Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 474
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Annu Rev Immunol ; 39: 279-311, 2021 04 26.
Article in English | MEDLINE | ID: mdl-33544645

ABSTRACT

The innate immune response is a rapid response to pathogens or danger signals. It is precisely activated not only to efficiently eliminate pathogens but also to avoid excessive inflammation and tissue damage. cis-Regulatory element-associated chromatin architecture shaped by epigenetic factors, which we define as the epiregulome, endows innate immune cells with specialized phenotypes and unique functions by establishing cell-specific gene expression patterns, and it also contributes to resolution of the inflammatory response. In this review, we focus on two aspects: (a) how niche signals during lineage commitment or following infection and pathogenic stress program epiregulomes by regulating gene expression levels, enzymatic activities, or gene-specific targeting of chromatin modifiers and (b) how the programed epiregulomes in turn mediate regulation of gene-specific expression, which contributes to controlling the development of innate cells, or the response to infection and inflammation, in a timely manner. We also discuss the effects of innate immunometabolic rewiring on epiregulomes and speculate on several future challenges to be encountered during the exploration of the master regulators of epiregulomes in innate immunity and inflammation.


Subject(s)
Immunity, Innate , Inflammation , Animals , Epigenesis, Genetic , Humans , Immunity, Innate/genetics , Inflammation/genetics
2.
Immunity ; 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38908373

ABSTRACT

Prolonged activation of the type I interferon (IFN-I) pathway leads to autoimmune diseases such as systemic lupus erythematosus (SLE). Metabolic regulation of cytokine signaling is critical for cellular homeostasis. Through metabolomics analyses of IFN-ß-activated macrophages and an IFN-stimulated-response-element reporter screening, we identified spermine as a metabolite brake for Janus kinase (JAK) signaling. Spermine directly bound to the FERM and SH2 domains of JAK1 to impair JAK1-cytokine receptor interaction, thus broadly suppressing JAK1 phosphorylation triggered by cytokines IFN-I, IFN-II, interleukin (IL)-2, and IL-6. Peripheral blood mononuclear cells (PBMCs) from individuals with SLE showing decreased spermine concentrations exhibited enhanced IFN-I and lupus gene signatures. Spermine treatment attenuated autoimmune pathogenesis in SLE and psoriasis mice and reduced IFN-I signaling in monocytes from individuals with SLE. We synthesized a spermine derivative (spermine derivative 1 [SD1]) and showed that it had a potent immunosuppressive function. Our findings reveal spermine as a metabolic checkpoint for cellular homeostasis and a potential immunosuppressive molecule for controlling autoimmune disease.

3.
Cell ; 175(5): 1336-1351.e17, 2018 11 15.
Article in English | MEDLINE | ID: mdl-30318148

ABSTRACT

As a critical step during innate response, the cytoplasmic ß subunit (IFN-γR2) of interferon-γ receptor (IFN-γR) is induced and translocates to plasma membrane to join α subunit to form functional IFN-γR to mediate IFN-γ signaling. However, the mechanism driving membrane translocation and its significance remain largely unknown. We found, unexpectedly, that mice deficient in E-selectin, an endothelial cell-specific adhesion molecule, displayed impaired innate activation of macrophages upon Listeria monocytogenes infection yet had increased circulating IFN-γ. Inflammatory macrophages from E-selectin-deficient mice had less surface IFN-γR2 and impaired IFN-γ signaling. BTK elicited by extrinsic E-selectin engagement phosphorylates cytoplasmic IFN-γR2, facilitating EFhd2 binding and promoting IFN-γR2 trafficking from Golgi to cell membrane. Our findings demonstrate that membrane translocation of cytoplasmic IFN-γR2 is required to activate macrophage innate response against intracellular bacterial infection, identifying the assembly of functional cytokine receptors on cell membrane as an important layer in innate activation and cytokine signaling.


