Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 69
Filter
1.
Mol Cell ; 81(5): 953-968.e9, 2021 03 04.
Article in English | MEDLINE | ID: mdl-33503407

ABSTRACT

While the role of transcription factors and coactivators in controlling enhancer activity and chromatin structure linked to gene expression is well established, the involvement of corepressors is not. Using inflammatory macrophage activation as a model, we investigate here a corepressor complex containing GPS2 and SMRT both genome-wide and at the Ccl2 locus, encoding the chemokine CCL2 (MCP-1). We report that corepressors co-occupy candidate enhancers along with the coactivators CBP (H3K27 acetylase) and MED1 (mediator) but act antagonistically by repressing eRNA transcription-coupled H3K27 acetylation. Genome editing, transcriptional interference, and cistrome analysis reveals that apparently related enhancer and silencer elements control Ccl2 transcription in opposite ways. 4C-seq indicates that corepressor depletion or inflammatory signaling functions mechanistically similarly to trigger enhancer activation. In ob/ob mice, adipose tissue macrophage-selective depletion of the Ccl2 enhancer-transcribed eRNA reduces metaflammation. Thus, the identified corepressor-eRNA-chemokine pathway operates inĀ vivo and suggests therapeutic opportunities by targeting eRNAs in immuno-metabolic diseases.


Subject(s)
Chemokine CCL2/genetics , Co-Repressor Proteins/genetics , Enhancer Elements, Genetic , Intracellular Signaling Peptides and Proteins/genetics , Nuclear Receptor Co-Repressor 2/genetics , Obesity/genetics , Silencer Elements, Transcriptional , Adipose Tissue/immunology , Adipose Tissue/pathology , Animals , CRISPR-Cas Systems , Chemokine CCL2/immunology , Co-Repressor Proteins/immunology , Gene Editing , Gene Expression Regulation/drug effects , HEK293 Cells , Histone Acetyltransferases/genetics , Histone Acetyltransferases/immunology , Histones/genetics , Histones/immunology , Humans , Intracellular Signaling Peptides and Proteins/immunology , Lipopolysaccharides/pharmacology , Macrophage Activation/drug effects , Male , Mediator Complex Subunit 1/genetics , Mediator Complex Subunit 1/immunology , Mice , Mice, Obese , Nuclear Receptor Co-Repressor 2/immunology , Obesity/immunology , Obesity/pathology , RAW 264.7 Cells , RNA, Untranslated/genetics , RNA, Untranslated/immunology , Signal Transduction
2.
Mol Cell ; 75(6): 1229-1242.e5, 2019 09 19.
Article in English | MEDLINE | ID: mdl-31377117

ABSTRACT

Interferon gamma (IFN-ƎĀ³), critical for host defense and tumor surveillance, requires tight control of its expression. Multiple cis-regulatory elements exist around Ifng along with a non-coding transcript, Ifng-as1 (also termed NeST). Here, we describe two genetic models generated to dissect the molecular functions of this locus and its RNA product. DNA deletion within the Ifng-as1 locus disrupted chromatin organization of the extended Ifng locus, impaired Ifng response, and compromised host defense. Insertion of a polyA signal ablated the Ifng-as1 full-length transcript and impaired host defense, while allowing proper chromatin structure. Transient knockdown of Ifng-as1 also reduced IFN-ƎĀ³ production. In humans, discordant expression of IFNG and IFNG-AS1 was evident in memory TĀ cells, with high expression of this long non-coding RNA (lncRNA) and low expression of the cytokine. These results establish Ifng-as1 as an important regulator of Ifng expression, as a DNA element and transcribed RNA, involved in dynamic and cell state-specific responses to infection.


Subject(s)
Gene Expression Regulation/immunology , Immunologic Memory , Infections/immunology , Interferon-gamma/immunology , RNA, Untranslated/immunology , T-Lymphocytes/immunology , Animals , Chromatin/genetics , Chromatin/immunology , Female , Gene Knockdown Techniques , Infections/genetics , Infections/pathology , Interferon-gamma/genetics , Mice , RNA, Untranslated/genetics , T-Lymphocytes/pathology
3.
Crit Rev Biochem Mol Biol ; 54(2): 133-152, 2019 04.
Article in English | MEDLINE | ID: mdl-31084369

