunology , HEK293 Cells , Hep G2 Cells , Homeostasis/immunology , Humans , Liver Neoplasms/metabolism , Promoter Regions, Genetic/immunology , Reactive Oxygen Species/immunology , Transcription Factors/immunology
4.
Int J Mol Sci ; 22(16)2021 Aug 23.
Article En | MEDLINE | ID: mdl-34445801

The cytoplasmic retinoic acid-inducible gene-I (RIG-I)-like receptors (RLRs) initiate interferon (IFN) production and antiviral gene expression in response to RNA virus infection. Consequently, RLR signalling is tightly regulated by both host and viral factors. Tripartite motif protein 25 (TRIM25) is an E3 ligase that ubiquitinates multiple substrates within the RLR signalling cascade, playing both ubiquitination-dependent and -independent roles in RIG-I-mediated IFN induction. However, additional regulatory roles are emerging. Here, we show a novel interaction between TRIM25 and another protein in the RLR pathway that is essential for type I IFN induction, DEAD-box helicase 3X (DDX3X). In vitro assays and knockdown studies reveal that TRIM25 ubiquitinates DDX3X at lysine 55 (K55) and that TRIM25 and DDX3X cooperatively enhance IFNB1 induction following RIG-I activation, but the latter is independent of TRIM25's catalytic activity. Furthermore, we found that the influenza A virus non-structural protein 1 (NS1) disrupts the TRIM25:DDX3X interaction, abrogating both TRIM25-mediated ubiquitination of DDX3X and cooperative activation of the IFNB1 promoter. Thus, our results reveal a new interplay between two RLR-host proteins that cooperatively enhance IFN-ß production. We also uncover a new and further mechanism by which influenza A virus NS1 suppresses host antiviral defence.


Antiviral Agents/immunology , DEAD Box Protein 58/immunology , DEAD-box RNA Helicases/immunology , Immunity/immunology , Receptors, Immunologic/immunology , Transcription Factors/immunology , Tripartite Motif Proteins/immunology , Ubiquitin-Protein Ligases/immunology , Cell Line , Gene Expression Regulation/immunology , HEK293 Cells , Humans , Influenza A virus/immunology , Interferons/immunology , Promoter Regions, Genetic/immunology , Protein Binding/immunology , Signal Transduction/immunology , Ubiquitination/immunology
5.
Cancer Lett ; 518: 115-126, 2021 10 10.
Article En | MEDLINE | ID: mdl-34098061

Cancer immunotherapies targeting the interaction between Programmed death 1 (PD-1) and Programmed death ligand 1 (PD-L1) have recently been approved for the treatment of multiple cancer types, including gastric cancer. However, not all patients respond to these therapies, while some eventually acquire resistance. A partial predictive biomarker for positive response to PD-1/PD-L1 therapy is PD-L1 expression, which has been shown to be under strict post-transcriptional control in cancer. By fractionating the PD-L1 3' untranslated region (3'UTR) into multiple overlapping fragments, we identified a small 100-nucleotide-long cis-acting region as being necessary and sufficient for post-transcriptional repression of PD-L1 expression in gastric cancer. In parallel, we performed a correlation analysis between PD-L1 expression and all host miRNAs in stomach cancer patient samples. A single miRNA, miR-105-5p, was predicted to bind to the identified cis-acting 3'UTR region and to negatively correlate with PD-L1 expression. Overexpression of miR-105-5p in gastric cancer cell lines resulted in decreased expression of PD-L1, both at the total protein and surface expression levels, and induced CD8+ T cell activation in co-culture assays. Finally, we show that expression of miR-105-5p in gastric cancer is partly controlled by DNA methylation of a cancer- and germline-specific promoter of its host gene, GABRA3. Dysregulation of miR-105-5p is observed in many cancer types and this study shows the importance of this miRNA in controlling the immunogenicity of cancer cells, thus highlighting it as a potential biomarker for PD-1/PD-L1 therapy and target for combinatorial immunotherapy.