Subject(s)
E-Selectin/metabolism , Immunity, Innate , Receptors, Interferon/metabolism , Animals , Calcium-Binding Proteins/antagonists & inhibitors , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cell Membrane/metabolism , E-Selectin/deficiency , E-Selectin/genetics , Golgi Apparatus/metabolism , Interferon-gamma/blood , Interferon-gamma/metabolism , Listeria/pathogenicity , Macrophage Activation , Macrophages/cytology , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Protein Transport , RAW 264.7 Cells , RNA Interference , RNA, Small Interfering/metabolism , Receptors, Interferon/deficiency , Receptors, Interferon/genetics , Signal Transduction , Interferon gamma Receptor
4.
Cell ; 173(4): 906-919.e13, 2018 05 03.
Article in English | MEDLINE | ID: mdl-29706547

ABSTRACT

The innate RNA sensor RIG-I is critical in the initiation of antiviral type I interferons (IFNs) production upon recognition of "non-self" viral RNAs. Here, we identify a host-derived, IFN-inducible long noncoding RNA, lnc-Lsm3b, that can compete with viral RNAs in the binding of RIG-I monomers and feedback inactivate the RIG-I innate function at late stage of innate response. Mechanistically, binding of lnc-Lsm3b restricts RIG-I protein's conformational shift and prevents downstream signaling, thereby terminating type I IFNs production. Multivalent structural motifs and long-stem structure are critical features of lnc-Lsm3b for RIG-I binding and inhibition. These data reveal a non-canonical self-recognition mode in the regulation of immune response and demonstrate an important role of an inducible "self" lncRNA acting as a potent molecular decoy actively saturating RIG-I binding sites to restrict the duration of "non-self" RNA-induced innate immune response and maintaining immune homeostasis, with potential utility in inflammatory disease management.


Subject(s)
DEAD Box Protein 58/metabolism , Immunity, Innate , RNA, Long Noncoding/metabolism , Animals , HEK293 Cells , Humans , Interferon-alpha/metabolism , Interferon-beta/metabolism , Macrophages/cytology , Macrophages/immunology , Macrophages/virology , Mice , Mice, Inbred C57BL , Protein Binding , RAW 264.7 Cells , RNA Interference , RNA, Double-Stranded/metabolism , RNA, Long Noncoding/antagonists & inhibitors , RNA, Long Noncoding/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Vesiculovirus/pathogenicity
5.
Cell ; 173(3): 634-648.e12, 2018 04 19.
Article in English | MEDLINE | ID: mdl-29606356

ABSTRACT

Identifying tumor-induced leukocyte subsets and their derived circulating factors has been instrumental in understanding cancer as a systemic disease. Nevertheless, how primary tumor-induced non-leukocyte populations in distal organs contribute to systemic spread remains poorly defined. Here, we report one population of tumor-inducible, erythroblast-like cells (Ter-cells) deriving from megakaryocyte-erythroid progenitor cells with a unique Ter-119+CD45-CD71+ phenotype. Ter-cells are enriched in the enlarged spleen of hosts bearing advanced tumors and facilitate tumor progression by secreting neurotrophic factor artemin into the blood. Transforming growth factor ß (TGF-ß) and Smad3 activation are important in Ter-cell generation. In vivo blockade of Ter-cell-derived artemin inhibits hepatocellular carcinoma (HCC) growth, and artemin deficiency abolishes Ter-cells' tumor-promoting ability. We confirm the presence of splenic artemin-positive Ter-cells in human HCC patients and show that significantly elevated serum artemin correlates with poor prognosis. We propose that Ter-cells and the secreted artemin play important roles in cancer progression with prognostic and therapeutic implications.


Subject(s)
Disease Progression , Erythroblasts/cytology , Nerve Tissue Proteins/blood , Spleen/cytology , Transforming Growth Factor beta/metabolism , Animals , Apoptosis , Carcinoma, Hepatocellular/metabolism , Cell Movement , Cell Proliferation , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Glial Cell Line-Derived Neurotrophic Factor Receptors/metabolism , Hep G2 Cells , Humans , Leukocyte Common Antigens/metabolism , Leukocytes/cytology , Liver Neoplasms/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Invasiveness/genetics , Signal Transduction
6.
Nat Immunol ; 21(4): 477-478, 2020 04.
Article in English | MEDLINE | ID: mdl-32099101

