Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.868
Filter
1.
Methods Mol Biol ; 2854: 253-264, 2025.
Article in English | MEDLINE | ID: mdl-39192135

ABSTRACT

Innate immunity is an important defense barrier for the human body. After viral pathogen-associated molecular patterns (PAMPs) are detected by host-pathogen recognition receptors (PRRs), the associated signaling pathways trigger the activation of the interferon (IFN) regulatory factor (IRF) family members and the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB). However, any gene defects among the signaling adaptors will compromise innate immune efficiency. Therefore, investigating genetic defects in the antiviral innate immune signaling pathway is important. We summarize the commonly used research methods related to antiviral immune gene defects and outline the relevant research protocols, which will help investigators study antiviral innate immunity.


Subject(s)
Immunity, Innate , Signal Transduction , Humans , Animals , Virus Diseases/immunology , Virus Diseases/genetics , Host-Pathogen Interactions/immunology , Host-Pathogen Interactions/genetics , NF-kappa B/metabolism , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , Receptors, Pattern Recognition/metabolism , Receptors, Pattern Recognition/genetics
2.
Cells ; 13(17)2024 Aug 31.
Article in English | MEDLINE | ID: mdl-39273037

ABSTRACT

The microenvironment of a cancer stem cell (CSC) niche is often found in coexistence with cancer-associated fibroblasts (CAFs). Here, we show the first in-depth analysis of the interaction between primary triple-negative breast cancer stem cells (BCSCs) with fibroblasts. Using 2D co-culture models with specific seeding ratios, we identified stromal fibroblast aggregation at the BCSC cluster periphery, and, on closer observation, the aggregated fibroblasts was found to encircle BCSC clusters in nematic organization. In addition, collagen type I and fibronectin accumulation were also found at the BCSC-stromal periphery. MACE-Seq analysis of BCSC-encapsulating fibroblasts displayed the transformation of stromal fibroblasts to CAFs and the upregulation of fibrosis regulating genes of which the Interferon Regulatory Factor 6 (IRF6) gene was identified. Loss of function experiments with the IRF6 gene decreased fibroblast encapsulation around BCSC clusters in 2D co-cultures. In BCSC xenografts, fibroblast IRF6 expression led to an increase in the stromal area and fibroblast density in tumors, in addition to a reduction in necrotic growth. Based on our findings, we propose that fibroblast IRF6 function is an important factor in the development of the stromal microenvironment and in sustaining the BCSC tumor niche.


Subject(s)
Coculture Techniques , Fibroblasts , Interferon Regulatory Factors , Neoplastic Stem Cells , Stromal Cells , Tumor Microenvironment , Up-Regulation , Humans , Female , Interferon Regulatory Factors/metabolism , Interferon Regulatory Factors/genetics , Stromal Cells/metabolism , Stromal Cells/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Animals , Fibroblasts/metabolism , Fibroblasts/pathology , Up-Regulation/genetics , Mice , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Breast Neoplasms/pathology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor
3.
Adv Immunol ; 163: 51-96, 2024.
Article in English | MEDLINE | ID: mdl-39271259

ABSTRACT

Interferon regulatory factor-8 (IRF8) is the lineage determining transcription factor for the type one classical dendritic cell (cDC1) subset, a terminal selector for plasmacytoid dendritic cells and important for the function of monocytes. Studies of Irf8 gene regulation have identified several enhancers controlling its activity during development of progenitors in the bone marrow that precisely regulate expression at distinct developmental stages. Each enhancer responds to distinct transcription factors that are expressed at each stage. IRF8 is first expressed in early progenitors that form the monocyte dendritic cell progenitor (MDP) in response to induction of the transcription factor CCAAT/enhancer-binding protein alpha (C/EBPα) acting at the Irf8 +56 kb enhancer. IRF8 levels increase further as the MDP transits into the common dendritic cell progenitor (CDP) in response to E protein activity at the Irf8 +41 kb enhancer. Upon Nfil3-induction in CDPs leading to specification of the cDC1 progenitor, abrupt induction of BATF3 forms the JUN/BATF3/IRF8 heterotrimer that activates the Irf8 +32 kb enhancer that sustains Irf8 autoactivation throughout the cDC1 lifetime. Deletions of each of these enhancers has revealed their stage dependent activation. Surprisingly, studies of compound heterozygotes for each combination of enhancer deletions revealed that activation of each subsequent enhancer requires the successful activation of the previous enhancer in strictly cis-dependent mechanism. Successful progression of enhancer activation is finely tuned to alter the functional accessibility of subsequent enhancers to factors active in the next stage of development. The molecular basis for these phenomenon is still obscure but could have implications for genomic regulation in a broader developmental context.


