Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Immunity ; 57(4): 649-673, 2024 Apr 09.
Article in English | MEDLINE | ID: mdl-38599164

ABSTRACT

Toll-like receptors (TLRs) are an evolutionarily conserved family in the innate immune system and are the first line of host defense against microbial pathogens by recognizing pathogen-associated molecular patterns (PAMPs). TLRs, categorized into cell surface and endosomal subfamilies, recognize diverse PAMPs, and structural elucidation of TLRs and PAMP complexes has revealed their intricate mechanisms. TLRs activate common and specific signaling pathways to shape immune responses. Recent studies have shown the importance of post-transcriptional regulation in TLR-mediated inflammatory responses. Despite their protective functions, aberrant responses of TLRs contribute to inflammatory and autoimmune disorders. Understanding the delicate balance between TLR activation and regulatory mechanisms is crucial for deciphering their dual role in immune defense and disease pathogenesis. This review provides an overview of recent insights into the history of TLR discovery, elucidation of TLR ligands and signaling pathways, and their relevance to various diseases.


Subject(s)
Pathogen-Associated Molecular Pattern Molecules , Toll-Like Receptors , Toll-Like Receptors/metabolism , Immunity, Innate/physiology , Signal Transduction , Gene Expression Regulation
2.
Int Immunol ; 2024 Apr 22.
Article in English | MEDLINE | ID: mdl-38646837

ABSTRACT

Interleukin-6 (IL-6) plays a crucial role in various cellular functions, including the innate and adaptive immune responses. Dysregulated expression of IL-6 is associated with hyperinflammation and chronic inflammatory diseases. In this study, we aimed to identify the enhancer regions responsible for robust Il6 mRNA expression in murine macrophages. Through comprehensive genome-wide ChIP-seq and ATAC-seq analyses, we identified two distinct clusters, termed E1 and E2 regions, located at -144 kb to -163 kb relative to the Il6 transcription start site in lipopolysaccharide (LPS)-activated murine macrophages. These clusters exhibited an accumulation of histone modification marks (H3K27ac and H3K4me1), as well as open chromatin, and were found to contain binding sites for the transcription factors PU.1, NF-κB, C/EBPß, and JunB. Upregulation of non-coding RNA (ncRNA) transcripts from the E1 and E2 regions was observed upon LPS stimulation, and repression of these ncRNAs resulted in abrogation of Il6 expression. Additionally, deletion of either E1 or E2 regions significantly impaired Il6 expression, while CRISPR/dCas9 activation-mediated recruitment of the co-activator p300 to the E1 and E2 regions facilitated Il6 expression. Collectively, our findings suggest that the E1 and E2 regions serve as putative enhancers for Il6 expression.

3.
Blood ; 143(3): 243-257, 2024 Jan 18.
Article in English | MEDLINE | ID: mdl-37922454

ABSTRACT

ABSTRACT: Regulation of lineage biases in hematopoietic stem and progenitor cells (HSPCs) is pivotal for balanced hematopoietic output. However, little is known about the mechanism behind lineage choice in HSPCs. Here, we show that messenger RNA (mRNA) decay factors regnase-1 (Reg1; Zc3h12a) and regnase-3 (Reg3; Zc3h12c) are essential for determining lymphoid fate and restricting myeloid differentiation in HSPCs. Loss of Reg1 and Reg3 resulted in severe impairment of lymphopoiesis and a mild increase in myelopoiesis in the bone marrow. Single-cell RNA sequencing analysis revealed that Reg1 and Reg3 regulate lineage directions in HSPCs via the control of a set of myeloid-related genes. Reg1- and Reg3-mediated control of mRNA encoding Nfkbiz, a transcriptional and epigenetic regulator, was essential for balancing lymphoid/myeloid lineage output in HSPCs in vivo. Furthermore, single-cell assay for transposase-accessible chromatin sequencing analysis revealed that Reg1 and Reg3 control the epigenetic landscape on myeloid-related gene loci in early stage HSPCs via Nfkbiz. Consistently, an antisense oligonucleotide designed to inhibit Reg1- and Reg3-mediated Nfkbiz mRNA degradation primed hematopoietic stem cells toward myeloid lineages by enhancing Nfkbiz expression. Collectively, the collaboration between posttranscriptional control and chromatin remodeling by the Reg1/Reg3-Nfkbiz axis governs HSPC lineage biases, ultimately dictating the fate of lymphoid vs myeloid differentiation.