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

7.
Nat Immunol ; 21(3): 355, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32034311

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

8.
Cell ; 170(3): 492-506.e14, 2017 Jul 27.
Article in English | MEDLINE | ID: mdl-28753426

ABSTRACT

Interferon-α (IFNα) signaling is essential for antiviral response via induction of IFN-stimulated genes (ISGs). Through a non-biased high-throughput RNAi screening of 711 known epigenetic modifiers in cellular models of IFNα-mediated inhibition of HBV replication, we identified methyltransferase SETD2 as a critical amplifier of IFNα-mediated antiviral immunity. Conditional knockout mice with hepatocyte-specific deletion of Setd2 exhibit enhanced HBV infection. Mechanistically, SETD2 directly mediates STAT1 methylation on lysine 525 via its methyltransferase activity, which reinforces IFN-activated STAT1 phosphorylation and antiviral cellular response. In addition, SETD2 selectively catalyzes the tri-methylation of H3K36 on promoters of some ISGs such as ISG15, leading to gene activation. Our study identifies STAT1 methylation on K525 catalyzed by the methyltransferase SETD2 as an essential signaling event for IFNα-dependent antiviral immunity and indicates potential of SETD2 in controlling viral infections.


Subject(s)
Hepatitis B virus/physiology , Hepatitis B, Chronic/immunology , Histone-Lysine N-Methyltransferase/metabolism , Interferon-alpha/immunology , STAT1 Transcription Factor/genetics , Animals , Cell Line , Cell Line, Tumor , Epigenesis, Genetic , Hepatitis B, Chronic/virology , Hepatocytes/metabolism , Histones/metabolism , Humans , Mice , Phosphorylation , Protein Domains , RNA Interference , Transcription, Genetic , Virus Replication
9.
Nat Immunol ; 20(12): 1621-1630, 2019 12.
Article in English | MEDLINE | ID: mdl-31740800

ABSTRACT

Interferon-γ (IFN-γ) is essential for the innate immune response to intracellular bacteria. Noncoding RNAs and RNA-binding proteins (RBPs) need to be further considered in studies of regulation of the IFN-γ-activated signaling pathway in macrophages. In the present study, we found that the microRNA miR-1 promoted IFN-γ-mediated clearance of Listeria monocytogenes in macrophages by indirectly stabilizing the Stat1 messenger RNA through the degradation of the cytoplasmic long noncoding RNA Sros1. Inducible degradation or genetic loss of Sros1 led to enhanced IFN-γ-dependent activation of the innate immune response. Mechanistically, Sros1 blocked the binding of Stat1 mRNA to the RBP CAPRIN1, which stabilized the Stat1 mRNA and, consequently, promoted IFN-γ-STAT1-mediated innate immunity. These observations shed light on the complex RNA-RNA regulatory networks involved in cytokine-initiated innate responses in host-pathogen interactions.


Subject(s)
Cytoplasm/metabolism , Listeria monocytogenes/physiology , Listeriosis/immunology , Macrophages/immunology , RNA, Long Noncoding/genetics , RNA, Messenger/genetics , STAT1 Transcription Factor/metabolism , Animals , Cell Cycle Proteins/metabolism , Immunity, Innate , Interferon-gamma/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics , Protein Binding , RAW 264.7 Cells , RNA Stability , RNA, Long Noncoding/metabolism , STAT1 Transcription Factor/genetics
10.
Nat Immunol ; 20(7): 812-823, 2019 07.
Article in English | MEDLINE | ID: mdl-31036902

ABSTRACT

The helicase RIG-I initiates an antiviral immune response after recognition of pathogenic RNA. TRIM25, an E3 ubiquitin ligase, mediates K63-linked ubiquitination of RIG-I, which is crucial for RIG-I downstream signaling and the antiviral innate immune response. The components and mode of the RIG-I-initiated innate signaling remain to be fully understood. Here we identify a novel long noncoding RNA (Lnczc3h7a) that binds to TRIM25 and promotes RIG-I-mediated antiviral innate immune responses. Depletion of Lnczc3h7a impairs RIG-I signaling and the antiviral innate response to RNA viruses in vitro and in vivo. Mechanistically, Lnczc3h7a binds to both TRIM25 and activated RIG-I, serving as a molecular scaffold for stabilization of the RIG-I-TRIM25 complex at the early stage of viral infection. Lnczc3h7a facilitates TRIM25-mediated K63-linked ubiquitination of RIG-I and thus promotes downstream signaling transduction. Our findings reveal that host RNAs can enhance the response of innate immune sensors to foreign RNAs, ensuring effective antiviral defense.