ABSTRACT

Ro60, also known as SS-A or TROVE2, is an evolutionarily conserved RNA-binding protein that is found in most animal cells, approximately 5% of sequenced prokaryotic genomes and some archaea. Ro60 is present in cells as both a free protein and as a component of a ribonucleoprotein complex, where its best-known partners are members of a class of noncoding RNAs called Y RNAs. Structural and biochemical analyses have revealed that Ro60 is a ring-shaped protein that binds Y RNAs on its outer surface. In addition to Y RNAs, Ro60 binds misfolded and aberrant noncoding RNAs in some animal cell nuclei. Although the fate of these defective Ro60-bound noncoding RNAs in animal cells is not well-defined, a bacterial Ro60 ortholog functions with 3' to 5' exoribonucleases to assist structured RNA degradation. Studies of Y RNAs have revealed that these RNAs regulate the subcellular localization of Ro60, tether Ro60 to effector proteins and regulate the access of other RNAs to its central cavity. As both mammalian cells and bacteria lacking Ro60 are sensitized to ultraviolet irradiation, Ro60 function may be important during exposure to some environmental stressors. Here we summarize the current knowledge regarding the functions of Ro60 and Y RNAs in animal cells and bacteria. Because the Ro60 RNP is a clinically important target of autoantibodies in patients with rheumatic diseases such as Sjogren's syndrome, systemic lupus erythematosus, and neonatal lupus, we also discuss potential roles for Ro60 RNPs in the initiation and pathogenesis of systemic autoimmune rheumatic disease.


Subject(s)
Autoimmunity , RNA, Untranslated/immunology , Ribonucleoproteins/immunology , Animals , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/metabolism , Humans , Lupus Erythematosus, Systemic/congenital , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/metabolism , Models, Molecular , Nucleic Acid Conformation , Protein Conformation , RNA Stability , RNA, Untranslated/analysis , RNA, Untranslated/metabolism , Ribonucleoproteins/analysis , Ribonucleoproteins/metabolism
4.
Semin Cancer Biol ; 65: 65-79, 2020 10.
Article in English | MEDLINE | ID: mdl-31733291

ABSTRACT

Substantial evolution in cancer therapy has been witnessed lately, steering mainly towards immunotherapeutic approaches, replacing or in combination with classical therapies. Whereas the use of various immunotherapy approaches, such as adoptive T cell therapy, genetically-modified T cells, or immune checkpoint inhibitors, has been a triumph for cancer immunotherapy, the great challenge is the ability of the immune system to sustain long lasting anti-tumor response. Additionally, epigenetic changes in a suppressive tumor microenvironment can pertain to T cell exhaustion, limiting their functionality. Noncoding RNAs (ncRNAs) have emerged over the last years as key players in epigenetic regulation. Among those, microRNAs (miRNAs) and long noncoding RNAs (lncRNAs) have been studied extensively for their potential role in regulating tumor immunity through direct regulation of genes involved in immune activation or suppression. In this review, we will provide an overview of contemporary approaches for cancer immunotherapy and will present the current state of knowledge implicating miRNAs and lncRNAs in regulating immune response against human cancer and their potential implications in resistance to cancer immunotherapy, with main emphasis on immune checkpoints regulation.


Subject(s)
Drug Resistance, Neoplasm/immunology , Immunotherapy/adverse effects , Neoplasms/drug therapy , RNA, Untranslated/immunology , Gene Expression Regulation, Neoplastic/genetics , Humans , Immunity/immunology , Neoplasms/immunology , Neoplasms/pathology , T-Lymphocytes/immunology , Tumor Microenvironment/immunology
5.
RNA ; 25(5): 539-556, 2019 05.
Article in English | MEDLINE | ID: mdl-30770398

ABSTRACT

The interferon-inducible protein kinase R (PKR) is a key component of host innate immunity that restricts viral replication and propagation. As one of the four eIF2α kinases that sense diverse stresses and direct the integrated stress response (ISR) crucial for cell survival and proliferation, PKR's versatile roles extend well beyond antiviral defense. Targeted by numerous host and viral regulators made of RNA and proteins, PKR is subject to multiple layers of endogenous control and external manipulation, driving its rapid evolution. These versatile regulators include not only the canonical double-stranded RNA (dsRNA) that activates the kinase activity of PKR, but also highly structured viral, host, and artificial RNAs that exert a full spectrum of effects. In this review, we discuss our deepening understanding of the allosteric mechanism that connects the regulatory and effector domains of PKR, with an emphasis on diverse structured RNA regulators in comparison to their protein counterparts. Through this analysis, we conclude that much of the mechanistic details that underlie this RNA-regulated kinase await structural and functional elucidation, upon which we can then describe a "PKR code," a set of structural and chemical features of RNA that are both descriptive and predictive for their effects on PKR.