B7-H1 Antigen/genetics , MicroRNAs/genetics , Stomach Neoplasms/genetics , Stomach Neoplasms/immunology , 3' Untranslated Regions/genetics , 3' Untranslated Regions/immunology , B7-H1 Antigen/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Line , Cell Line, Tumor , DNA Methylation/genetics , DNA Methylation/immunology , Gene Expression/genetics , Gene Expression/immunology , HEK293 Cells , Humans , MicroRNAs/immunology , Promoter Regions, Genetic/genetics , Promoter Regions, Genetic/immunology
6.
Immunity ; 54(5): 947-961.e8, 2021 05 11.
Article En | MEDLINE | ID: mdl-33930308

The transcription factor Foxp3 plays crucial roles for Treg cell development and function. Conserved non-coding sequences (CNSs) at the Foxp3 locus control Foxp3 transcription, but how they developmentally contribute to Treg cell lineage specification remains obscure. Here, we show that among Foxp3 CNSs, the promoter-upstream CNS0 and the intergenic CNS3, which bind distinct transcription factors, were activated at early stages of thymocyte differentiation prior to Foxp3 promoter activation, with sequential genomic looping bridging these regions and the promoter. While deletion of either CNS0 or CNS3 partially compromised thymic Treg cell generation, deletion of both completely abrogated the generation and impaired the stability of Foxp3 expression in residual Treg cells. As a result, CNS0 and CNS3 double-deleted mice succumbed to lethal systemic autoimmunity and inflammation. Thus, hierarchical and coordinated activation of Foxp3 CNS0 and CNS3 initiates and stabilizes Foxp3 gene expression, thereby crucially controlling Treg cell development, maintenance, and consequently immunological self-tolerance.


Enhancer Elements, Genetic/immunology , Forkhead Transcription Factors/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cell Differentiation/immunology , Cell Lineage/immunology , Gene Expression Regulation/immunology , Humans , Immune Tolerance/immunology , Lymphocyte Activation/immunology , Male , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic/immunology , Self Tolerance/immunology
7.
Nat Commun ; 12(1): 537, 2021 01 22.
Article En | MEDLINE | ID: mdl-33483487

Targeting chromatin regulators to specific genomic locations for gene control is emerging as a powerful method in basic research and synthetic biology. However, many chromatin regulators are large, making them difficult to deliver and combine in mammalian cells. Here, we develop a strategy for gene control using small nanobodies that bind and recruit endogenous chromatin regulators to a gene. We show that an antiGFP nanobody can be used to simultaneously visualize GFP-tagged chromatin regulators and control gene expression, and that nanobodies against HP1 and DNMT1 can silence a reporter gene. Moreover, combining nanobodies together or with other regulators, such as DNMT3A or KRAB, can enhance silencing speed and epigenetic memory. Finally, we use the slow silencing speed and high memory of antiDNMT1 to build a signal duration timer and recorder. These results set the basis for using nanobodies against chromatin regulators for controlling gene expression and epigenetic memory.


Chromatin/immunology , Epigenesis, Genetic/immunology , Gene Expression Regulation/immunology , Promoter Regions, Genetic/immunology , Single-Domain Antibodies/immunology , Algorithms , Animals , Chromatin/genetics , Gene Silencing/immunology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/immunology , Green Fluorescent Proteins/metabolism , Humans , Models, Genetic , Promoter Regions, Genetic/genetics , Signal Transduction/genetics , Signal Transduction/immunology , Single-Domain Antibodies/metabolism
8.
Front Immunol ; 12: 694105, 2021.
Article En | MEDLINE | ID: mdl-35069519

Filovirus family consists of highly pathogenic viruses that have caused fatal outbreaks especially in many African countries. Previously, research focus has been on Ebola, Sudan and Marburg viruses leaving other filoviruses less well studied. Filoviruses, in general, pose a significant global threat since they are highly virulent and potentially transmissible between humans causing sporadic infections and local or widespread epidemics. Filoviruses have the ability to downregulate innate immunity, and especially viral protein 24 (VP24), VP35 and VP40 have variably been shown to interfere with interferon (IFN) gene expression and signaling. Here we systematically analyzed the ability of VP24 proteins of nine filovirus family members to interfere with retinoic acid-inducible gene I (RIG-I) and melanoma differentiation-associated antigen 5 (MDA5) induced IFN-ß and IFN-λ1 promoter activation. All VP24 proteins were localized both in the cell cytoplasm and nucleus in variable amounts. VP24 proteins of Zaire and Sudan ebolaviruses, Lloviu, Taï Forest, Reston, Marburg and Bundibugyo viruses (EBOV, SUDV, LLOV, TAFV, RESTV, MARV and BDBV, respectively) were found to inhibit both RIG-I and MDA5 stimulated IFN-ß and IFN-λ1 promoter activation. The inhibition takes place downstream of interferon regulatory factor 3 phosphorylation suggesting the inhibition to occur in the nucleus. VP24 proteins of Mengla (MLAV) or Bombali viruses (BOMV) did not inhibit IFN-ß or IFN-λ1 promoter activation. Six ebolavirus VP24s and Lloviu VP24 bound tightly, whereas MARV and MLAV VP24s bound weakly, to importin α5, the subtype that regulates the nuclear import of STAT complexes. MARV and MLAV VP24 binding to importin α5 was very weak. Our data provides new information on the innate immune inhibitory mechanisms of filovirus VP24 proteins, which may contribute to the pathogenesis of filovirus infections.