Subject(s)
Bone Marrow , Hematopoietic Stem Cells , Cell Lineage/genetics , Hematopoietic Stem Cells/metabolism , Bone Marrow/metabolism , Hematopoiesis/genetics , RNA, Messenger/metabolism , Cell Differentiation/genetics
4.
Int Rev Immunol ; 42(4): 274-286, 2023.
Article in English | MEDLINE | ID: mdl-35499950

ABSTRACT

Inflammation plays indispensable roles in building the immune responses such as acquired immunity against harmful pathogens. Furthermore, it is essential for maintaining biological homeostasis in ever-changing conditions. Pattern-recognition receptors (PRRs) reside in cell membranes, endosomes or cytoplasm, and function as triggers for inflammatory responses. Binding of pathogen- or self-derived components, such as DNA, to PRRs activates downstream signaling cascades, resulting in the production of a series of pro-inflammatory cytokines and type I interferons (IFNs). While these series of responses are essential for host defense, the unexpected release of DNA from the nucleus or mitochondria of host cells can lead to autoimmune and autoinflammatory diseases. In this review, we focus on DNA-sensing mechanisms via PRRs and the disorders and extraordinary conditions caused by self-derived DNA.


Subject(s)
Immunity, Innate , Receptors, Pattern Recognition , Humans , Receptors, Pattern Recognition/metabolism , Cytokines/metabolism , Adaptive Immunity , DNA/genetics
5.
Cell Rep ; 41(11): 111828, 2022 12 13.
Article in English | MEDLINE | ID: mdl-36516765

ABSTRACT

Lung CD8+ memory T cells play central roles in protective immunity to respiratory viruses, such as influenza A virus (IAV). Here, we find that alveolar macrophages (AMs) function as antigen-presenting cells that support the expansion of lung CD8+ memory T cells. Intranasal antigen administration to mice subcutaneously immunized with antigen results in a rapid expansion of antigen-specific CD8+ T cells in the lung, which is dependent on antigen cross-presentation by AMs. AMs highly express interleukin-18 (IL-18), which mediates subsequent formation of CD103+CD8+ resident memory T (TRM) cells in the lung. In a mouse model of IAV infection, AMs are required for expansion of virus-specific CD8+ T cells and CD103+CD8+ TRM cells and inhibiting virus replication in the lungs during secondary infection. These results suggest that AMs instruct a rapid expansion of antigen-specific CD8+ T cells in lung, which protect the host from respiratory virus infection.


Subject(s)
Influenza A virus , Orthomyxoviridae Infections , Mice , Animals , Macrophages, Alveolar , CD8-Positive T-Lymphocytes , Immunologic Memory , Cross-Priming , Lung
6.
Front Cell Infect Microbiol ; 12: 910654, 2022.
Article in English | MEDLINE | ID: mdl-35734577

ABSTRACT

Pattern recognition receptors (PRRs) play critical roles in recognizing pathogen-derived nucleic acids and inducing innate immune responses, such as inflammation and type I interferon production. PRRs that recognize nucleic acids include members of endosomal Toll-like receptors, cytosolic retinoic acid inducible gene I-like receptors, cyclic GMP-AMP synthase, absent in melanoma 2-like receptors, and nucleotide binding oligomerization domain-like receptors. Aberrant recognition of self-derived nucleic acids by these PRRs or unexpected activation of downstream signaling pathways results in the constitutive production of type I interferons and inflammatory cytokines, which lead to the development of autoimmune or autoinflammatory diseases. In this review, we focus on the nucleic acid-sensing machinery and its pathophysiological roles in various inflammatory diseases.