Subject(s)
Host-Pathogen Interactions/genetics , RNA, Double-Stranded/genetics , RNA, Untranslated/genetics , Virus Diseases/genetics , eIF-2 Kinase/genetics , Allosteric Regulation , Animals , Base Sequence , Binding Sites , Gene Expression Regulation , Host-Pathogen Interactions/immunology , Humans , Immunity, Innate , Interferons/genetics , Interferons/immunology , Protein Binding , Protein Interaction Domains and Motifs , Protein Structure, Secondary , RNA, Double-Stranded/chemistry , RNA, Double-Stranded/immunology , RNA, Untranslated/chemistry , RNA, Untranslated/immunology , Virus Diseases/immunology , Virus Diseases/virology , Virus Replication , eIF-2 Kinase/chemistry , eIF-2 Kinase/immunology
6.
Int J Mol Sci ; 22(24)2021 Dec 17.
Article in English | MEDLINE | ID: mdl-34948375

ABSTRACT

Pediatric autoimmune liver disorders include autoimmune hepatitis (AIH), autoimmune sclerosing cholangitis (ASC), and de novo AIH after liver transplantation. AIH is an idiopathic disease characterized by immune-mediated hepatocyte injury associated with the destruction of liver cells, causing inflammation, liver failure, and fibrosis, typically associated with autoantibodies. The etiology of AIH is not entirely unraveled, but evidence supports an intricate interaction among genetic variants, environmental factors, and epigenetic modifications. The pathogenesis of AIH comprises the interaction between specific genetic traits and molecular mimicry for disease development, impaired immunoregulatory mechanisms, including CD4+ T cell population and Treg cells, alongside other contributory roles played by CD8+ cytotoxicity and autoantibody production by B cells. These findings delineate an intricate pathway that includes gene to gene and gene to environment interactions with various drugs, viral infections, and the complex microbiome. Epigenetics emphasizes gene expression through hereditary and reversible modifications of the chromatin architecture without interfering with the DNA sequence. These alterations comprise DNA methylation, histone transformations, and non-coding small (miRNA) and long (lncRNA) RNA transcriptions. The current first-line therapy comprises prednisolone plus azathioprine to induce clinical and biochemical remission. Further understanding of the cellular and molecular mechanisms encountered in AIH may depict their impact on clinical aspects, detect biomarkers, and guide toward novel, effective, and better-targeted therapies with fewer side effects.


Subject(s)
Hepatitis, Autoimmune/pathology , Liver/pathology , Animals , Azathioprine/therapeutic use , DNA Methylation/drug effects , Epigenesis, Genetic/drug effects , Glucocorticoids/therapeutic use , Hepatitis, Autoimmune/drug therapy , Hepatitis, Autoimmune/genetics , Hepatitis, Autoimmune/immunology , Humans , Immunosuppressive Agents/therapeutic use , Liver/immunology , Liver/metabolism , Prednisolone/therapeutic use , RNA, Untranslated/genetics , RNA, Untranslated/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology
7.
Int J Mol Sci ; 22(4)2021 Feb 18.
Article in English | MEDLINE | ID: mdl-33670458

ABSTRACT

Interferons (IFNs) are a crucial component in the innate immune response. Especially the IFN-Ɵ signaling operates in most cell types and plays a key role in the first line of defense upon pathogen intrusion. The induction of IFN-Ɵ should be tightly controlled, because its hyperactivation can lead to tissue damage or autoimmune diseases. Activation of the IFN-Ɵ promoter needs Interferon Regulatory Factor 3 (IRF3), together with Nuclear Factor kappa-light-chain-enhancer of activated B cells (NF-κB) and Activator Protein 1 (AP-1). Here we report that a human noncoding RNA, nc886, is a novel suppressor for the IFN-Ɵ signaling and inflammation. Upon treatment with several pathogen-associated molecular patterns and viruses, nc886 suppresses the activation of IRF3 and also inhibits NF-κB and AP-1 via inhibiting Protein Kinase R (PKR). These events lead to decreased expression of IFN-Ɵ and resultantly IFN-stimulated genes. nc886's role might be to restrict the IFN-Ɵ signaling from hyperactivation. Since nc886 expression is regulated by epigenetic and environmental factors, nc886 might explain why innate immune responses to pathogens are variable depending on biological settings.