Subject(s)
Dendritic Cells , Enhancer Elements, Genetic , Interferon Regulatory Factors , Humans , Interferon Regulatory Factors/metabolism , Interferon Regulatory Factors/genetics , Animals , Dendritic Cells/immunology , Dendritic Cells/metabolism , Enhancer Elements, Genetic/genetics , Cell Differentiation , Gene Expression Regulation , Super Enhancers
4.
Cell Commun Signal ; 22(1): 395, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39123188

ABSTRACT

Esophageal cancer is common worldwide, with ESCC being the most frequent tumor in East Asia. Tumor-associated macrophages are an important component of the ESCC microenvironment. SUMOylation is a post-translational modification of proteins, and SUMO-specific proteases (SENPs) play an important role in de-SUMOylation. In human patients, we discovered that the levels of SENP3 were upregulated in the tumor-associated macrophages. Furthermore, the loss of SENP3 enhanced the alternative activation of macrophages in the 4-NQO-induced ESCC mice model. This is the first study to identify SENP3-mediated macrophage polarization via the de-SUMOylation of interferon regulatory factor 4 (IRF4) at the K349 site. Alternative activation of macrophages increases the migration and invasion potential of ESCC cells and promotes their progression in vivo. Moreover, patients with relatively low SENP3 expression in macrophages exhibit higher primary PET SUVmax value and lymph node metastasis rates. In summary, this study revealed that SENP3-mediated IRF4 de-SUMOylation is crucial for the alternative activation of macrophages and influences the progression of ESCC.


Subject(s)
Cysteine Endopeptidases , Interferon Regulatory Factors , Macrophage Activation , Sumoylation , Animals , Female , Humans , Male , Mice , Cell Line, Tumor , Cell Movement , Cysteine Endopeptidases/metabolism , Cysteine Endopeptidases/genetics , Disease Progression , Esophageal Squamous Cell Carcinoma/pathology , Esophageal Squamous Cell Carcinoma/metabolism , Esophageal Squamous Cell Carcinoma/genetics , Interferon Regulatory Factors/metabolism , Interferon Regulatory Factors/genetics , Macrophages/metabolism , Tumor-Associated Macrophages/metabolism
5.
J Med Virol ; 96(8): e29879, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39169736

ABSTRACT

Interferon regulatory factor 5 (IRF5) is a key transcription factor in inflammatory and immune responses, with its dysregulation linked to autoimmune diseases. Using bioinformatic approaches, including Basic Local Alignment Search Tool (BLAST) for sequence similarity searches, BLAST-Like Alignment Tool (BLAT) for genome-wide alignments, and several phylogenetics software, such as Multiple Alignment using Fast Fourier Transform (MAFFT), for phylogenetic analyses, we characterized the structure, origin, and evolutionary history of the human IRF5 pseudogene 1 (IRF5P1). Our analyses reveal that IRF5P1 is a chimeric processed pseudogene containing sequences derived from multiple sources, including IRF5-like sequences from disparate organisms. We find that IRF5P1 is specific to higher primates, likely originating through an ancient retroviral integration event approximately 60 million years ago. Interestingly, IRF5P1 resides within the triple QxxK/R motif-containing (TRIQK) gene, and its antisense strand is predominantly expressed as part of the TRIQK pre-messenger RNA (mRNA). Analysis of publicly available RNA-seq data suggests potential expression of antisense IRF5P1 RNA. We hypothesize that this antisense RNA may regulate IRF5 expression through complementary binding to IRF5 mRNA, with human genetic variants potentially modulating this interaction. The conservation of IRF5P1 in the primate lineage suggests its positive effects on primate evolution and innate immunity. This study highlights the importance of investigating pseudogenes and their potential regulatory roles in shaping lineage-specific immune adaptations.


Subject(s)
Evolution, Molecular , Interferon Regulatory Factors , Phylogeny , Primates , Pseudogenes , Pseudogenes/genetics , Animals , Humans , Interferon Regulatory Factors/genetics , Primates/genetics , Computational Biology/methods , Sequence Alignment
6.
Nat Immunol ; 25(9): 1663-1677, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39179932

ABSTRACT

Early B cell lymphopoiesis depends on E2A, Ebf1, Pax5 and Ikaros family members. In the present study, we used acute protein degradation in mice to identify direct target genes of these transcription factors in pro-B, small pre-B and immature B cells. E2A, Ebf1 and Pax5 predominantly function as transcriptional activators by inducing open chromatin at their target genes, have largely unique functions and are essential for early B cell maintenance. Ikaros and Aiolos act as dedicated repressors to cooperatively control early B cell development. The surrogate light-chain genes Igll1 and Vpreb1 are directly activated by Ebf1 and Pax5 in pro-B cells and directly repressed by Ikaros and Aiolos in small pre-B cells. Pax5 and E2A contribute to V(D)J recombination by activating Rag1, Rag2, Dntt, Irf4 and Irf8. Similar to Pax5, Ebf1 also represses the cohesin-release factor gene Wapl to mediate prolonged loop extrusion across the Igh locus. In summary, in vivo protein degradation has provided unprecedented insight into the control of early B cell lymphopoiesis by five transcription factors.