Subject(s)
DEAD Box Protein 58/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Immunity, Innate/genetics , RNA, Long Noncoding/genetics , Transcription Factors/genetics , Animals , Cell Line , Humans , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Macrophages, Peritoneal/virology , Mice , Models, Biological , RNA Interference , RNA Viruses/immunology , Signal Transduction , Virus Diseases/genetics , Virus Diseases/immunology , Virus Diseases/metabolism , Virus Diseases/virology
13.
Nat Immunol ; 19(1): 41-52, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29242538

ABSTRACT

Prolonged activation of interferon-STAT1 signaling is closely related to inflammatory autoimmune disorders, and therefore the identification of negative regulators of these pathways is important. Through high-content screening of 115 mouse RING-domain E3 ligases, we identified the E3 ubiquitin ligase RNF2 as a potent inhibitor of interferon-dependent antiviral responses. RNF2 deficiency substantially enhanced interferon-stimulated gene (ISG) expression and antiviral responses. Mechanistically, nuclear RNF2 directly bound to STAT1 after interferon stimulation and increased K33-linked polyubiquitination of the DNA-binding domain of STAT1 at position K379, in addition to promoting the disassociation of STAT1/STAT2 from DNA and consequently suppressing ISG transcription. Our study provides insight into the regulation of interferon-dependent responses via a previously unrecognized post-translational modification of STAT1 in the nucleus.


Subject(s)
DNA/metabolism , Interferon Type I/pharmacology , Lysine/metabolism , Polycomb Repressive Complex 1/metabolism , STAT1 Transcription Factor/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Antiviral Agents/pharmacology , Cell Line , Gene Expression/drug effects , Lysine/genetics , Macrophages/metabolism , Macrophages/virology , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Polycomb Repressive Complex 1/genetics , Protein Binding/drug effects , STAT1 Transcription Factor/genetics , STAT2 Transcription Factor/genetics , STAT2 Transcription Factor/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitination/drug effects , Vesicular Stomatitis/genetics , Vesicular Stomatitis/prevention & control , Vesicular Stomatitis/virology , Vesicular stomatitis Indiana virus/physiology
14.
Nat Immunol ; 18(10): 1094-1103, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28846086

ABSTRACT

DEAD-box (DDX) helicases are vital for the recognition of RNA and metabolism and are critical for the initiation of antiviral innate immunity. Modification of RNA is involved in many biological processes; however, its role in antiviral innate immunity has remained unclear. Here we found that nuclear DDX member DDX46 inhibited the production of type I interferons after viral infection. DDX46 bound Mavs, Traf3 and Traf6 transcripts (which encode signaling molecules involved in antiviral responses) via their conserved CCGGUU element. After viral infection, DDX46 recruited ALKBH5, an 'eraser' of the RNA modification N6-methyladenosine (m6A), via DDX46's DEAD helicase domain to demethylate those m6A-modified antiviral transcripts. It consequently enforced their retention in the nucleus and therefore prevented their translation and inhibited interferon production. DDX46 also suppressed antiviral innate immunity in vivo. Thus, DDX46 inhibits antiviral innate responses by entrapping selected antiviral transcripts in the nucleus by erasing their m6A modification, a modification normally required for export from the nucleus and translation.