Subject(s)
Gene Expression Regulation/immunology , Interferon Regulatory Factor-3/immunology , Interferon Type I/immunology , RNA, Untranslated/immunology , Animals , Cell Line, Tumor , HCT116 Cells , HEK293 Cells , Host-Pathogen Interactions/immunology , Humans , Immunity, Innate/genetics , Immunity, Innate/immunology , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/metabolism , Interferon Type I/genetics , Interferon Type I/metabolism , Mice , NF-kappa B/immunology , NF-kappa B/metabolism , Promoter Regions, Genetic/genetics , RAW 264.7 Cells , RNA, Untranslated/genetics , Signal Transduction/immunology , Transcription Factor AP-1/immunology , Transcription Factor AP-1/metabolism , Viruses/immunology , eIF-2 Kinase/genetics , eIF-2 Kinase/immunology , eIF-2 Kinase/metabolism
8.
J Immunol ; 198(4): 1595-1605, 2017 02 15.
Article in English | MEDLINE | ID: mdl-28077601

ABSTRACT

Among innovative adjuvants conferring a Th1-shift, RNAdjuvant is a promising candidate. This adjuvant consists of a 547-nt uncapped noncoding ssRNA containing polyU repeats that is stabilized by a cationic carrier peptide. Whereas vaccination of mice with an influenza subunit vaccine induced moderate virus-specific IgG1, vaccination together with RNAdjuvant significantly enhanced this IgG1 and additionally promoted the formation of IgG2b/c, which is indicative of Th1 responses. Furthermore, such sera neutralized influenza virus, whereas this effect was not detected upon vaccination with the subunit vaccine alone. Similarly, upon vaccination with virus-like particles displaying vesicular stomatitis virus G protein, RNAdjuvant promoted the formation of virus-specific IgG2b/c and enhanced neutralizing IgG responses to an extent that mice were protected against lethal virus infection. RNAdjuvant induced dendritic cells to upregulate activation markers and produce IFN-I. Although these effects were strictly TLR7 dependent, RNAdjuvant-mediated augmentation of vaccine responses needed concurrent TLR and RIG-I-like helicase signaling. This was indicated by the absence of the adjuvant effect in vaccinated MyD88-/-Cardif-/- mice, which are devoid of TLR (with the exception of TLR3) and RIG-I-like helicase signaling, whereas in vaccinated MyD88-/- mice the adjuvant effect was reduced. Notably, i.m. RNAdjuvant injection induced local IFN-I responses and did not induce systemic effects, implying good tolerability and a favorable safety profile for RNAdjuvant.


Subject(s)
Adjuvants, Immunologic , Immunoglobulin G/blood , Influenza Vaccines/immunology , Membrane Glycoproteins/immunology , RNA, Untranslated/immunology , Toll-Like Receptor 7/immunology , Vaccines, Virus-Like Particle/immunology , Adjuvants, Immunologic/adverse effects , Animals , Antibodies, Viral/blood , DEAD Box Protein 58/immunology , DEAD Box Protein 58/metabolism , Immunoglobulin G/immunology , Influenza Vaccines/administration & dosage , Membrane Glycoproteins/administration & dosage , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred BALB C , Myeloid Differentiation Factor 88/deficiency , Myeloid Differentiation Factor 88/metabolism , Th1 Cells/immunology , Toll-Like Receptor 7/metabolism , Vaccination , Vaccines, Subunit/immunology , Vaccines, Virus-Like Particle/administration & dosage , Viral Envelope Proteins/administration & dosage , Viral Envelope Proteins/immunology
9.
Proc Natl Acad Sci U S A ; 112(49): 15154-9, 2015 Dec 08.
Article in English | MEDLINE | ID: mdl-26575629

ABSTRACT

Recent studies have demonstrated abundant transcription of a set of noncoding RNAs (ncRNAs) preferentially within tumors as opposed to normal tissue. Using an approach from statistical physics, we quantify global transcriptome-wide motif use for the first time, to our knowledge, in human and murine ncRNAs, determining that most have motif use consistent with the coding genome. However, an outlier subset of tumor-associated ncRNAs, typically of recent evolutionary origin, has motif use that is often indicative of pathogen-associated RNA. For instance, we show that the tumor-associated human repeat human satellite repeat II (HSATII) is enriched in motifs containing CpG dinucleotides in AU-rich contexts that most of the human genome and human adapted viruses have evolved to avoid. We demonstrate that a key subset of these ncRNAs functions as immunostimulatory "self-agonists" and directly activates cells of the mononuclear phagocytic system to produce proinflammatory cytokines. These ncRNAs arise from endogenous repetitive elements that are normally silenced, yet are often very highly expressed in cancers. We propose that the innate response in tumors may partially originate from direct interaction of immunogenic ncRNAs expressed in cancer cells with innate pattern recognition receptors, and thereby assign a previously unidentified danger-associated function to a set of dark matter repetitive elements. These findings potentially reconcile several observations concerning the role of ncRNA expression in cancers and their relationship to the tumor microenvironment.