Subject(s)
B-Lymphocytes , Basic Helix-Loop-Helix Transcription Factors , Ikaros Transcription Factor , Lymphopoiesis , PAX5 Transcription Factor , Trans-Activators , Animals , Ikaros Transcription Factor/metabolism , Ikaros Transcription Factor/genetics , PAX5 Transcription Factor/metabolism , PAX5 Transcription Factor/genetics , Mice , Trans-Activators/metabolism , Trans-Activators/genetics , B-Lymphocytes/metabolism , B-Lymphocytes/immunology , Lymphopoiesis/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Proteolysis , Precursor Cells, B-Lymphoid/metabolism , Precursor Cells, B-Lymphoid/immunology , Mice, Inbred C57BL , Mice, Knockout , Cell Differentiation , Transcription Factor 7-Like 1 Protein/metabolism , Transcription Factor 7-Like 1 Protein/genetics , Transcription Factor 3/metabolism , Transcription Factor 3/genetics , V(D)J Recombination , Interferon Regulatory Factors/metabolism , Interferon Regulatory Factors/genetics , Immunoglobulin Light Chains, Surrogate/metabolism , Immunoglobulin Light Chains, Surrogate/genetics , Transcription, Genetic
7.
Sci Rep ; 14(1): 20146, 2024 08 30.
Article in English | MEDLINE | ID: mdl-39209978

ABSTRACT

Microglia are resident immune cells in the central nervous system, including the retina that surveil the environment for damage and infection. Following retinal damage, microglia undergo morphological changes, migrate to the site of damage, and express and secrete pro-inflammatory signals. In the zebrafish retina, inflammation induces the reprogramming and proliferation of Müller glia and the regeneration of neurons following damage or injury. Immunosuppression or pharmacological ablation of microglia reduce or abolish Müller glia proliferation. We evaluated the retinal architecture and retinal regeneration in adult zebrafish irf8 mutants, which have significantly depleted numbers of microglia. We show that irf8 mutants have normal retinal structure at 3 months post fertilization (mpf) and 6 mpf but fewer cone photoreceptors by 10 mpf. Surprisingly, light-induced photoreceptor ablation induced Müller glia proliferation in irf8 mutants and cone and rod photoreceptor regeneration. Light-damaged retinas from both wild-type and irf8 mutants show upregulated expression of mmp-9, il8, and tnfß pro-inflammatory cytokines. Our data demonstrate that adult zebrafish irf8 mutants can regenerate normally following acute retinal injury. These findings suggest that microglia may not be essential for retinal regeneration in zebrafish and that other mechanisms can compensate for the reduction in microglia numbers.


Subject(s)
Interferon Regulatory Factors , Microglia , Retina , Zebrafish , Animals , Interferon Regulatory Factors/metabolism , Interferon Regulatory Factors/genetics , Microglia/metabolism , Retina/metabolism , Retina/pathology , Mutation , Regeneration , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Retinal Cone Photoreceptor Cells/metabolism , Retinal Cone Photoreceptor Cells/pathology , Photoreceptor Cells, Vertebrate/metabolism , Photoreceptor Cells, Vertebrate/pathology , Cell Proliferation , Light , Ependymoglial Cells/metabolism , Ependymoglial Cells/pathology
8.
Front Immunol ; 15: 1439465, 2024.
Article in English | MEDLINE | ID: mdl-39211041

ABSTRACT

The economic importance of lumpfish (Cyclopterus lumpus) is increasing, but several aspects of its immune responses are not well understood. To discover genes and mechanisms involved in the lumpfish antiviral response, fish were intraperitoneally injected with either the viral mimic polyinosinic:polycytidylic acid [poly(I:C)] or phosphate-buffered saline (PBS; vehicle control), and head kidneys were sampled 24 hours post-injection (hpi) for transcriptomic analyses. RNA sequencing (RNA-Seq) (adjusted p-value <0.05) identified 4,499 upregulated and 3,952 downregulated transcripts in the poly(I:C)-injected fish compared to the PBS-injected fish. Eighteen genes identified as differentially expressed by RNA-Seq were included in a qPCR study that confirmed the upregulation of genes encoding proteins with antiviral immune response functions (e.g., rsad2) and the downregulation of genes (e.g., jarid2b) with potential cellular process functions. In addition, transcript expression levels of 12 members of the interferon regulatory factor (IRF) family [seven of which were identified as poly(I:C)-responsive in this RNA-Seq study] were analyzed using qPCR. Levels of irf1a, irf1b, irf2, irf3, irf4b, irf7, irf8, irf9, and irf10 were significantly higher and levels of irf4a and irf5 were significantly lower in the poly(I:C)-injected fish compared to the PBS-injected fish. This research and associated new genomic resources enhance our understanding of the genes and molecular mechanisms underlying the lumpfish response to viral mimic stimulation and help identify possible therapeutic targets and biomarkers for viral infections in this species.