Subject(s)
Interferon Type I , Nucleic Acids , Immunity, Innate , Interferon Type I/metabolism , Nucleic Acids/metabolism , Receptors, Pattern Recognition , Toll-Like Receptors
7.
Genes Cells ; 27(7): 482-492, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35467779

ABSTRACT

Lipopolysaccharide on gram negative bacteria can be detected by Toll-like receptor 4 (TLR4) to elicit a series of innate immune responses, leading to inflammation to eliminate the targeted pathogen. However, dysregulation in the responses results in excessive inflammation. The 1'-acetoxychavicol acetate (ACA) is a bioactive compound originated from Alpinia species known to have anti-inflammatory and apoptosis-inducing properties. Here, we found that ACA inhibits lipopolysaccharide-induced expression and production of proinflammatory cytokines such as interleukin 6 and TNFα by macrophages. ACA suppresses the activation of NF-κB and MAP kinases in TLR4 signaling. Moreover, ACA also inhibits TLR4-mediated induction of type I interferon by suppressing IRF3 activation. In lipopolysaccharide-challenged mice, ACA treatment successfully increased the survival of mice and alleviated inflammation in the lung. Thus, ACA is a potential anti-inflammatory agent to regulate excessive inflammation.


Subject(s)
Benzyl Alcohols , Inflammation , Toll-Like Receptor 4 , Animals , Benzyl Alcohols/pharmacology , Cytokines/metabolism , Inflammation/drug therapy , Lipopolysaccharides/pharmacology , Mice , NF-kappa B/metabolism , Toll-Like Receptor 4/metabolism
8.
iScience ; 25(4): 104118, 2022 Apr 15.
Article in English | MEDLINE | ID: mdl-35402874

ABSTRACT

The innate immune system is an immediate defense against infectious pathogens by the production of inflammatory cytokines and other mediators. Deficiencies of epigenetic regulatory enzymes, such as Tet1 and Dnmt1, cause dysregulation of cytokine expression. However, it is unclear if DNA methylation at a single CpG dinucleotide in a specific gene locus can regulate gene expression. In this study, we demonstrated that CpG+286 and CpG+348 in exon 2 of the Il6 gene are similar in various primary mouse cells. In lipopolysaccharide-stimulated condition, hypomethylated CpG+286 promoted Il6 expression whereas deletion of CpG+348 led to a reduction in Il6 expression associated with enhanced CTCF binding to the Il6 locus. Moreover, hypomethylation at CpG+286 in alveolar macrophages from aged mice led to higher Il6 expression in response to LPS compared with young mice. Thus, DNA methylation at specific CpG dinucleotides plays an important regulatory role in Il6 expression.

9.
Int Immunol ; 33(7): 373-386, 2021 06 18.
Article in English | MEDLINE | ID: mdl-33830232

ABSTRACT

The nucleotide-binding oligomerization domain-like receptor (NLR) family pyrin domain containing (NLRP) 3 inflammasome is a multiprotein complex that triggers Caspase-1-mediated IL-1ß production and pyroptosis, and its dysregulation is associated with the pathogenesis of inflammatory diseases. 1'-Acetoxychavicol acetate (ACA) is a natural compound in the rhizome of tropical ginger Alpinia species with anti-microbial, anti-allergic and anti-cancer properties. In this study, we found that ACA suppressed NLRP3 inflammasome activation in mouse bone marrow-derived macrophages and human THP-1 monocytes. ACA inhibited Caspase-1 activation and IL-1ß production by NLRP3 agonists such as nigericin, monosodium urate (MSU) crystals, and ATP. Moreover, it suppressed oligomerization of the adapter molecule, apoptosis-associated speck-like protein containing a CARD (ASC), and Caspase-1-mediated cleavage of pyroptosis executor Gasdermin D. Mechanistically, ACA inhibited generation of mitochondrial reactive oxygen species (ROS) and prevented release of oxidized mitochondrial DNA, which trigger NLRP3 inflammasome activation. ACA also prevented NLRP3 inflammasome activation in vivo, as evidenced in the MSU crystal-induced peritonitis and dextran sodium sulfate-induced colitis mouse models accompanied by decreased Caspase-1 activation. Thus, ACA is a potent inhibitor of the NLRP3 inflammasome for prevention of NLRP3-associated inflammatory diseases.