Subject(s)
Neoplasms/genetics , RNA, Untranslated/immunology , Animals , Humans , Immunity, Innate , Mice , Neoplasms/immunology
10.
Proc Natl Acad Sci U S A ; 112(46): E6293-300, 2015 Nov 17.
Article in English | MEDLINE | ID: mdl-26578789

ABSTRACT

Telomeric repeat-containing RNA (TERRA) has been identified as a telomere-associated regulator of chromosome end protection. Here, we report that TERRA can also be found in extracellular fractions that stimulate innate immune signaling. We identified extracellular forms of TERRA in mouse tumor and embryonic brain tissue, as well as in human tissue culture cell lines using RNA in situ hybridization. RNA-seq analyses revealed TERRA to be among the most highly represented transcripts in extracellular fractions derived from both normal and cancer patient blood plasma. Cell-free TERRA (cfTERRA) could be isolated from the exosome fractions derived from human lymphoblastoid cell line (LCL) culture media. cfTERRA is a shorter form (Ć¢ĀˆĀ¼200 nt) of cellular TERRA and copurifies with CD63- and CD83-positive exosome vesicles that could be visualized by cyro-electron microscopy. These fractions were also enriched for histone proteins that physically associate with TERRA in extracellular ChIP assays. Incubation of cfTERRA-containing exosomes with peripheral blood mononuclear cells stimulated transcription of several inflammatory cytokine genes, including TNFα, IL6, and C-X-C chemokine 10 (CXCL10) Exosomes engineered with elevated TERRA or liposomes with synthetic TERRA further stimulated inflammatory cytokines, suggesting that exosome-associated TERRA augments innate immune signaling. These findings imply a previously unidentified extrinsic function for TERRA and a mechanism of communication between telomeres and innate immune signals in tissue and tumor microenvironments.


Subject(s)
Exosomes/immunology , Immunity, Innate , Neoplasms/immunology , RNA, Untranslated/immunology , Signal Transduction/immunology , Telomere , Animals , Antigens, CD/blood , Antigens, CD/genetics , Antigens, CD/immunology , Cell Line, Tumor , Cytokines/blood , Cytokines/genetics , Cytokines/immunology , Exosomes/genetics , Exosomes/metabolism , Histones/blood , Histones/genetics , Histones/immunology , Humans , Immunoglobulins/blood , Immunoglobulins/genetics , Immunoglobulins/immunology , Inflammation/blood , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Membrane Glycoproteins/blood , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mice , Neoplasms/blood , Neoplasms/genetics , Neoplasms/pathology , RNA, Untranslated/blood , RNA, Untranslated/genetics , Signal Transduction/genetics , Tetraspanin 30/blood , Tetraspanin 30/genetics , Tetraspanin 30/immunology , CD83 Antigen
11.
Immunol Rev ; 253(1): 97-111, 2013 May.
Article in English | MEDLINE | ID: mdl-23550641

ABSTRACT

RNA interference (RNAi) is an ancient process by which non-coding RNAs regulate gene expression in a sequence-specific manner. The core components of RNAi are small regulatory RNAs, approximately 21-30 nucleotides in length, including small interfering RNAs (siRNAs) and microRNAs (miRNAs). The past two decades have seen considerable progress in our understanding of the molecular mechanisms underlying the biogenesis of siRNAs and miRNAs. Recent advances have also revealed the crucial regulatory roles played by small RNAs in such diverse processes as development, homeostasis, innate immunity, and oncogenesis. Accumulating evidence indicates that RNAi initially evolved as a host defense mechanism against viruses and transposons. The ability of the host small RNA biogenesis machinery to recognize viral double-stranded RNA replication intermediates and transposon transcripts is critical to this process, as is small RNA-guided targeting of RNAs via complementary base pairing. Collectively, these properties confer unparalleled specificity and precision to RNAi-mediated gene silencing as an effective antiviral mechanism.


Subject(s)
Host-Pathogen Interactions , RNA, Untranslated/immunology , RNA, Viral/immunology , Virus Diseases/immunology , Viruses/immunology , Animals , Biological Evolution , Gene Expression Regulation , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , RNA Interference , Virus Diseases/genetics , Viruses/genetics
12.
PLoS Pathog ; 10(7): e1004242, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24992036

ABSTRACT

Viral RNA-host protein interactions are critical for replication of flaviviruses, a genus of positive-strand RNA viruses comprising major vector-borne human pathogens including dengue viruses (DENV). We examined three conserved host RNA-binding proteins (RBPs) G3BP1, G3BP2 and CAPRIN1 in dengue virus (DENV-2) infection and found them to be novel regulators of the interferon (IFN) response against DENV-2. The three RBPs were required for the accumulation of the protein products of several interferon stimulated genes (ISGs), and for efficient translation of PKR and IFITM2 mRNAs. This identifies G3BP1, G3BP2 and CAPRIN1 as novel regulators of the antiviral state. Their antiviral activity was antagonized by the abundant DENV-2 non-coding subgenomic flaviviral RNA (sfRNA), which bound to G3BP1, G3BP2 and CAPRIN1, inhibited their activity and lead to profound inhibition of ISG mRNA translation. This work describes a new and unexpected level of regulation for interferon stimulated gene expression and presents the first mechanism of action for an sfRNA as a molecular sponge of anti-viral effectors in human cells.