Subject(s)
Head Kidney , Interferon Regulatory Factors , Poly I-C , Transcriptome , Animals , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , Head Kidney/immunology , Head Kidney/metabolism , Poly I-C/immunology , Perciformes/immunology , Perciformes/genetics , Gene Expression Profiling , Fish Proteins/genetics , Fish Proteins/immunology , Fish Diseases/immunology , Fish Diseases/virology , Fishes/immunology , Fishes/genetics
9.
Microb Pathog ; 195: 106896, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39208957

ABSTRACT

BACKGROUND: Bovine herpesvirus 1 (BoHV-1) is a major pathogen that affects the global bovine population, primarily inducing respiratory and reproductive disorders. Its ability to establish latent infections in neuronal cells and to reactivate under certain conditions poses a continual threat to uninfected hosts. In this study, we aimed to analyze the replication characteristics of BoHV-1 in neuronal cells, as well as the effects of viral replication on host cell immunity and physiology. METHODS: Using the Neuro-2a neuronal-origin cell line as a model, we explored the dynamics of BoHV-1 replication and analyzed differential gene expression profiles post-BoHV-1 infection using high-throughput RNA sequencing. RESULTS: BoHV-1 demonstrated restricted replication in Neuro-2a cells. BoHV-1 induced apoptotic pathways and enhanced the transcription of interferon-stimulated genes and interferon regulatory factors while suppressing the complement cascade in Neuro-2a cells. CONCLUSIONS: Different from BoHV-1 infection in other non-highly differentiated somatic cells result in viral dominance, BoHV-1 regulated the innate immune response in neuronal cells formed a "virus-nerve cell" relative equilibrium state, which may account for the restricted replication of BoHV-1 in neuronal cells, leading to a latent infection. These findings provide a foundation for further research into the mechanism underlying BoHV-1-induced latent infection in nerve cells.


Subject(s)
Gene Expression Profiling , Herpesvirus 1, Bovine , Immunity, Innate , Neurons , Virus Replication , Herpesvirus 1, Bovine/immunology , Herpesvirus 1, Bovine/genetics , Herpesvirus 1, Bovine/physiology , Animals , Cattle , Neurons/virology , Neurons/immunology , Cell Line , Mice , Herpesviridae Infections/virology , Herpesviridae Infections/immunology , Herpesviridae Infections/veterinary , Apoptosis , Transcriptome , Virus Latency , Host-Pathogen Interactions/immunology , Cattle Diseases/virology , Cattle Diseases/immunology , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , High-Throughput Nucleotide Sequencing
10.
Commun Biol ; 7(1): 896, 2024 Jul 24.
Article in English | MEDLINE | ID: mdl-39043941

ABSTRACT

The central nervous system (CNS) includes anatomically distinct macrophage populations including parenchyma microglia and CNS-associated macrophages (CAMs) localized at the interfaces like meninges and perivascular space, which play specialized roles for the maintenance of the CNS homeostasis with the help of precisely controlled gene expressions. However, the transcriptional machinery that determines their cell-type specific states of microglia and CAMs remains poorly understood. Here we show, by myeloid cell-specific deletion of transcription factors, IRF8 and MAFB, that both adult microglia and CAMs utilize IRF8 to maintain their core gene signatures, although the genes altered by IRF8 deletion are different in the two macrophage populations. By contrast, MAFB deficiency robustly affected the gene expression profile of adult microglia, whereas CAMs are almost independent of MAFB. Our data suggest that distinct transcriptional machineries regulate different macrophages in the CNS.


Subject(s)
Central Nervous System , Interferon Regulatory Factors , Macrophages , MafB Transcription Factor , MafB Transcription Factor/genetics , MafB Transcription Factor/metabolism , Animals , Macrophages/metabolism , Interferon Regulatory Factors/metabolism , Interferon Regulatory Factors/genetics , Mice , Central Nervous System/metabolism , Microglia/metabolism , Mice, Knockout , Mice, Inbred C57BL , Transcription, Genetic , Gene Expression Regulation
11.
Adv Exp Med Biol ; 1459: 379-403, 2024.
Article in English | MEDLINE | ID: mdl-39017853