Subject(s)
Benzyl Alcohols/pharmacology , Inflammasomes/drug effects , Inflammasomes/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Reactive Oxygen Species/metabolism , Animals , Caspase 1/metabolism , Cells, Cultured , Disease Models, Animal , Humans , Inflammation/metabolism , Interleukin-1beta/metabolism , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Monocytes/drug effects , Monocytes/metabolism , Peritonitis/drug therapy , Peritonitis/metabolism , Phagocytosis/drug effects , Pyroptosis/drug effects , THP-1 Cells/drug effects , THP-1 Cells/metabolism
10.
Biochem Biophys Res Commun ; 530(4): 699-705, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32768188

ABSTRACT

Interleukin-33 (IL-33) is a member of the IL-1 cytokine family and plays critical roles in facilitating type-2 immune responses. IL-33 is localized in the nucleus and released to the extracellular milieu during cell death, although the precise mechanisms underlying IL-33 mobilization remain unclear. Here, we found that nigericin, a toxin derived from Streptomyces hygroscopicus, promoted IL-33 translocation from the nucleus to the cytosol before extracellular release. This translocation was inhibited by chelating Ca2+ with EGTA or membrane protection by glycine treatment. Ca2+ ionophore A23187 stimulation caused IL-33 translocation to the cytoplasm but was not sufficient for extracellular release. However, IL-33 release was induced by detergent treatment, which indicates that membrane rupture is required for IL-33 release. The pore-forming pyroptosis executor gasdermin D was cleaved following nigericin stimulation, and overexpression of the cleaved gasdermin D-N-terminal fragment that forms the membrane pore sufficiently induced IL-33 release, which was blocked by EGTA and glycine. Together, these findings suggest that Ca2+-dependent signals and gasdermin D pore formation are required for robust IL-33 production.


Subject(s)
Calcium/immunology , Interleukin-33/immunology , Nigericin/immunology , Streptomyces/immunology , Animals , Cells, Cultured , HEK293 Cells , Humans , Interleukin-33/analysis , Intracellular Signaling Peptides and Proteins/immunology , Mice, Inbred C57BL , Phosphate-Binding Proteins/immunology
11.
Front Immunol ; 11: 625833, 2020.
Article in English | MEDLINE | ID: mdl-33633744

ABSTRACT

Recognition of pathogen-derived nucleic acids by pattern-recognition receptors (PRRs) is essential for eliciting antiviral immune responses by inducing the production of type I interferons (IFNs) and proinflammatory cytokines. Such responses are a prerequisite for mounting innate and pathogen-specific adaptive immune responses. However, host cells also use nucleic acids as carriers of genetic information, and the aberrant recognition of self-nucleic acids by PRRs is associated with the onset of autoimmune or autoinflammatory diseases. In this review, we describe the mechanisms of nucleic acid sensing by PRRs, including Toll-like receptors, RIG-I-like receptors, and DNA sensor molecules, and their signaling pathways as well as the disorders caused by uncontrolled or unnecessary activation of these PRRs.


Subject(s)
DNA/immunology , Immunity, Innate , Interferon Type I/immunology , Receptors, Pattern Recognition/immunology , Signal Transduction/immunology , Animals , Humans , Inflammation/immunology , Inflammation/pathology
12.
Biochem Biophys Res Commun ; 521(4): 1077-1082, 2020 01 22.
Article in English | MEDLINE | ID: mdl-31733835

ABSTRACT

RIG-I-like receptors (RLRs) are cytoplasmic sensors for viral RNA that elicit antiviral innate immune responses. RLR signaling culminates in the activation of the protein kinase TBK1, which mediates phosphorylation and nuclear translocation of IRF3 that regulates expression of type I interferon genes. Here, we found that Nucleoporin 93 (Nup93), components of nuclear pore complex (NPC), plays an important role in RLR-mediated antiviral responses. Nup93-deficient RAW264.7 macrophage cells exhibited decreased expression of Ifnb1 and Cxcl10 genes after treatment with a synthetic RLR agonist stimulation as well as Newcastle Disease Virus infection. Silencing Nup93 in murine primary macrophages and embryonic fibroblasts also resulted in reduced expression of these genes. IRF3 nuclear translocation during RLR signaling was impaired in Nup93-deficient RAW264.7 cells. Notably, the activation of TBK1 during RLR signaling was also decreased in Nup93-deficient cells. We found that Nup93 formed a complex with TBK1, and Nup93 overexpression enhanced TBK1-mediated IFNß promoter activation. Taken together, our findings suggest that Nup93 regulates antiviral innate immunity by enhancing TBK1 activity and IRF3 nuclear translocation.