Subject(s)
Carrier Proteins/immunology , Cell Cycle Proteins/immunology , Dengue Virus/immunology , Protein Biosynthesis/immunology , RNA, Messenger/immunology , RNA, Untranslated/immunology , RNA, Viral/immunology , Adaptor Proteins, Signal Transducing , Animals , Base Sequence , Carrier Proteins/genetics , Cell Cycle Proteins/genetics , Cricetinae , DNA Helicases , Humans , Membrane Proteins/genetics , Membrane Proteins/immunology , Molecular Sequence Data , Poly-ADP-Ribose Binding Proteins , RNA Helicases , RNA Recognition Motif Proteins , RNA, Messenger/genetics , RNA, Untranslated/genetics , RNA, Viral/genetics , RNA-Binding Proteins , eIF-2 Kinase/genetics , eIF-2 Kinase/immunology
13.
J Immunol ; 193(7): 3746-54, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25187654

ABSTRACT

Our previous studies have shown that DNase I hypersensitive sites 1 and 2 (HS1-2) and HS3-6 within the mouse Vκ-Jκ intervening region are essential for controlling locus contraction and creating a diverse Ab repertoire. In this article, we demonstrate that a 6.3-kb deletion encompassing HS1-6 altogether not only leads to the predictable sums of these phenotypes, but also results in a novel hyperelevation of transcription of proximal Vκ genes, in both pre-B and splenic B cells. These findings reveal previously unrecognized additional functions for cis-elements within the Vκ-Jκ intervening region, namely, prevention of the production of massive levels of noncoding RNA species by silencing transcription of germline proximal Vκ genes in both developing and mature B cells.


Subject(s)
Immunoglobulin Joining Region/immunology , Immunoglobulin kappa-Chains/immunology , Precursor Cells, B-Lymphoid/immunology , Spleen/immunology , Transcription, Genetic/immunology , Animals , Gene Silencing/immunology , Immunoglobulin Joining Region/genetics , Immunoglobulin kappa-Chains/genetics , Mice , Mice, Mutant Strains , Precursor Cells, B-Lymphoid/cytology , RNA, Untranslated/genetics , RNA, Untranslated/immunology , Spleen/cytology , Transcription, Genetic/genetics
14.
Adv Exp Med Biol ; 909: 69-138, 2016.
Article in English | MEDLINE | ID: mdl-27240457

ABSTRACT

Biological response modifiers (BRMs) emerge as a lay of new compounds or approaches used in improving cancer immunotherapy. Evidences highlight that cytokines, Toll-like receptor (TLR) signaling, and noncoding RNAs are of crucial roles in modulating antitumor immune response and cancer-related chronic inflammation, and BRMs based on them have been explored. In particular, besides some cytokines like IFN-α and IL-2, several Toll-like receptor (TLR) agonists like BCG, MPL, and imiquimod are also licensed to be used in patients with several malignancies nowadays, and the first artificial small noncoding RNA (microRNA) mimic, MXR34, has entered phase I clinical study against liver cancer, implying their potential application in cancer therapy. According to amounts of original data, this chapter will review the regulatory roles of TLR signaling, some noncoding RNAs, and several key cytokines in cancer and cancer-related immune response, as well as the clinical cases in cancer therapy based on them.


Subject(s)
Aminoquinolines/therapeutic use , Antineoplastic Agents/therapeutic use , Gene Expression Regulation, Neoplastic/immunology , Immunologic Factors/therapeutic use , Immunotherapy/methods , Neoplasms/therapy , Clinical Trials as Topic , Humans , Imiquimod , Interferon-alpha/genetics , Interferon-alpha/immunology , Interleukin-2/genetics , Interleukin-2/immunology , Mycobacterium bovis/genetics , Mycobacterium bovis/immunology , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/pathology , RNA, Untranslated/genetics , RNA, Untranslated/immunology , RNA, Untranslated/therapeutic use , Receptors, Thrombopoietin/genetics , Receptors, Thrombopoietin/immunology , Signal Transduction , Toll-Like Receptors/agonists , Toll-Like Receptors/genetics , Toll-Like Receptors/immunology
15.
Z Rheumatol ; 75(4): 402-5, 2016 May.
Article in German | MEDLINE | ID: mdl-27115697

ABSTRACT

Non-coding regulatory ribonucleic acids (RNA), including microRNA, long non-coding RNA and circular RNA, can influence the expression of genes mediating inflammatory processes and therefore affect the course and progression of chronic inflammatory diseases. Recent studies using antisense oligonucleotides suggest that such non-coding regulatory RNAs are suitable as novel therapeutic target molecules for the treatment of inflammatory rheumatic diseases.