DEAD Box Protein 58/immunology , Filoviridae/immunology , Interferon Type I/immunology , Interferon-Induced Helicase, IFIH1/immunology , Interferons/immunology , Interleukins/immunology , Promoter Regions, Genetic/immunology , Receptors, Immunologic/immunology , Viral Proteins/immunology , Cell Line, Tumor , DEAD Box Protein 58/genetics , Filoviridae/genetics , Gene Expression Regulation/immunology , HEK293 Cells , Humans , Interferon Type I/genetics , Interferon-Induced Helicase, IFIH1/genetics , Interferons/genetics , Interleukins/genetics , Receptors, Immunologic/genetics , Viral Proteins/genetics
9.
Front Immunol ; 12: 722188, 2021.
Article En | MEDLINE | ID: mdl-35111150

Protein arginine transferase 5 (PRMT5) has been implicated as an important modulator of tumorigenesis as it promotes tumor cell proliferation, invasion, and metastasis. Studies have largely focused on PRMT5 regulating intrinsic changes in tumors; however, the effects of PRMT5 on the tumor microenvironment and particularly immune cells are largely unknown. Here we found that targeting PRMT5 by genetic or pharmacological inhibition reduced lung tumor progression in immunocompromised mice; however, the effects were weakened in immunocompetent mice. PRMT5 inhibition not only decreased tumor cell survival but also increased the tumor cell expression of CD274 in vitro and in vivo, which activated the PD1/PD-L1 axis and eliminated CD8+T cell antitumor immunity. Mechanistically, PRMT5 regulated CD274 gene expression through symmetric dimethylation of histone H4R3, increased deposition of H3R4me2s on CD274 promoter loci, and inhibition of CD274 gene expression. Targeting PRMT5 reduced this inhibitory effect and promoted CD274 expression in lung cancer. However, PRMT5 inhibitors represent a double-edged sword as they may selectively kill cancer cells but may also disrupt the antitumor immune response. The combination of PRMT5 inhibition and ani-PD-L1 therapy resulted in an increase in the number and enhanced the function of tumor-infiltrating T cells. Our findings address an unmet clinical need in which combining PRMT5 inhibition with anti-PD-L1 therapy could be a promising strategy for lung cancer treatment.


B7-H1 Antigen/genetics , Lung Neoplasms/genetics , Protein-Arginine N-Methyltransferases/genetics , Animals , B7-H1 Antigen/immunology , CD8-Positive T-Lymphocytes/immunology , Carcinogenesis/genetics , Carcinogenesis/immunology , Cell Line, Tumor , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/immunology , Humans , Lung Neoplasms/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Promoter Regions, Genetic/genetics , Promoter Regions, Genetic/immunology , Protein-Arginine N-Methyltransferases/immunology , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Xenograft Model Antitumor Assays/methods
10.
Front Immunol ; 12: 771216, 2021.
Article En | MEDLINE | ID: mdl-35058921