Subject(s)
Antiviral Agents/metabolism , Immunity, Innate , Newcastle disease virus/physiology , Nuclear Pore Complex Proteins/metabolism , Animals , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Gene Knockdown Techniques , HEK293 Cells , Humans , Immunity, Innate/drug effects , Interferon Regulatory Factor-3/metabolism , Mice , Newcastle disease virus/drug effects , Nuclear Pore Complex Proteins/deficiency , Poly I-C/pharmacology , Protein Binding/drug effects , Protein Serine-Threonine Kinases/metabolism , Protein Transport/drug effects , RAW 264.7 Cells
13.
J Biol Chem ; 294(21): 8412-8423, 2019 05 24.
Article in English | MEDLINE | ID: mdl-30944173

ABSTRACT

The innate immune system plays an essential role in initial recognition of pathogen infection by producing inflammatory cytokines and type I interferons. cGAS is a cytoplasmic sensor for DNA derived from DNA viruses. cGAS binding with DNA induces the production of cGAMP, a second messenger that associates with STING in endoplasmic reticulum (ER). STING changes its cellular distribution from ER to perinuclear Golgi, where it activates the protein kinase TBK1 that catalyzes the phosphorylation of IRF3. Here we found that STING trafficking is regulated by myotubularin-related protein (MTMR) 3 and MTMR4, members of protein tyrosine phosphatases that dephosphorylate 3' position in phosphatidylinositol (PtdIns) and generate PtdIns5P from PtdIns3,5P2 and PtdIns from PtdIns3P. We established MTMR3 and MTMR4 double knockout (DKO) RAW264.7 macrophage cells and found that they exhibited increased type I interferon production after interferon-stimulatory DNA (ISD) stimulation and herpes simplex virus 1 infection concomitant with enhanced IRF3 phosphorylation. In DKO cells, STING rapidly trafficked from ER to Golgi after ISD stimulation. Notably, DKO cells exhibited enlarged cytosolic puncta positive for PtdIns3P and STING was aberrantly accumulated in this puncta. Taken together, these results suggest that MTMR3 and MTMR4 regulate the production of PtdIns3P, which plays a critical role in suppressing DNA-mediated innate immune responses via modulating STING trafficking.


Subject(s)
DNA, Viral/immunology , Herpesvirus 1, Human/immunology , Immunity, Innate , Macrophages/immunology , Membrane Proteins/immunology , Phosphatidylinositol Phosphates/immunology , Protein Tyrosine Phosphatases, Non-Receptor/immunology , Animals , DNA, Viral/genetics , Herpesvirus 1, Human/genetics , Membrane Proteins/genetics , Mice , Phosphatidylinositol Phosphates/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/immunology , Protein Transport/genetics , Protein Transport/immunology , Protein Tyrosine Phosphatases, Non-Receptor/genetics , RAW 264.7 Cells
14.
J Immunol ; 200(11): 3814-3824, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29678949

ABSTRACT

Retinoic acid-inducible gene I (RIG-I)-like receptors (RLRs), RIG-I, and melanoma differentiation-associated gene 5 (MDA5) play a critical role in inducing antiviral innate immune responses by activating IFN regulatory factor 3 (IRF3) and NF-κB, which regulates the transcription of type I IFN and inflammatory cytokines. Antiviral innate immune responses are also regulated by posttranscriptional and translational mechanisms. In this study, we identified an RNA-binding protein HuR as a regulator for RLR signaling. Overexpression of HuR, but not of other Hu members, increased IFN-ß promoter activity. HuR-deficient macrophage cells exhibited decreased Ifnb1 expression after RLR stimulation, whereas they showed normal induction after stimulation with bacterial LPS or immunostimulatory DNA. Moreover, HuR-deficient cells displayed impaired nuclear translocation of IRF3 after RLR stimulation. In HuR-deficient cells, the mRNA expression of Polo-like kinase (PLK) 2 was markedly reduced. We found that HuR bound to the 3' untranslated region of Plk2 mRNA and increased its stabilization. PLK2-deficient cells also showed reduced IRF3 nuclear translocation and Ifnb mRNA expression during RLR signaling. Together, these findings suggest that HuR bolsters RLR-mediated IRF3 nuclear translocation by controlling the stability of Plk2 mRNA.


Subject(s)
Antiviral Agents/immunology , ELAV-Like Protein 1/immunology , Immunity, Innate/immunology , Protein Serine-Threonine Kinases/immunology , RNA, Messenger/immunology , 3' Untranslated Regions/immunology , Animals , Cell Line , DEAD Box Protein 58/immunology , DNA/immunology , HEK293 Cells , Humans , Interferon Regulatory Factor-3/immunology , Interferon Type I/immunology , Interferon-Induced Helicase, IFIH1/immunology , Interferon-beta/immunology , Mice , Mice, Inbred C57BL , NF-kappa B/immunology , RAW 264.7 Cells , Signal Transduction/immunology
15.
Crit Rev Immunol ; 38(4): 279-301, 2018.
Article in English | MEDLINE | ID: mdl-30806244

ABSTRACT

The innate immune system serves as the first line of defense to protect the host from pathogen infection. As a first step, the pattern recognition receptors (PRRs) recognize pathogen-associated molecular patterns (PAMPs), such as non-self DNA derived from pathogens, and damage-associated molecular patterns (DAMPs), such as self DNA released from damaged or injured cells. Sensing of such DNAs elicits innate immune responses through the production of type I interferons (IFNs) and proinflammatory cytokines resulting from the activation of interferon regulatory factor 3 (IRF3) and nuclear factor kappa B (NF-κB), respectively. These cytokines are key players in interlinking innate and adaptive immune responses. However, defects in DNA sensors and their signaling cascades lead to dysregulation of immune responses, autoimmune diseases, and cancer progression. Here we provide an update on DNA signaling pathways in response to pathogen infection and cell injury, and on the roles of regulators in governing the immune system and maintaining host homeostasis. We also discuss the evasion of immunosurveillance by pathogens.


Subject(s)
DNA/immunology , Immunity, Innate/immunology , Receptors, Pattern Recognition/immunology , Signal Transduction/immunology , Animals , Cytokines/biosynthesis , Cytokines/immunology , Humans , Interferon Type I/biosynthesis , Interferon Type I/immunology
16.
Cell Rep ; 19(8): 1614-1630, 2017 05 23.
Article in English | MEDLINE | ID: mdl-28538180

ABSTRACT

Iron metabolism is regulated by transcriptional and post-transcriptional mechanisms. The mRNA of the iron-controlling gene, transferrin receptor 1 (TfR1), has long been believed to be negatively regulated by a yet-unidentified endonuclease. Here, we show that the endonuclease Regnase-1 is critical for the degradation of mRNAs involved in iron metabolism in vivo. First, we demonstrate that Regnase-1 promotes TfR1 mRNA decay. Next, we show that Regnase-1-/- mice suffer from severe iron deficiency anemia, although hepcidin expression is downregulated. The iron deficiency anemia is induced by a defect in duodenal iron uptake. We reveal that duodenal Regnase-1 controls the expression of PHD3, which impairs duodenal iron uptake via HIF2α suppression. Finally, we show that Regnase-1 is a HIF2α-inducible gene and thus provides a positive feedback loop for HIF2α activation via PHD3. Collectively, these results demonstrate that Regnase-1-mediated regulation of iron-related transcripts is essential for the maintenance of iron homeostasis.


Subject(s)
Antigens, CD/metabolism , Homeostasis , Iron/metabolism , Procollagen-Proline Dioxygenase/metabolism , RNA Stability , Receptors, Transferrin/metabolism , Ribonucleases/metabolism , Anemia/metabolism , Anemia/pathology , Animals , Antigens, CD/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Duodenum/metabolism , Ferritins/metabolism , Mice , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Transferrin/genetics , Response Elements/genetics , Ribonucleases/deficiency , Transcription, Genetic
17.
Int Rev Immunol ; 36(2): 74-88, 2017 03 04.
Article in English | MEDLINE | ID: mdl-28333574

ABSTRACT

During viral and bacterial infections, pathogen-derived cytosolic nucleic acids are recognized by the intracellular RNA sensors retinoic acid-inducible gene I and melanoma-differentiated gene 5 and intracellular DNA sensors, including cyclic-di-GMP-AMP synthase, absent in melanoma 2, interferon (IFN)-gamma inducible protein 16, polymerase III, and so on. Binding of intracellular nucleic acids to these sensors activates downstream signaling cascades, resulting in the production of type I IFNs and pro-inflammatory cytokines to induce appropriate systematic immune responses. While these sensors also recognize endogenous nucleic acids and activate immune responses, they can discriminate between self- and non-self-nucleic acids. However, dysfunction of these sensors or failure of regulatory mechanisms causes aberrant activation of immune response and autoimmune disorders. In this review, we focus on how intracellular immune sensors recognize exogenous nucleic acids and activate the innate immune system, and furthermore, how autoimmune diseases result from dysfunction of these sensors.


Subject(s)
Adenylyl Cyclases/metabolism , Autoimmune Diseases/immunology , DEAD Box Protein 58/metabolism , DNA-Binding Proteins/metabolism , Interferon-Induced Helicase, IFIH1/metabolism , Nucleic Acids/immunology , Animals , Cytosol/metabolism , Host-Parasite Interactions , Humans , Immunity, Innate , Receptors, Immunologic , Receptors, Pattern Recognition/metabolism , Self Tolerance , Signal Transduction
18.
J Immunol ; 195(2): 519-27, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-26041538

ABSTRACT

Akirin2, an evolutionarily conserved nuclear protein, is an important factor regulating inflammatory gene transcription in mammalian innate immune cells by bridging the NF-κB and SWI/SNF complexes. Although Akirin is critical for Drosophila immune responses, which totally rely on innate immunity, the mammalian NF-κB system is critical not only for the innate but also for the acquired immune system. Therefore, we investigated the role of mouse Akirin2 in acquired immune cells by ablating Akirin2 function in B lymphocytes. B cell-specific Akirin2-deficient (Cd19(Cre/+)Akirin2(fl/fl)) mice showed profound decrease in the splenic follicular (FO) and peritoneal B-1, but not splenic marginal zone (MZ), B cell numbers. However, both Akirin2-deficient FO and MZ B cells showed severe proliferation defect and are prone to undergo apoptosis in response to TLR ligands, CD40, and BCR stimulation. Furthermore, B cell cycling was defective in the absence of Akirin2 owing to impaired expression of genes encoding cyclin D and c-Myc. Additionally, Brg1 recruitment to the Myc and Ccnd2 promoter was severely impaired in Akirin2-deficient B cells. Cd19(Cre/+)Akirin2(fl/fl) mice showed impaired in vivo immune responses to T-dependent and -independent Ags. Collectively, these results demonstrate that Akirin2 is critical for the mitogen-induced B cell cycle progression and humoral immune responses by controlling the SWI/SNF complex, further emphasizing the significant function of Akirin2 not only in the innate, but also in adaptive immune cells.