Subject(s)
Inflammation/genetics , Inflammation/immunology , MicroRNAs/genetics , MicroRNAs/immunology , RNA, Untranslated/genetics , RNA, Untranslated/immunology , Chronic Disease , Evidence-Based Medicine , Humans , Immunogenetic Phenomena , Regulatory Sequences, Ribonucleic Acid/genetics , Regulatory Sequences, Ribonucleic Acid/immunology
16.
Infect Immun ; 83(3): 863-75, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25510881

ABSTRACT

Pseudomonas aeruginosa is an opportunistic pathogen that requires iron to cause infection, but it also must regulate the uptake of iron to avoid iron toxicity. The iron-responsive PrrF1 and PrrF2 small regulatory RNAs (sRNAs) are part of P. aeruginosa's iron regulatory network and affect the expression of at least 50 genes encoding iron-containing proteins. The genes encoding the PrrF1 and PrrF2 sRNAs are encoded in tandem in P. aeruginosa, allowing for the expression of a distinct, heme-responsive sRNA named PrrH that appears to regulate genes involved in heme metabolism. Using a combination of growth, mass spectrometry, and gene expression analysis, we showed that the ΔprrF1,2 mutant, which lacks expression of the PrrF and PrrH sRNAs, is defective for both iron and heme homeostasis. We also identified phuS, encoding a heme binding protein involved in heme acquisition, and vreR, encoding a previously identified regulator of P. aeruginosa virulence genes, as novel targets of prrF-mediated heme regulation. Finally, we showed that the prrF locus encoding the PrrF and PrrH sRNAs is required for P. aeruginosa virulence in a murine model of acute lung infection. Moreover, we showed that inoculation with a ΔprrF1,2 deletion mutant protects against future challenge with wild-type P. aeruginosa. Combined, these data demonstrate that the prrF-encoded sRNAs are critical regulators of P. aeruginosa virulence.


Subject(s)
Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial , Iron/metabolism , Pseudomonas Infections/prevention & control , Pseudomonas aeruginosa/genetics , RNA, Untranslated/metabolism , Acute Disease , Animals , Bacterial Proteins/genetics , Base Sequence , Carrier Proteins/genetics , Carrier Proteins/metabolism , Heme/metabolism , Heme-Binding Proteins , Hemeproteins/genetics , Hemeproteins/metabolism , Homeostasis , Humans , Immunization , Lung/microbiology , Lung/pathology , Mice , Molecular Sequence Data , Pseudomonas Infections/immunology , Pseudomonas Infections/microbiology , Pseudomonas Infections/pathology , Pseudomonas aeruginosa/immunology , Pseudomonas aeruginosa/metabolism , Pseudomonas aeruginosa/pathogenicity , RNA, Untranslated/administration & dosage , RNA, Untranslated/genetics , RNA, Untranslated/immunology , Sequence Deletion , Virulence
17.
Immunol Cell Biol ; 93(3): 271-6, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25776989

ABSTRACT

Autoimmune diseases are complex disorders of largely unknown etiology. Genetic studies have identified a limited number of causal genes from a marginal number of individuals, and demonstrated a high degree of discordance in monozygotic twins. Studies have begun to reveal epigenetic contributions to these diseases, primarily through the study of DNA methylation, but chromatin and non-coding RNA changes are also emerging. Moving forward an integrative analysis of genomic, transcriptomic and epigenomic data, with the latter two coming from specific cell types, will provide an understanding that has been missed from genetics alone. We provide an overview of the current state of the field and vision for deriving the epigenomics of autoimmunity.


Subject(s)
Autoimmune Diseases/genetics , Epigenesis, Genetic , Epigenomics/methods , Animals , Autoimmune Diseases/immunology , Autoimmunity/genetics , Chromatin Assembly and Disassembly , DNA Methylation , Epigenomics/trends , Humans , RNA, Untranslated/immunology
18.
Immunol Cell Biol ; 93(3): 233-44, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25559622

ABSTRACT

The interaction of innate immune cells with pathogens leads to changes in gene expression that elicit our body's first line of defense against infection. Although signaling pathways and transcription factors have a central role, it is becoming increasingly clear that epigenetic factors, in the form of DNA or histone modifications, as well as noncoding RNAs, are critical for generating the necessary cell lineage as well as context-specific gene expression in diverse innate immune cell types. Much of the epigenetic landscape is set during cellular differentiation; however, pathogens and other environmental triggers also induce changes in histone modifications that can either promote tolerance or 'train' innate immune cells for a more robust antigen-independent secondary response. Here we review the important contribution of epigenetic factors to the initiation, maintenance and training of innate immune responses. In addition, we explore how pathogens have hijacked these mechanisms for their benefit and the potential of small molecules targeting chromatin machinery as a way to boost or subdue the innate immune response in disease.