Chronic asthma is characterized by airway inflammation and irreversible airway remodeling. Epithelial-mesenchymal transition (EMT) is a typical pathological change of airway remodeling. Our previous research demonstrated miR-23b inhibited airway smooth muscle proliferation while the function of miR-23b-3p has not been reported yet. Besides, miRNA is regulated by many factors, including DNA methylation. The function of miR-23b-3p and whether it is regulated by DNA methylation are worth exploring. Balb/c mice were given OVA sensitization to develop the asthmatic model. Expression of miR-23b-3p and EMT markers were measured by RT-qPCR, WB and immunohistochemistry (IHC). DNA methylation was detected by methylation-specific PCR (MSP) and the MassARRAY System. Asthmatic mice and TGF-ß1-stimulated bronchial epithelial cells (BEAS-2B) showed EMT with increased miR-23b-3p. Overexpression of miR-23b-3p promoted EMT and migration, while inhibition of miR-23b-3p reversed these transitions. DNA methyltransferases were decreased in asthmatic mice. MSP and MassARRAY System detected the promotor of miR-23b showed DNA hypomethylation. DNA methyltransferase inhibitor 5'-AZA-CdZ increased the expression of miR-23b-3p. Meanwhile, PTEN was identified as a target gene of miR-23b-3p. Our results indicated that promotor hypomethylation mediated upregulation of miR-23b-3p targets PTEN to promote EMT in chronic asthma. miR-23b-3p and DNA methylation might be potential therapeutic targets for irreversible airway remodeling.


Asthma/immunology , Bronchi/immunology , DNA Methylation/immunology , Epithelial-Mesenchymal Transition/immunology , MicroRNAs/immunology , PTEN Phosphohydrolase/immunology , Promoter Regions, Genetic/immunology , Animals , Asthma/genetics , Cell Line , Chronic Disease , Humans , Male , Mice , Mice, Inbred BALB C , MicroRNAs/genetics , PTEN Phosphohydrolase/genetics
11.
PLoS Pathog ; 16(6): e1008555, 2020 06.
Article En | MEDLINE | ID: mdl-32579593

Exhaustion is a dysfunctional state of cytotoxic CD8+ T cells (CTL) observed in chronic infection and cancer. Current in vivo models of CTL exhaustion using chronic viral infections or cancer yield very few exhausted CTL, limiting the analysis that can be done on these cells. Establishing an in vitro system that rapidly induces CTL exhaustion would therefore greatly facilitate the study of this phenotype, identify the truly exhaustion-associated changes and allow the testing of novel approaches to reverse or prevent exhaustion. Here we show that repeat stimulation of purified TCR transgenic OT-I CTL with their specific peptide induces all the functional (reduced cytokine production and polyfunctionality, decreased in vivo expansion capacity) and phenotypic (increased inhibitory receptors expression and transcription factor changes) characteristics of exhaustion. Importantly, in vitro exhausted cells shared the transcriptomic characteristics of the gold standard of exhaustion, CTL from LCMV cl13 infections. Gene expression of both in vitro and in vivo exhausted CTL was distinct from T cell anergy. Using this system, we show that Tcf7 promoter DNA methylation contributes to TCF1 downregulation in exhausted CTL. Thus this novel in vitro system can be used to identify genes and signaling pathways involved in exhaustion and will facilitate the screening of reagents that prevent/reverse CTL exhaustion.


CD8-Positive T-Lymphocytes/immunology , DNA Methylation/immunology , Hepatocyte Nuclear Factor 1-alpha/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Promoter Regions, Genetic/immunology , Animals , CD8-Positive T-Lymphocytes/pathology , Hepatocyte Nuclear Factor 1-alpha/genetics , Lymphocytic Choriomeningitis/genetics , Lymphocytic Choriomeningitis/pathology , Lymphocytic choriomeningitis virus/genetics , Mice , Mice, Transgenic , Signal Transduction/genetics , Signal Transduction/immunology
12.
Cytokine ; 133: 155161, 2020 09.
Article En | MEDLINE | ID: mdl-32531745

Mammalian interferon-induced protein with tetratricopeptide repeats family proteins (IFITs) play important roles in host innate immune response to viruses. Recently, studies have shown that IFIT from poultry also plays a crucial part in antiviral function. This study first reports the regulation of duck Tembusu virus (DTMUV) replication by IFIT5 and the effect of duck IFIT5 (duIFIT5) on the innate immune response after DTMUV infection. Firstly, duIFIT5 was obviously increased in duck embryo fibroblast cells (DEFs) infected with DTMUV. Compared to the negative control, we found that in the duIFIT5-overexpressing group, the DTMUV titer at 24 h post infection (hpi) was significantly reduced, but the viral titer was strikingly increased at 48 hpi. Moreover, overexpression of duIFIT5 could significantly inhibit IFN-ß transcription and IFN-ß promoter activation at indicated time points after DTMUV infection. Further, in DTMUV-infected or poly(I:C)-stimulated DEFs, overexpression of duIFIT5 also significantly inhibited the activation of NF-κB and IRF7 promoters, as well as the activation of downstream IFN induced the interferon-stimulated response element (ISRE) promoter. Meanwhile, the transcription level of antiviral protein Mx, but not OASL, was obviously decreased at various time points. The opposite results were obtained by knockdown of duIFIT5 in DTMUV-infected or poly(I:C)-stimulated DEFs. Compared to the negative control, knockdown of duIFIT5 promoted DTMUV titer and DTMUV envelope (E) protein expression at 24 hpi, but DTMUV titer and E protein expression was markedly decreased at 48 hpi. Additionally, the promoters of IFN-ß, NF-κB, IRF7 and ISRE were significantly activated in the duIFIT5 knockdown group. Collectively, duIFIT5 differentially regulates DTMUV replication and inhibits virus-triggered innate immune response.


Flavivirus/immunology , Immunity, Innate/immunology , Neoplasm Proteins/immunology , Virus Replication/immunology , Animals , Antiviral Agents/immunology , Ducks , Fibroblasts/immunology , Interferon-beta/immunology , NF-kappa B/immunology , Poly I-C/immunology , Promoter Regions, Genetic/immunology , Signal Transduction/immunology
13.
J Leukoc Biol ; 108(4): 1329-1337, 2020 10.
Article En | MEDLINE | ID: mdl-32421902

CD8 T cells play a crucial role in immune responses to virus infections and tumors. Naïve CD8 T lymphocytes after TCR stimulation undergo differentiation into CTLs and memory cells, which are essential sources of IFN-γ. We investigated IFN-γ production by CD8 T cell subsets found in nonimmune mice. A minor fraction of in vitro TCR-stimulated CD8 T cells produce IFN-γ, and it is regulated at the transcriptional level. Antigen inexperienced C57BL/6 mice present the coexistence of 2 populations. The main population exhibits a CD44low CD122low profile, which is compatible with naïve lymphocytes. The minor expresses a phenotype of immunologic memory, CD44hi CD122hi . Both subsets are able to produce IL-2 in response to TCR activation, but only the memory-like population is responsible for IFN-γ production. Similar to memory CD8 T cells, CD44hi CD8+ T cells also present a higher level of the transcriptional factor Eomes and a lower level of T-bet (Tbx21) mRNA than CD44low CD8+ T cells. The presence of the CD44hi CD8+ T cell population in nonimmune OT-I transgenic mice reveals that the population is generated independently of antigenic stimulation. CpG methylation is an efficient epigenetic mechanism for gene silencing. DNA methylation at posttranscriptional CpG sites in the Ifng promoter is higher in CD44low CD8+ T cells than in CD44hi CD8+ T cells. Thus, memory-like CD8 T cells have a distinct epigenetic pattern in the Ifng promoter and can rapidly produce IFN-γ in response to TCR stimulation.


CD8-Positive T-Lymphocytes/immunology , Immunologic Memory , Interferon-gamma/immunology , Animals , CD8-Positive T-Lymphocytes/cytology , CpG Islands/immunology , Hyaluronan Receptors/genetics , Hyaluronan Receptors/immunology , Interferon-gamma/genetics , Interleukin-2 Receptor beta Subunit/genetics , Interleukin-2 Receptor beta Subunit/immunology , Male , Mice , Mice, Transgenic , Promoter Regions, Genetic/genetics , Promoter Regions, Genetic/immunology , T-Box Domain Proteins/genetics , T-Box Domain Proteins/immunology
14.
Fish Shellfish Immunol ; 101: 99-105, 2020 Jun.
Article En | MEDLINE | ID: mdl-32224278

Interferons play an important role in the fish innate immune system against viral infection by inducing the interferon stimulated genes, such as Mx gene. We cloned the MxII gene promoter from orange-spotted grouper and found three MxII gene promoters. All of them contained two interferon stimulated response elements (ISREs), and three dinucleotide repeat sequences located at 5' end of ISREs. Interestingly, there is a polymorphic GT repeat element located upstream of these ISREs. The three MxII gene promoters respectively contained 27, 29, and 31 GT repeats, namely EcMx_27, EcMx_29, and EcMx_31. To determine whether GT repeat element influence the MxII gene expression, the MxII gene promoters were subcloned into promoterless reporter plasmid and transfected into grouper kidney (GK) cells. The results showed that a significant induction by poly(I:C) was detected in GK cells transfected with pEcMx_31 (2.65 folds) whereas there was no induction in GK cells transfected with pEcMx_27 and pEcMx_29. However, the significant induction by nervous necrosis virus (NNV) infection was found in GK cells separately transfected with three reporter plasmids. These results suggest that the GT repeat element plays an important role in modulation of MxII gene expression and the induction by poly(I:C) and NNV may be mediated through different signal transduction pathways.


Bass/immunology , Fish Diseases/immunology , Poly I-C/administration & dosage , Polymorphism, Genetic , Promoter Regions, Genetic/immunology , RNA Virus Infections/veterinary , Animals , Kidney/metabolism , Nodaviridae/physiology , RNA Virus Infections/immunology
15.
J Immunol ; 204(9): 2503-2513, 2020 05 01.
Article En | MEDLINE | ID: mdl-32205424

Macrophages are critical for the initiation and resolution of the inflammatory phase of wound healing. In diabetes, macrophages display a prolonged inflammatory phenotype preventing tissue repair. TLRs, particularly TLR4, have been shown to regulate myeloid-mediated inflammation in wounds. We examined macrophages isolated from wounds of patients afflicted with diabetes and healthy controls as well as a murine diabetic model demonstrating dynamic expression of TLR4 results in altered metabolic pathways in diabetic macrophages. Further, using a myeloid-specific mixed-lineage leukemia 1 (MLL1) knockout (Mll1f/fLyz2Cre+ ), we determined that MLL1 drives Tlr4 expression in diabetic macrophages by regulating levels of histone H3 lysine 4 trimethylation on the Tlr4 promoter. Mechanistically, MLL1-mediated epigenetic alterations influence diabetic macrophage responsiveness to TLR4 stimulation and inhibit tissue repair. Pharmacological inhibition of the TLR4 pathway using a small molecule inhibitor (TAK-242) as well as genetic depletion of either Tlr4 (Tlr4-/- ) or myeloid-specific Tlr4 (Tlr4f/fLyz2Cre+) resulted in improved diabetic wound healing. These results define an important role for MLL1-mediated epigenetic regulation of TLR4 in pathologic diabetic wound repair and suggest a target for therapeutic manipulation.


Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/immunology , Epigenesis, Genetic/genetics , Macrophages/physiology , Toll-Like Receptor 4/genetics , Wound Healing/genetics , Aged , Animals , Epigenesis, Genetic/immunology , Female , Histones/genetics , Histones/immunology , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation Mediators/immunology , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Myeloid-Lymphoid Leukemia Protein/genetics , Myeloid-Lymphoid Leukemia Protein/immunology , Promoter Regions, Genetic/genetics , Promoter Regions, Genetic/immunology , Toll-Like Receptor 4/immunology , Wound Healing/immunology
16.
J Immunol ; 204(9): 2589-2599, 2020 05 01.
Article En | MEDLINE | ID: mdl-32198140

S100A8 is a damage-associated molecular pattern protein released by monocytes, playing a decisive role in the development of inflammation. Nonresolving inflammation is viewed as a driving force in tumorigenesis, and its role in tumor immune escape also attracted attentions. PD-1/PD-L1 axis is a critical determinant of physiological immune homeostasis, and anti-PD-1 or PD-L1 therapy has becoming the most exciting field of oncology. Multiple regulation mechanisms have been contributed to PD-L1 expression modulation including inflammatory mediators. In this study we reported that S100A8 significantly induced PD-L1 expression in monocytes/macrophages but not in tumor cells. S100A8 induced PD-L1 transcription through the TLR4 receptor and multiple crucial pathways of inflammation process. S100A8 modulated the histone modification of the PD-L1 promoter in monocytes/macrophages. S100A8-pretreated macrophages had immunosuppressive function and attenuated the antitumor ability of CTLs both in vitro and in vivo. A highly positive correlation existed between S100A8 expression and PD-L1 expression in human cancer specimens. To our knowledge, our study uncovers a novel molecular mechanism for regulating PD-L1 transcription by an inflammatory mediator S100A8, and reveals the importance of comprehensive understanding the role of inflammation in tumorigenesis as well as in tumor immune escape.


B7-H1 Antigen/immunology , Calgranulin A/immunology , Inflammation/immunology , Macrophages/immunology , Neoplasms/genetics , Tumor Escape/immunology , Animals , Carcinogenesis/immunology , Cell Line , Cell Line, Tumor , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, SCID , Monocytes/immunology , Promoter Regions, Genetic/immunology , RAW 264.7 Cells , THP-1 Cells , Toll-Like Receptor 4/immunology , Transcription, Genetic/immunology
17.
J Immunol ; 204(5): 1214-1224, 2020 03 01.
Article En | MEDLINE | ID: mdl-31980574

Leukocytes are rapidly recruited to sites of inflammation via interactions with the vascular endothelium. The steroid hormone dehydroepiandrosterone (DHEA) exerts anti-inflammatory properties; however, the underlying mechanisms are poorly understood. In this study, we show that an anti-inflammatory mechanism of DHEA involves the regulation of developmental endothelial locus 1 (DEL-1) expression. DEL-1 is a secreted homeostatic factor that inhibits ß2-integrin-dependent leukocyte adhesion, and the subsequent leukocyte recruitment and its expression is downregulated upon inflammation. Similarly, DHEA inhibited leukocyte adhesion to the endothelium in venules of the inflamed mouse cremaster muscle. Importantly, in a model of lung inflammation, DHEA limited neutrophil recruitment in a DEL-1-dependent manner. Mechanistically, DHEA counteracted the inhibitory effect of inflammation on DEL-1 expression. Indeed, whereas TNF reduced DEL-1 expression and secretion in endothelial cells by diminishing C/EBPß binding to the DEL-1 gene promoter, DHEA counteracted the inhibitory effect of TNF via activation of tropomyosin receptor kinase A (TRKA) and downstream PI3K/AKT signaling that restored C/EBPß binding to the DEL-1 promoter. In conclusion, DHEA restrains neutrophil recruitment by reversing inflammation-induced downregulation of DEL-1 expression. Therefore, the anti-inflammatory DHEA/DEL-1 axis could be harnessed therapeutically in the context of inflammatory diseases.


Calcium-Binding Proteins/immunology , Cell Adhesion Molecules/immunology , Dehydroepiandrosterone/pharmacology , Leukocytes/immunology , Signal Transduction/immunology , Animals , CCAAT-Enhancer-Binding Protein-beta/immunology , CD18 Antigens/immunology , Cell Adhesion/immunology , Endothelium, Vascular/immunology , Female , Gene Expression Regulation/immunology , Leukocytes/cytology , Mice , Phosphatidylinositol 3-Kinases/immunology , Promoter Regions, Genetic/immunology , Proto-Oncogene Proteins c-akt/immunology , Receptor, trkA/immunology
18.
J Immunol ; 204(5): 1201-1213, 2020 03 01.
Article En | MEDLINE | ID: mdl-31932499

Vitamin D can modulate the innate and adaptive immune system. Vitamin D deficiency has been associated with various autoimmune diseases. Th9 cells are implicated in the pathogenesis of numerous autoimmune diseases. Thus, we investigated the role of calcitriol (active metabolite of vitamin D) in the regulation of Th9 cell differentiation. In this study, we have unraveled the molecular mechanisms of calcitriol-mediated regulation of Th9 cell differentiation. Calcitriol significantly diminished IL-9 secretion from murine Th9 cells associated with downregulated expression of the Th9-associated transcription factor, PU.1. Ectopic expression of VDR in Th9 cells attenuated the percentage of IL-9-secreting cells. VDR associated with PU.1 in Th9 cells. Using a series of mutations, we were able to dissect the VDR domain involved in the regulation of the Il9 gene. The VDR-PU.1 interaction prevented the accessibility of PU.1 to the Il9 gene promoter, thereby restricting its expression. However, the expression of Foxp3, regulatory T cell-specific transcription factor, was enhanced in the presence of calcitriol in Th9 cells. When Th9 cells are treated with both calcitriol and trichostatin A (histone deacetylase inhibitor), the level of IL-9 reached to the level of wild-type untreated Th9 cells. Calcitriol attenuated specific histone acetylation at the Il9 gene. In contrast, calcitriol enhanced the recruitment of the histone modifier HDAC1 at the Il9 gene promoter. In summary, we have identified that calcitriol blocked the access of PU.1 to the Il9 gene by reducing its expression and associating with it as well as regulated the chromatin of the Il9 gene to regulate expression.


Calcitriol/pharmacology , Cell Differentiation/drug effects , Gene Expression Regulation/drug effects , Histone Deacetylase 1/immunology , Interleukin-9/immunology , Proto-Oncogene Proteins/immunology , T-Lymphocytes, Regulatory/immunology , Trans-Activators/immunology , Acetylation/drug effects , Animals , Cell Differentiation/immunology , Female , Gene Expression Regulation/immunology , Histones/immunology , Mice , Promoter Regions, Genetic/immunology , Receptors, Calcitriol/immunology , T-Lymphocytes, Regulatory/cytology
19.
J Immunol ; 204(5): 1173-1187, 2020 03 01.
Article En | MEDLINE | ID: mdl-31996458

Homogeneous populations of mature differentiated primary cell types can display variable responsiveness to extracellular stimuli, although little is known about the underlying mechanisms that govern such heterogeneity at the level of gene expression. In this article, we show that morphologically homogenous human endothelial cells exhibit heterogeneous expression of VCAM1 after TNF-α stimulation. Variability in VCAM1 expression was not due to stochasticity of intracellular signal transduction but rather to preexisting established heterogeneous states of promoter DNA methylation that were generationally conserved through mitosis. Variability in DNA methylation of the VCAM1 promoter resulted in graded RelA/p65 and RNA polymerase II binding that gave rise to a distribution of VCAM1 transcription in the population after TNF-α stimulation. Microarray analysis and single-cell RNA sequencing revealed that a number of cytokine-inducible genes shared this heterogeneous response pattern. These results show that heritable epigenetic heterogeneity is fundamental in inflammatory signaling and highlight VCAM1 as a metastable epiallele.


Epigenesis, Genetic/immunology , Human Umbilical Vein Endothelial Cells/immunology , Human Umbilical Vein Endothelial Cells/cytology , Humans , Promoter Regions, Genetic/immunology , RNA Polymerase II/genetics , RNA Polymerase II/immunology , Transcription Factor RelA/genetics , Transcription Factor RelA/immunology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/immunology
20.
Iran J Allergy Asthma Immunol ; 19(5): 509-516, 2020 Oct 18.
Article En | MEDLINE | ID: mdl-33463118

Vitamin D plays a variety of physiological functions, such as regulating mineral homeostasis. More recently, it has emerged as an immunomodulator player, affecting several types of immune cells, such as regulatory T (Treg) cells. It has been reported that vitamin D exerts some mediatory effects through an epigenetic mechanism. In this study, the impacts of calcitriol, the active form of vitamin D, on the methylation of the conserved non-coding sequence 2 (CNS2) region of the forkhead box P3 (Foxp3) gene promoter, were evaluated. Fourteen C57BL/6 mice were recruited in this study and divided into two intervention and control groups. The CD4+ T cells were isolated from mice splenocytes. The expression of Foxp3, IL-10, and transforming growth factor-beta (TGF-ß1) genes were relatively quantified by real-time PCR technique, and the DNA methylation percentage of every CpG site in the CNS2 region was measured individually by bisulfite-sequencing PCR. Vitamin D Intervention significantly (p<0.05) could increase the expression of Foxp3, IL-10, and TGF-ß1 gene in the CD4+ T cells of mice comparing with the control group. Meanwhile, methylation of the CNS2 region of Foxp3 promoter was significantly decreased in three of ten CpG sites in the vitamin D group compared to the control group. The results of this study showed that vitamin D can engage the methylation process to induce Foxp3 gene expression and probably Treg cytokines profile. Further researches are needed to discover the precise epigenetic mechanisms by which vitamin D modulates the immune system.


CD4-Positive T-Lymphocytes/immunology , Calcitriol/immunology , DNA Methylation/immunology , Forkhead Transcription Factors/immunology , Immunologic Factors/immunology , Animals , Epigenesis, Genetic/immunology , Female , Interleukin-10/immunology , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic/immunology , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta/immunology , Transforming Growth Factor beta1/immunology
...