ABSTRACT

Myocyte enhancer factor 2 (MEF2) is a key transcription factor (TF) in skeletal, cardiac, and neural tissue development and includes four isoforms: MEF2A, MEF2B, MEF2C, and MEF2D. These isoforms significantly affect embryonic development, nervous system regulation, muscle cell differentiation, B- and T-cell development, thymocyte selection, and effects on tumorigenesis and leukemia. This chapter describes the multifaceted roles of MEF2 family proteins, covering embryonic development, nervous system regulation, and muscle cell differentiation. It further elucidates the contribution of MEF2 to various blood and immune cell functions. Specifically, in B-cell precursor acute lymphoblastic leukemia (BCP-ALL), MEF2D is aberrantly expressed and forms a fusion protein with BCL9, CSF1R, DAZAP1, HNRNPUL1, and SS18. These fusion proteins are closely related to the pathogenesis of leukemia. In addition, it specifically introduces the regulatory effect of MEF2D fusion protein on the proliferation and growth of B-cell acute lymphoblastic leukemia (B-ALL) cells. Finally, we detail the positive feedback loop between MEF2D and IRF8 that significantly promotes the progression of acute myeloid leukemia (AML) and the importance of the ZMYND8-BRD4 interaction in regulating the IRF8 and MYC transcriptional programs. The MEF2D-CEBPE axis is highlighted as a key transcriptional mechanism controlling the block of leukemic cell self-renewal and differentiation in AML. This chapter starts with the structure and function of MEF2 family proteins, specifically summarizing and analyzing the role of MEF2D in B-ALL and AML, mediating the complex molecular mechanisms of transcriptional regulation and exploring their implications for human health and disease.


Subject(s)
MEF2 Transcription Factors , MEF2 Transcription Factors/metabolism , MEF2 Transcription Factors/genetics , Humans , Animals , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Cell Differentiation/genetics , Gene Expression Regulation, Leukemic , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Cell Proliferation/genetics
12.
Life Sci Alliance ; 7(9)2024 Sep.
Article in English | MEDLINE | ID: mdl-38960622

ABSTRACT

A pleiotropic immunoregulatory cytokine, TGF-ß, signals via the receptor-regulated SMADs: SMAD2 and SMAD3, which are constitutively expressed in normal cells. Here, we show that selective repression of SMAD3 induces cDC differentiation from the CD115+ common DC progenitor (CDP). SMAD3 was expressed in haematopoietic cells including the macrophage DC progenitor. However, SMAD3 was specifically down-regulated in CD115+ CDPs, SiglecH- pre-DCs, and cDCs, whereas SMAD2 remained constitutive. SMAD3-deficient mice showed a significant increase in cDCs, SiglecH- pre-DCs, and CD115+ CDPs compared with the littermate control. SMAD3 repressed the mRNA expression of FLT3 and the cDC-related genes: IRF4 and ID2. We found that one of the SMAD transcriptional corepressors, c-SKI, cooperated with phosphorylated STAT3 at Y705 and S727 to repress the transcription of SMAD3 to induce cDC differentiation. These data indicate that STAT3 and c-Ski induce cDC differentiation by repressing SMAD3: the repressor of the cDC-related genes during the developmental stage between the macrophage DC progenitor and CD115+ CDP.


Subject(s)
Cell Differentiation , DNA-Binding Proteins , Dendritic Cells , Proto-Oncogene Proteins , STAT3 Transcription Factor , Smad3 Protein , Animals , Mice , Dendritic Cells/metabolism , Dendritic Cells/cytology , fms-Like Tyrosine Kinase 3/genetics , fms-Like Tyrosine Kinase 3/metabolism , Inhibitor of Differentiation Protein 2/genetics , Inhibitor of Differentiation Protein 2/metabolism , Interferon Regulatory Factors/metabolism , Interferon Regulatory Factors/genetics , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/genetics , Signal Transduction , Smad2 Protein/metabolism , Smad2 Protein/genetics , Smad3 Protein/metabolism , Smad3 Protein/genetics , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics , DNA-Binding Proteins/metabolism
13.
J Transl Med ; 22(1): 678, 2024 Jul 24.
Article in English | MEDLINE | ID: mdl-39049031

ABSTRACT

BACKGROUND: Patients with lung adenocarcinoma (LUAD) have a low response rate to immune checkpoint blockade. It is highly important to explore the tumor immune escape mechanism of LUAD patients and expand the population of patients who may benefit from immunotherapy. METHODS: Based on 954 bulk RNA-seq data of LUAD patients and 15 single-cell RNA-seq data, the relationships between tumor immune dysfunction and exclusion (TIDE) scores and survival prognosis in each patient were calculated and evaluated, and the immune escape mechanism affecting the independent prognosis of LUAD patients was identified. Functional enrichment analysis explored the antitumour immune response and biological behavior of tumor cells among different LUAD groups. Single-cell annotation and pseudotemporal analysis were used to explore the target molecules and immune escape mechanisms of LUAD. RESULTS: Myeloid-derived suppressor cells (MDSCs) and IRF8 were identified as risk and protective factors for the independent prognosis of LUAD patients, respectively. In the tumor microenvironment of patients with high infiltration of MDSCs, the antitumor immune response is significantly suppressed, while tumor cell division, proliferation, and distant metastasis are significantly enhanced. Single-cell RNA-seq analysis revealed that IRF8 is an important regulator of MDSC differentiation in LUAD myeloid cells. In addition, IRF8 may regulate the differentiation of MDSCs through the IL6-JAK-STAT3 signalling pathway. CONCLUSIONS: IRF8 deficiency impairs the normal development of LUAD myeloid cells and induces their differentiation into MDSCs, thereby accelerating the immune escape of LUAD cells. IRF8-targeted activation to inhibit the formation of MDSCs may be a new target for immunotherapy in LUAD.


Subject(s)
Adenocarcinoma of Lung , Interferon Regulatory Factors , Lung Neoplasms , Myeloid-Derived Suppressor Cells , Tumor Microenvironment , Humans , Myeloid-Derived Suppressor Cells/immunology , Adenocarcinoma of Lung/immunology , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/pathology , Interferon Regulatory Factors/metabolism , Interferon Regulatory Factors/genetics , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Lung Neoplasms/genetics , Tumor Microenvironment/immunology , Prognosis , Female , Gene Expression Regulation, Neoplastic , Signal Transduction , Male , Tumor Escape , Immune Evasion , Single-Cell Analysis , Cell Differentiation
14.
Sci Adv ; 10(28): eadk2091, 2024 Jul 12.
Article in English | MEDLINE | ID: mdl-38996030

ABSTRACT

The mechanism by which interferon regulatory factor 8 (IRF8) mutation contributes to lymphomagenesis is unknown. We modeled IRF8 variants in B cell lymphomas and found that they affected the expression of regulators of antigen presentation. Expression of IRF8 mutants in murine B cell lymphomas suppressed CD4, but not CD8, activation elicited by antigen presentation and downmodulated CD74 and human leukocyte antigen (HLA) DM, intracellular regulators of antigen peptide processing/loading in the major histocompatibility complex (MHC) II. Concordantly, mutant IRF8 bound less efficiently to the promoters of these genes. Mice harboring IRF8 mutant lymphomas displayed higher tumor burden and remodeling of the tumor microenvironment, typified by depletion of CD4, CD8, and natural killer cells, increase in regulatory T cells and T follicular helper cells. Deconvolution of bulk RNA sequencing data from IRF8-mutant human diffuse large B cell lymphoma (DLBCL) recapitulated part of the immune remodeling detected in mice. We concluded that IRF8 mutations contribute to DLBCL biology by facilitating immune escape.


Subject(s)
Antigen Presentation , Antigens, Differentiation, B-Lymphocyte , Histocompatibility Antigens Class II , Interferon Regulatory Factors , Mutation , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , Animals , Antigen Presentation/immunology , Antigen Presentation/genetics , Humans , Mice , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class II/metabolism , Antigens, Differentiation, B-Lymphocyte/genetics , Antigens, Differentiation, B-Lymphocyte/metabolism , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/immunology , Tumor Microenvironment/immunology , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/immunology , Lymphoma, Large B-Cell, Diffuse/pathology , Cell Line, Tumor , Tumor Escape/genetics , Gene Expression Regulation, Neoplastic
15.
Nat Immunol ; 25(8): 1432-1444, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38969872

ABSTRACT

Memory B cells (MBCs) differentiate into plasma cells (PCs) or germinal centers (GCs) upon antigen recall. How this decision is programmed is not understood. We found that the relative strength between two antagonistic transcription factors, B lymphocyte-induced maturation protein 1 (BLIMP1) and BTB domain and CNC homolog 2 (BACH2), progressively increases in favor of BLIMP1 in antigen-responding B cells through the course of primary responses. MBC subsets that preferentially produce secondary GCs expressed comparatively higher BACH2 but lower BLIMP1 than those predisposed for PC development. Skewing the BLIMP1-BACH2 balance in otherwise fate-predisposed MBC subsets could switch their fate preferences. Underlying the changing BLIMP1-over-BACH2 balance, we observed progressively increased accessibilities at chromatin loci that are specifically opened in PCs, particularly those that contain interferon-sensitive response elements (ISREs) and are controlled by interferon regulatory factor 4 (IRF4). IRF4 is upregulated by B cell receptor, CD40 or innate receptor signaling and it induces graded levels of PC-specifying epigenetic imprints according to the strength of stimulation. By analyzing history-stamped GC B cells, we found progressively increased chromatin accessibilities at PC-specific, IRF4-controlled gene loci over time. Therefore, the cumulative stimulation history of B cells is epigenetically recorded in an IRF4-dependent manner, determines the relative strength between BLIMP1 and BACH2 in individual MBCs and dictates their probabilities to develop into GCs or PCs upon restimulation.


Subject(s)
Basic-Leucine Zipper Transcription Factors , Cell Differentiation , Epigenesis, Genetic , Germinal Center , Immunologic Memory , Interferon Regulatory Factors , Memory B Cells , Plasma Cells , Positive Regulatory Domain I-Binding Factor 1 , Positive Regulatory Domain I-Binding Factor 1/metabolism , Positive Regulatory Domain I-Binding Factor 1/genetics , Animals , Interferon Regulatory Factors/metabolism , Interferon Regulatory Factors/genetics , Mice , Basic-Leucine Zipper Transcription Factors/metabolism , Basic-Leucine Zipper Transcription Factors/genetics , Memory B Cells/immunology , Memory B Cells/metabolism , Plasma Cells/immunology , Plasma Cells/metabolism , Germinal Center/immunology , Germinal Center/metabolism , Mice, Inbred C57BL , Signal Transduction , Lymphocyte Activation/genetics
16.
J Immunol ; 213(5): 743-752, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39058321

ABSTRACT

IFN regulatory factors (IRFs) are transcription factors that mediate homeostatic mechanisms of host defense against pathogens. In addition to IRF1-9, which are conserved across vertebrates, teleost fishes have two other IRFs, IRF10 and IRF11. In zebrafish (Danio rerio), IRF10 represses the expression of IFNφ1 and IFNφ3, whereas IRF11 exerts the opposite effect. In this study, we found IRF10 could significantly inhibit the expression of IFNφ1 and IFNφ3 induced by IFN11 to synergistically regulate type I IFN expression. To clarify the synergistically regulatory mechanism of IRF10 and IRF11 in type I IFN expression, we determined and analyzed the crystal structures of the DNA-binding domains (DBDs) of zebrafish IRF10 and IRF11 bound to DNA, as well as IRF11 DBD in apo form. The interactions of IRF10-DBD and IRF11-DBD with DNA backbone were elaborated in detail. Further analysis showed that IRF10 and IRF11 have the same binding patterns and comparable affinities with the IFN-sensitive response elements of IFNφ1 and IFNφ3 promoters. Therefore, IRF10 could function as a controlling factor for IRF11 by competitive binding of the IFN-sensitive response elements to coregulate the host IFN response. Accordingly, similar to IRF1 and IRF2 in mammals, IRF10 and IRF11 act as another pair of negative and positive regulators to balance the antiviral responses in fish.


Subject(s)
DNA , Interferon Regulatory Factors , Zebrafish , Animals , Zebrafish/immunology , DNA/immunology , Interferon Regulatory Factors/metabolism , Interferon Regulatory Factors/genetics , Crystallography, X-Ray , Zebrafish Proteins/metabolism , Zebrafish Proteins/genetics , Protein Binding , Gene Expression Regulation , Interferon Type I/metabolism , Interferon Type I/immunology , Interferons/metabolism , Interferons/immunology
17.
J Immunoassay Immunochem ; 45(5): 432-451, 2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39051937

ABSTRACT

BACKGROUND AND OBJECTIVES: The type I interferon (IFN) signature has been found to be overactivated in many systemic autoimmune diseases. This may be explained by impaired regulation of interferon-stimulated genes (ISGs) as well as interferon-induced protein 44 (IFI44) expression via their regulatory mechanisms via interferon regulatory factors (IRFs). PATIENTS AND METHODS: This case-control study includes two groups: 50 RA patients and 50 healthy controls. The quantification of IFI44 and IRF4 expression levels by the real-time PCR technique was estimated. Disease Activity Score-28 (DAS-28) was estimated for RA patients only. RESULTS: Among the RA patients, there were statistically significant increased ESR, CRP, TLC, RF, and anti-CCP levels (p value < 0.001) and significant increased expression of the IFI44 and IRF4 genes (p value < 0.001). There was a significant positive correlation between the IFI44 and IRF4, and there was a significant correlation between both and ESR and anti-CCP among RA patients. At a cutoff point of 1.95, IFI44 shows higher sensitivity and specificity values than IRF4 for the diagnosis of RA. CONCLUSION: IFI44 was more sensitive for RA diagnosis than IRF4. IFI44 and IRF4 overexpression could be promising predictors of RA diagnosis and might become useful clinical tools to guide therapeutic strategies.


Subject(s)
Arthritis, Rheumatoid , Interferon Regulatory Factors , Humans , Interferon Regulatory Factors/genetics , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/diagnosis , Male , Female , Middle Aged , Case-Control Studies , Adult , Intracellular Signaling Peptides and Proteins/genetics , Antigens , Cytoskeletal Proteins
18.
Epigenetics ; 19(1): 2374988, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39003776

ABSTRACT

Early detection is crucial for increasing the survival rate of gastric cancer (GC). We aimed to identify a methylated cell-free DNA (cfDNA) marker panel for detecting GC. The differentially methylated CpGs (DMCs) were selected from datasets of The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. The selected DMCs were validated and further selected in tissue samples (40 gastric cancer and 36 healthy white blood cell samples) and in a quarter sample volume of plasma samples (37 gastric cancer, 12 benign gastric disease, and 43 healthy individuals). The marker combination selected was then evaluated in a normal sample volume of plasma samples (35 gastric cancer, 39 control diseases, and 40 healthy individuals) using real-time methylation-specific PCR (MSP). The analysis of the results compared methods based on 2-ΔΔCt values and Ct values. In the results, 30 DMCs were selected through bioinformatics methods, and then 5 were selected for biological validation. The marker combination of two fragments of IRF4 (IRF4-1 and IRF4-2) and one of ZEB2 was selected due to its good performance. The Ct-based method was selected for its good results and practical advantages. The assay, IRF4-1 and IRF4-2 in one fluorescence channel and ZEB2 in another, obtained 74.3% sensitivity for the GC group at any stage, at 92.4% specificity. In conclusion, the panel of IRF4 and ZEB2 in plasma cfDNA demonstrates good diagnostic performance and application potential in clinical settings.


Subject(s)
Biomarkers, Tumor , Cell-Free Nucleic Acids , DNA Methylation , Interferon Regulatory Factors , Stomach Neoplasms , Zinc Finger E-box Binding Homeobox 2 , Humans , Stomach Neoplasms/genetics , Stomach Neoplasms/blood , Stomach Neoplasms/diagnosis , Interferon Regulatory Factors/genetics , Biomarkers, Tumor/genetics , Biomarkers, Tumor/blood , Female , Male , Zinc Finger E-box Binding Homeobox 2/genetics , Zinc Finger E-box Binding Homeobox 2/metabolism , Cell-Free Nucleic Acids/genetics , Middle Aged , Aged , Adult
19.
Cancer Cell ; 42(8): 1326-1328, 2024 Aug 12.
Article in English | MEDLINE | ID: mdl-39029465

ABSTRACT

In Cancer Cell, Bolomsky et al., Duplaquet et al., and He et al. identify cancers that are dependent on the BAF chromatin remodeling complex, specifically IRF4-driven multiple myeloma and POU2F3-subtype small cell lung cancer, highlighting potential therapeutic applications for BAF complex inhibitors/degraders.


Subject(s)
Transcription Factors , Humans , Transcription Factors/metabolism , Transcription Factors/genetics , Chromatin Assembly and Disassembly , Interferon Regulatory Factors/metabolism , Interferon Regulatory Factors/genetics , Multiple Myeloma/genetics , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Neoplasms/genetics , Neoplasms/metabolism , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/metabolism , Small Cell Lung Carcinoma/pathology
20.
Front Immunol ; 15: 1415565, 2024.
Article in English | MEDLINE | ID: mdl-38989285

ABSTRACT

How the microbiome regulates responses of systemic innate immune cells is unclear. In the present study, our purpose was to document a novel mechanism by which the microbiome mediates crosstalk with the systemic innate immune system. We have identified a family of microbiome Bacteroidota-derived lipopeptides-the serine-glycine (S/G) lipids, which are TLR2 ligands, access the systemic circulation, and regulate proinflammatory responses of splenic monocytes. To document the role of these lipids in regulating systemic immunity, we used oral gavage with an antibiotic to decrease the production of these lipids and administered exogenously purified lipids to increase the systemic level of these lipids. We found that decreasing systemic S/G lipids by decreasing microbiome Bacteroidota significantly enhanced splenic monocyte proinflammatory responses. Replenishing systemic levels of S/G lipids via exogenous administration returned splenic monocyte responses to control levels. Transcriptomic analysis demonstrated that S/G lipids regulate monocyte proinflammatory responses at the level of gene expression of a small set of upstream inhibitors of TLR and NF-κB pathways that include Trem2 and Irf4. Consistent with enhancement in proinflammatory cytokine responses, decreasing S/G lipids lowered gene expression of specific pathway inhibitors. Replenishing S/G lipids normalized gene expression of these inhibitors. In conclusion, our results suggest that microbiome-derived S/G lipids normally establish a level of buffered signaling activation necessary for well-regulated innate immune responses in systemic monocytes. By regulating gene expression of inflammatory pathway inhibitors such as Trem2, S/G lipids merit broader investigation into the potential dysfunction of other innate immune cells, such as microglia, in diseases such as Alzheimer's disease.


Subject(s)
Monocytes , Signal Transduction , Monocytes/immunology , Monocytes/metabolism , Monocytes/drug effects , Animals , Mice , Microbiota/immunology , Mice, Inbred C57BL , Immunity, Innate , Toll-Like Receptor 2/metabolism , Gene Expression Regulation/drug effects , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/genetics , Lipopeptides/pharmacology , Receptors, Immunologic/metabolism , Receptors, Immunologic/genetics , NF-kappa B/metabolism , Inflammation/immunology , Interferon Regulatory Factors/metabolism , Interferon Regulatory Factors/genetics , Male , Lipids , Spleen/immunology , Spleen/metabolism , Cytokines/metabolism , Female
SELECTION OF CITATIONS
SEARCH DETAIL