Subject(s)
B-Lymphocytes/immunology , Cell Lineage/immunology , Immunity, Humoral , Lymphocyte Activation , Repressor Proteins/immunology , Animals , Antigens, CD19/genetics , Antigens, CD19/immunology , Apoptosis , B-Lymphocytes/cytology , CD40 Antigens/genetics , CD40 Antigens/immunology , Cell Proliferation , Cyclin D/genetics , Cyclin D/immunology , Cyclin D2/genetics , Cyclin D2/immunology , DNA Helicases/genetics , DNA Helicases/immunology , Gene Expression Regulation , Mice , Mice, Knockout , NF-kappa B/genetics , NF-kappa B/immunology , Nuclear Proteins/genetics , Nuclear Proteins/immunology , Promoter Regions, Genetic , Protein Transport , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/immunology , Repressor Proteins/deficiency , Repressor Proteins/genetics , Signal Transduction , Toll-Like Receptors/genetics , Toll-Like Receptors/immunology , Transcription Factors/genetics , Transcription Factors/immunology
19.
Cell ; 161(5): 1058-1073, 2015 May 21.
Article in English | MEDLINE | ID: mdl-26000482

ABSTRACT

Regnase-1 and Roquin are RNA binding proteins essential for degradation of inflammation-related mRNAs and maintenance of immune homeostasis. However, their mechanistic relationship has yet to be clarified. Here, we show that, although Regnase-1 and Roquin regulate an overlapping set of mRNAs via a common stem-loop structure, they function in distinct subcellular locations: ribosome/endoplasmic reticulum and processing-body/stress granules, respectively. Moreover, Regnase-1 specifically cleaves and degrades translationally active mRNAs and requires the helicase activity of UPF1, similar to the decay mechanisms of nonsense mRNAs. In contrast, Roquin controls translationally inactive mRNAs, independent of UPF1. Defects in both Regnase-1 and Roquin lead to large increases in their target mRNAs, although Regnase-1 tends to control the early phase of inflammation when mRNAs are more actively translated. Our findings reveal that differential regulation of mRNAs by Regnase-1 and Roquin depends on their translation status and enables elaborate control of inflammation.


Subject(s)
Inflammation/metabolism , RNA Stability , RNA, Messenger/metabolism , Ribonucleases/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Base Sequence , Codon, Terminator , HeLa Cells , Humans , Inflammation/genetics , Inflammation/immunology , Mice , Molecular Sequence Data , NIH 3T3 Cells , Nucleic Acid Conformation , Polyribosomes/metabolism , Protein Biosynthesis , RNA, Messenger/chemistry , Ribosomal Proteins/metabolism , Trans-Activators/metabolism
20.
J Biol Chem ; 290(2): 1269-80, 2015 Jan 09.
Article in English | MEDLINE | ID: mdl-25451939

ABSTRACT

RIG-I-like receptors (RLRs), including retinoic acid-inducible gene-I (RIG-I) and MDA5, constitute a family of cytoplasmic RNA helicases that senses viral RNA and mounts antiviral innate immunity by producing type I interferons and inflammatory cytokines. Despite their essential roles in antiviral host defense, RLR signaling is negatively regulated to protect the host from excessive inflammation and autoimmunity. Here, we identified ADP-ribosylation factor-like protein 5B (Arl5B), an Arl family small GTPase, as a regulator of RLR signaling through MDA5 but not RIG-I. Overexpression of Arl5B repressed interferon ß promoter activation by MDA5 but not RIG-I, and its knockdown enhanced MDA5-mediated responses. Furthermore, Arl5B-deficient mouse embryonic fibroblast cells exhibited increased type I interferon expression in response to MDA5 agonists such as poly(I:C) and encephalomyocarditis virus. Arl5B-mediated negative regulation of MDA5 signaling does not require its GTP binding ability but requires Arl5B binding to the C-terminal domain of MDA5, which prevents interaction between MDA5 and poly(I:C). Our results, therefore, suggest that Arl5B is a negative regulator for MDA5.


Subject(s)
ADP-Ribosylation Factors/metabolism , DEAD-box RNA Helicases/metabolism , Immunity, Innate/genetics , Interferon Type I/biosynthesis , ADP-Ribosylation Factors/genetics , ADP-Ribosylation Factors/immunology , Animals , Autoimmunity/genetics , DEAD Box Protein 58 , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/immunology , Humans , Interferon-Induced Helicase, IFIH1 , Interferon-beta/genetics , Mice , Promoter Regions, Genetic/genetics , RNA Helicases/immunology , RNA, Viral/immunology , Receptors, Immunologic , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...