Subject(s)
Adenine/analogs & derivatives , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Immunity, Innate/genetics , Transcription, Genetic , Adenine/metabolism , AlkB Homolog 5, RNA Demethylase/genetics , AlkB Homolog 5, RNA Demethylase/metabolism , Animals , Binding Sites , Cell Line , Cell Nucleus/metabolism , Gene Expression , Gene Knockdown Techniques , Humans , Interferon Type I/biosynthesis , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Knockout , Nucleotide Motifs , Protein Binding , RNA Transport , RNA, Messenger/genetics , RNA, Messenger/metabolism , Vesicular Stomatitis/genetics , Vesicular Stomatitis/immunology , Vesicular Stomatitis/metabolism , Vesiculovirus/physiology , Virus Replication
15.
Nat Immunol ; 18(12): 1361, 2017 11 16.
Article in English | MEDLINE | ID: mdl-29144498

ABSTRACT

This corrects the article DOI: 10.1038/ni.3830.

16.
Immunity ; 53(6): 1168-1181.e7, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33326766

ABSTRACT

Viruses have evolved multiple strategies to evade elimination by the immune system. Here we examined the contribution of host long noncoding RNAs (lncRNAs) in viral immune evasion. By functional screening of lncRNAs whose expression decreased upon viral infection of macrophages, we identified a lncRNA (lncRNA-GM, Gene Symbol: AK189470.1) that promoted type I interferon (IFN-I) production and inhibited viral replication. Deficiency of lncRNA-GM in mice increased susceptibility to viral infection and impaired IFN-I production. Mechanistically, lncRNA-GM bound to glutathione S-transferase M1 (GSTM1) and blocked GSTM1 interaction with the kinase TBK1, reducing GSTM1-mediated S-glutathionylation of TBK1. Decreased S-glutathionylation enhanced TBK1 activity and downstream production of antiviral mediators. Viral infection reprogrammed intracellular glutathione metabolism and furthermore, an oxidized glutathione mimetic could inhibit TBK1 activity and promote viral replication. Our findings reveal regulation of TBK1 by S-glutathionylation and provide insight into the viral mediated metabolic changes that impact innate immunity and viral evasion.


Subject(s)
Glutathione/metabolism , Immune Evasion , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/metabolism , RNA, Long Noncoding/metabolism , Animals , Glutathione Transferase/metabolism , Humans , Immunity, Innate , Interferon Regulatory Factor-3/metabolism , Interferon Type I/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice , RNA, Long Noncoding/genetics , Signal Transduction , Virus Diseases/genetics , Virus Diseases/immunology , Virus Diseases/metabolism , Virus Replication
17.
Nat Immunol ; 22(11): 1360-1362, 2021 11.
Article in English | MEDLINE | ID: mdl-34671145
18.
Nat Immunol ; 17(10): 1167-75, 2016 10.
Article in English | MEDLINE | ID: mdl-27548433

ABSTRACT

CD8α(+) dendritic cells (DCs) are specialized at cross-presenting extracellular antigens on major histocompatibility complex (MHC) class I molecules to initiate cytotoxic T lymphocyte (CTL) responses; however, details of the mechanisms that regulate cross-presentation remain unknown. We found lower expression of the lectin family member Siglec-G in CD8α(+) DCs, and Siglec-G deficient (Siglecg(-/-)) mice generated more antigen-specific CTLs to inhibit intracellular bacterial infection and tumor growth. MHC class I-peptide complexes were more abundant on Siglecg(-/-) CD8α(+) DCs than on Siglecg(+/+) CD8α(+) DCs. Mechanistically, phagosome-expressed Siglec-G recruited the phosphatase SHP-1, which dephosphorylated the NADPH oxidase component p47(phox) and inhibited the activation of NOX2 on phagosomes. This resulted in excessive hydrolysis of exogenous antigens, which led to diminished formation of MHC class I-peptide complexes for cross-presentation. Therefore, Siglec-G inhibited DC cross-presentation by impairing such complex formation, and our results add insight into the regulation of cross-presentation in adaptive immunity.


Subject(s)
Cross-Priming , Dendritic Cells/immunology , Lectins/metabolism , Listeria monocytogenes/immunology , Listeriosis/immunology , Neoplasms, Experimental/immunology , Receptors, Antigen, B-Cell/metabolism , T-Lymphocytes, Cytotoxic/immunology , Animals , Antigens/metabolism , CD8 Antigens/metabolism , Histocompatibility Antigens Class I/metabolism , Lectins/genetics , Lymphocyte Activation , Melanoma, Experimental , Mice , Mice, Inbred C57BL , Mice, Knockout , NADPH Oxidases/metabolism , Peptide Fragments/metabolism , Phagocytosis/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Receptors, Antigen, B-Cell/genetics , Sialic Acid Binding Immunoglobulin-like Lectins , Signal Transduction , Tumor Burden/genetics
19.
Nat Immunol ; 17(7): 806-15, 2016 07.
Article in English | MEDLINE | ID: mdl-27240213

ABSTRACT

The DNA methyltransferase Dnmt3a has high expression in terminally differentiated macrophages; however, its role in innate immunity remains unknown. Here we report that deficiency in Dnmt3a selectively impaired the production of type I interferons triggered by pattern-recognition receptors (PRRs), but not that of the proinflammatory cytokines TNF and IL-6. Dnmt3a-deficient mice exhibited enhanced susceptibility to viral challenge. Dnmt3a did not directly regulate the transcription of genes encoding type I interferons; instead, it increased the production of type I interferons through an epigenetic mechanism by maintaining high expression of the histone deacetylase HDAC9. In turn, HDAC9 directly maintained the deacetylation status of the key PRR signaling molecule TBK1 and enhanced its kinase activity. Our data add mechanistic insight into the crosstalk between epigenetic modifications and post-translational modifications in the regulation of PRR signaling and activation of antiviral innate immune responses.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/metabolism , Immunity, Innate , Macrophages/immunology , Rhabdoviridae Infections/immunology , Vesicular stomatitis Indiana virus/immunology , Acetylation , Animals , DNA Methyltransferase 3A , Epigenesis, Genetic , HEK293 Cells , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Humans , Interferon Type I/metabolism , Macrophages/virology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Protein Serine-Threonine Kinases/metabolism , RAW 264.7 Cells , Receptors, Pattern Recognition/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Signal Transduction
20.
Immunity ; 50(3): 600-615.e15, 2019 03 19.
Article in English | MEDLINE | ID: mdl-30824325

ABSTRACT

CCR7 chemokine receptor stimulation induces rapid but transient dendritic cell (DC) migration toward draining lymph nodes, which is critical for the initiation of protective immunity and maintenance of immune homeostasis. The mechanisms for terminating CCR7-mediated DC migration remain incompletely understood. Here we have identified a long non-coding RNA lnc-Dpf3 whose feedback restrained CCR7-mediated DC migration. CCR7 stimulation upregulated lnc-Dpf3 via removing N6-methyladenosine (m6A) modification to prevent RNA degradation. DC-specific lnc-Dpf3 deficiency increased CCR7-mediated DC migration, leading to exaggerated adaptive immune responses and inflammatory injuries. Mechanistically, CCR7 stimulation activated the HIF-1α transcription factor pathway in DCs, leading to metabolic reprogramming toward glycolysis for DC migration. lnc-Dpf3 directly bound to HIF-1α and suppressed HIF-1α-dependent transcription of the glycolytic gene Ldha, thus inhibiting DC glycolytic metabolism and migratory capacity. We demonstrate a critical role for CCR7-inducible lnc-Dpf3 in coupling epigenetic and metabolic pathways to feedback-control DC migration and inflammatory responses.


Subject(s)
Cell Movement/genetics , DNA-Binding Proteins/genetics , Glycolysis/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Receptors, CCR7/genetics , Transcription Factors/genetics , Adaptive Immunity/genetics , Animals , Cell Line , Dendritic Cells/pathology , Epigenesis, Genetic/genetics , Gene Expression Regulation/genetics , HEK293 Cells , Humans , Inflammation/genetics , Inflammation/pathology , Lymph Nodes/pathology , Metabolic Networks and Pathways/genetics , Mice , Mice, Inbred C57BL , Transcription, Genetic/genetics , Up-Regulation/genetics
SELECTION OF CITATIONS
SEARCH DETAIL