Subject(s)
Epigenesis, Genetic , Host-Pathogen Interactions/genetics , Immunity, Innate/genetics , RNA, Untranslated/immunology , Animals , Cell Differentiation , Cell Lineage , Cellular Microenvironment , DNA Methylation , Gene Expression Regulation/immunology , Histones/metabolism , Humans , Immunomodulation , Protein Processing, Post-Translational , Signal Transduction/immunology
19.
Circ Res ; 113(5): 588-602, 2013 Aug 16.
Article in English | MEDLINE | ID: mdl-23948584

ABSTRACT

Understanding of the roles of noncoding RNAs (ncRNAs) within complex organisms has fundamentally changed. It is increasingly possible to use ncRNAs as diagnostic and therapeutic tools in medicine. Regarding disease pathogenesis, it has become evident that confinement to the analysis of protein-coding regions of the human genome is insufficient because ncRNA variants have been associated with important human diseases. Thus, inclusion of noncoding genomic elements in pathogenetic studies and their consideration as therapeutic targets is warranted. We consider aspects of the evolutionary and discovery history of ncRNAs, as far as they are relevant for the identification and selection of ncRNAs with likely therapeutic potential. Novel therapeutic strategies are based on ncRNAs, and we discuss here RNA interference as a highly versatile tool for gene silencing. RNA interference-mediating RNAs are small, but only parts of a far larger spectrum encompassing ncRNAs up to many kilobasepairs in size. We discuss therapeutic options in cardiovascular medicine offered by ncRNAs and key issues to be solved before clinical translation. Convergence of multiple technical advances is highlighted as a prerequisite for the translational progress achieved in recent years. Regarding safety, we review properties of RNA therapeutics, which may immunologically distinguish them from their endogenous counterparts, all of which underwent sophisticated evolutionary adaptation to specific biological contexts. Although our understanding of the noncoding human genome is only fragmentary to date, it is already feasible to develop RNA interference against a rapidly broadening spectrum of therapeutic targets and to translate this to the clinical setting under certain restrictions.


Subject(s)
Cardiovascular Diseases/therapy , Genetic Therapy/methods , Molecular Targeted Therapy/methods , RNA Interference , RNA, Untranslated/therapeutic use , Animals , Cardiovascular Diseases/genetics , Dependovirus/genetics , Dependovirus/immunology , Drug Evaluation, Preclinical , Drug Stability , Forecasting , Genetic Therapy/adverse effects , Genetic Vectors/genetics , Genetic Vectors/immunology , Genetic Vectors/therapeutic use , Genome-Wide Association Study , Humans , MicroRNAs/adverse effects , MicroRNAs/immunology , MicroRNAs/physiology , MicroRNAs/therapeutic use , Molecular Targeted Therapy/adverse effects , RNA Processing, Post-Transcriptional , RNA, Small Interfering/adverse effects , RNA, Small Interfering/immunology , RNA, Small Interfering/pharmacology , RNA, Small Interfering/physiology , RNA, Small Interfering/therapeutic use , RNA, Untranslated/adverse effects , RNA, Untranslated/classification , RNA, Untranslated/immunology , RNA, Untranslated/pharmacology , RNA, Untranslated/physiology , Substrate Specificity , Transcriptome , Translational Research, Biomedical
20.
Nat Rev Genet ; 9(7): 503-7, 2008 07.
Article in English | MEDLINE | ID: mdl-18490927

ABSTRACT

It has been known for decades that some clinically important viruses encode abundant amounts of non-coding RNAs (ncRNAs) during infection. Until recently, the number of viral ncRNAs identified was few and their functions were mostly unknown. Although our understanding is still in its infancy, several recent reports have identified new functions for viral microRNAs and larger ncRNAs. These results so far show that different classes of viral ncRNAs act to autoregulate viral gene expression and evade host antiviral defences such as apoptosis and the immune response.


Subject(s)
RNA, Untranslated/genetics , RNA, Untranslated/immunology , Viruses/genetics , Viruses/immunology , Animals , Gene Expression Regulation, Viral , Humans , RNA Interference , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL