Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 110
Filter
1.
J Clin Invest ; 131(4)2021 02 15.
Article in English | MEDLINE | ID: mdl-33320835

ABSTRACT

Unlike pathogens, which attack the host, commensal bacteria create a state of friendly coexistence. Here, we identified a mechanism of bacterial adaptation to the host niche, where they reside. Asymptomatic carrier strains were shown to inhibit RNA polymerase II (Pol II) in host cells by targeting Ser2 phosphorylation, a step required for productive mRNA elongation. Assisted by a rare, spontaneous loss-of-function mutant from a human carrier, the bacterial NlpD protein was identified as a Pol II inhibitor. After internalization by host cells, NlpD was shown to target constituents of the Pol II phosphorylation complex (RPB1 and PAF1C), attenuating host gene expression. Therapeutic efficacy of a recombinant NlpD protein was demonstrated in a urinary tract infection model, by reduced tissue pathology, accelerated bacterial clearance, and attenuated Pol II-dependent gene expression. The findings suggest an intriguing, evolutionarily conserved mechanism for bacterial modulation of host gene expression, with a remarkable therapeutic potential.


Subject(s)
Escherichia coli Infections , Escherichia coli Proteins , Escherichia coli , Gene Expression Regulation, Bacterial/immunology , Lipoproteins , RNA Polymerase II , Urinary Tract Infections , Animals , Cell Line, Tumor , Escherichia coli/genetics , Escherichia coli/immunology , Escherichia coli Infections/genetics , Escherichia coli Infections/immunology , Escherichia coli Proteins/genetics , Escherichia coli Proteins/immunology , Female , Humans , Lipoproteins/genetics , Lipoproteins/immunology , Mice , RNA Polymerase II/genetics , RNA Polymerase II/immunology , Urinary Tract Infections/genetics , Urinary Tract Infections/immunology
2.
J Immunol ; 204(5): 1173-1187, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31996458

ABSTRACT

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


Subject(s)
Epigenesis, Genetic/immunology , Human Umbilical Vein Endothelial Cells/immunology , Human Umbilical Vein Endothelial Cells/cytology , Humans , Promoter Regions, Genetic/immunology , RNA Polymerase II/genetics , RNA Polymerase II/immunology , Transcription Factor RelA/genetics , Transcription Factor RelA/immunology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/immunology
3.
Proc Natl Acad Sci U S A ; 115(39): E9162-E9171, 2018 09 25.
Article in English | MEDLINE | ID: mdl-30201712

ABSTRACT

Epigenetic memory for signal-dependent transcription has remained elusive. So far, the concept of epigenetic memory has been largely limited to cell-autonomous, preprogrammed processes such as development and metabolism. Here we show that IFNß stimulation creates transcriptional memory in fibroblasts, conferring faster and greater transcription upon restimulation. The memory was inherited through multiple cell divisions and led to improved antiviral protection. Of ∼2,000 IFNß-stimulated genes (ISGs), about half exhibited memory, which we define as memory ISGs. The rest, designated nonmemory ISGs, did not show memory. Surprisingly, mechanistic analysis showed that IFN memory was not due to enhanced IFN signaling or retention of transcription factors on the ISGs. We demonstrated that this memory was attributed to accelerated recruitment of RNA polymerase II and transcription/chromatin factors, which coincided with acquisition of the histone H3.3 and H3K36me3 chromatin marks on memory ISGs. Similar memory was observed in bone marrow macrophages after IFNγ stimulation, suggesting that IFN stimulation modifies the shape of the innate immune response. Together, external signals can establish epigenetic memory in mammalian cells that imparts lasting adaptive performance upon various somatic cells.


Subject(s)
Bone Marrow Cells/immunology , Cell Division/immunology , Epigenesis, Genetic/immunology , Immunity, Innate , Interferon-beta/immunology , Macrophages/immunology , Signal Transduction/immunology , Transcription, Genetic/immunology , Animals , Bone Marrow Cells/cytology , Cell Division/genetics , Chromatin/genetics , Chromatin/immunology , Histones/genetics , Histones/immunology , Interferon-beta/genetics , Macrophages/cytology , Mice , Mice, Mutant Strains , RNA Polymerase II/genetics , RNA Polymerase II/immunology , Signal Transduction/genetics , Transcription Factors/genetics , Transcription Factors/immunology
4.
Proc Natl Acad Sci U S A ; 115(39): E9172-E9181, 2018 09 25.
Article in English | MEDLINE | ID: mdl-30206152

ABSTRACT

Genome-wide investigations of host-pathogen interactions are often limited by analyses of mixed populations of infected and uninfected cells, which lower sensitivity and accuracy. To overcome these obstacles and identify key mechanisms by which Zika virus (ZIKV) manipulates host responses, we developed a system that enables simultaneous characterization of genome-wide transcriptional and epigenetic changes in ZIKV-infected and neighboring uninfected primary human macrophages. We demonstrate that transcriptional responses in ZIKV-infected macrophages differed radically from those in uninfected neighbors and that studying the cell population as a whole produces misleading results. Notably, the uninfected population of macrophages exhibits the most rapid and extensive changes in gene expression, related to type I IFN signaling. In contrast, infected macrophages exhibit a delayed and attenuated transcriptional response distinguished by preferential expression of IFNB1 at late time points. Biochemical and genomic studies of infected macrophages indicate that ZIKV infection causes both a targeted defect in the type I IFN response due to degradation of STAT2 and reduces RNA polymerase II protein levels and DNA occupancy, particularly at genes required for macrophage identity. Simultaneous evaluation of transcriptomic and epigenetic features of infected and uninfected macrophages thereby reveals the coincident evolution of dominant proviral or antiviral mechanisms, respectively, that determine the outcome of ZIKV exposure.


Subject(s)
Immunity, Innate , Macrophages/immunology , Zika Virus Infection/immunology , Zika Virus/immunology , Bystander Effect , Female , Humans , Interferon-beta/genetics , Interferon-beta/immunology , Macrophages/pathology , Male , Proteolysis , RNA Polymerase II/genetics , RNA Polymerase II/immunology , STAT2 Transcription Factor/genetics , STAT2 Transcription Factor/immunology , Zika Virus Infection/pathology
5.
Nucleic Acids Res ; 46(17): 9011-9026, 2018 09 28.
Article in English | MEDLINE | ID: mdl-30137501

ABSTRACT

DExD/H-box helicase 9 (DHX9), or RNA helicase A (RHA), is an abundant multifunctional nuclear protein. Although it was previously reported to act as a cytosolic DNA sensor in plasmacytoid dendritic cells (pDCs), the role and molecular mechanisms of action of DHX9 in cells that are not pDCs during DNA virus infection are not clear. Here, a macrophage-specific knockout and a fibroblast-specific knockdown of DHX9 impaired antiviral innate immunity against DNA viruses, leading to increased virus replication. DHX9 enhanced NF-κB-mediated transactivation in the nucleus, which required its ATPase-dependent helicase (ATPase/helicase) domain, but not the cytosolic DNA-sensing domain. In addition, DNA virus infection did not induce cytoplasmic translocation of nuclear DHX9 in macrophages and fibroblasts. Nuclear DHX9 was associated with a multiprotein complex including both NF-κB p65 and RNA polymerase II (RNAPII) in chromatin containing NF-κB-binding sites. DHX9 was essential for the recruitment of RNAPII rather than NF-κB p65, to the corresponding promoters; this function also required its ATPase/helicase activity. Taken together, our results show a critical role of nuclear DHX9 (as a transcription coactivator) in the stimulation of NF-κB-mediated innate immunity against DNA virus infection, independently of DHX9's DNA-sensing function.


Subject(s)
DEAD-box RNA Helicases/genetics , DNA, Viral/genetics , Host-Pathogen Interactions/genetics , Immunity, Innate , NF-kappa B/genetics , RNA Polymerase II/genetics , Animals , Chlorocebus aethiops , DEAD-box RNA Helicases/deficiency , DEAD-box RNA Helicases/immunology , DNA, Viral/immunology , Dendritic Cells/immunology , Dendritic Cells/virology , Female , Gammaherpesvirinae/genetics , Gammaherpesvirinae/growth & development , Gammaherpesvirinae/immunology , Gene Expression Regulation , HEK293 Cells , HeLa Cells , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/growth & development , Herpesvirus 1, Human/immunology , Host-Pathogen Interactions/immunology , Humans , Macrophages/immunology , Macrophages/virology , Male , Mice , Mice, Transgenic , Mouse Embryonic Stem Cells/immunology , Mouse Embryonic Stem Cells/virology , NF-kappa B/immunology , NIH 3T3 Cells , Primary Cell Culture , RNA Polymerase II/immunology , Signal Transduction , Vero Cells , Virus Replication
6.
Cell ; 172(4): 811-824.e14, 2018 02 08.
Article in English | MEDLINE | ID: mdl-29395325

ABSTRACT

Type I interferon (IFN) is produced when host sensors detect foreign nucleic acids, but how sensors differentiate self from nonself nucleic acids, such as double-stranded RNA (dsRNA), is incompletely understood. Mutations in ADAR1, an adenosine-to-inosine editing enzyme of dsRNA, cause Aicardi-Goutières syndrome, an autoinflammatory disorder associated with spontaneous interferon production and neurologic sequelae. We generated ADAR1 knockout human cells to explore ADAR1 substrates and function. ADAR1 primarily edited Alu elements in RNA polymerase II (pol II)-transcribed mRNAs, but not putative pol III-transcribed Alus. During the IFN response, ADAR1 blocked translational shutdown by inhibiting hyperactivation of PKR, a dsRNA sensor. ADAR1 dsRNA binding and catalytic activities were required to fully prevent endogenous RNA from activating PKR. Remarkably, ADAR1 knockout neuronal progenitor cells exhibited MDA5 (dsRNA sensor)-dependent spontaneous interferon production, PKR activation, and cell death. Thus, human ADAR1 regulates sensing of self versus nonself RNA, allowing pathogen detection while avoiding autoinflammation.


Subject(s)
Adenosine Deaminase/metabolism , Alu Elements , Autoimmune Diseases of the Nervous System/metabolism , Nervous System Malformations/metabolism , Neural Stem Cells/metabolism , Protein Biosynthesis , RNA, Double-Stranded/metabolism , RNA-Binding Proteins/metabolism , Adenosine Deaminase/genetics , Adenosine Deaminase/immunology , Autoimmune Diseases of the Nervous System/genetics , Autoimmune Diseases of the Nervous System/immunology , Cell Death/genetics , Cell Death/immunology , Gene Knockout Techniques , HEK293 Cells , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Interferon-Induced Helicase, IFIH1/genetics , Interferon-Induced Helicase, IFIH1/immunology , Interferon-Induced Helicase, IFIH1/metabolism , Nervous System Malformations/genetics , Nervous System Malformations/immunology , Neural Stem Cells/cytology , Neural Stem Cells/immunology , Neural Stem Cells/pathology , RNA Polymerase II/genetics , RNA Polymerase II/immunology , RNA Polymerase II/metabolism , RNA, Double-Stranded/genetics , RNA, Double-Stranded/immunology , RNA-Binding Proteins/genetics , RNA-Binding Proteins/immunology , eIF-2 Kinase/genetics , eIF-2 Kinase/immunology , eIF-2 Kinase/metabolism
7.
Mol Immunol ; 91: 8-16, 2017 11.
Article in English | MEDLINE | ID: mdl-28858629

ABSTRACT

B cell activation is dependent on a large increase in transcriptional output followed by focused expression on secreted immunoglobulin as the cell transitions to an antibody producing plasma cell. The rapid transcriptional induction is facilitated by the release of poised RNA pol II into productive elongation through assembly of the super elongation complex (SEC). We report that a SEC component, the Eleven -nineteen Lysine-rich leukemia (ELL) family member 3 (ELL3) is dynamically up-regulated in mature and activated human B cells followed by suppression as B cells transition to plasma cells in part mediated by the transcription repressor PRDM1. Burkitt's lymphoma and a sub-set of Diffuse Large B cell lymphoma cell lines abundantly express ELL3. Depletion of ELL3 in the germinal center derived lymphomas results in severe disruption of DNA replication and cell division along with increased DNA damage and cell death. This restricted utilization and survival dependence reveal a key step in B cell activation and indicate a potential therapeutic target against B cell lymphoma's with a germinal center origin.


Subject(s)
B-Lymphocytes/immunology , Cell Division/immunology , Gene Expression Regulation/immunology , Transcriptional Elongation Factors/immunology , Cell Division/genetics , Cell Survival/genetics , Cell Survival/immunology , DNA Replication/genetics , DNA Replication/immunology , Humans , Jurkat Cells , RNA Polymerase II/genetics , RNA Polymerase II/immunology , Transcriptional Elongation Factors/genetics
8.
J Immunol ; 199(9): 3249-3260, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28931603

ABSTRACT

Members of the IFN-inducible PYHIN protein family, such as absent in melanoma-2 and IFN-γ-inducible protein (IFI)16, bind dsDNA and form caspase-1-activating inflammasomes that are important in immunity to cytosolic bacteria, DNA viruses, or HIV. IFI16 has also been shown to regulate transcription of type I IFNs during HSV infection. The role of other members of the PYHIN protein family in the regulation of immune responses is much less clear. In this study, we identified an immune-regulatory function for a member of the murine PYHIN protein family, p205 (also called Ifi205). Examination of immune responses induced by dsDNA and other microbial ligands in bone marrow-derived macrophages lacking p205 revealed that inflammasome activation by dsDNA, as well as ligands that engage the NLRP3 inflammasome, was severely compromised in these cells. Further analysis revealed that p205-knockdown cells showed reduced expression of apoptosis-associated speck-like molecule containing CARD domain (Asc) at the protein and RNA levels. p205 knockdown resulted in reduced binding of actively transcribing RNA polymerase II to the endogenous Asc gene, resulting in decreased transcription and processing of Asc pre-mRNA. Deletion of p205 in B16 melanoma cells using CRISPR/Cas9 showed a similar loss of Asc expression. Ectopic expression of p205 induced expression of an Asc promoter-luciferase reporter gene. Together, these findings suggest that p205 controls expression of Asc mRNA to regulate inflammasome responses. These findings expand on our understanding of immune-regulatory roles for the PYHIN protein family.


Subject(s)
Apoptosis Regulatory Proteins/immunology , Gene Expression Regulation/immunology , Inflammasomes/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Animals , Apoptosis Regulatory Proteins/genetics , CARD Signaling Adaptor Proteins , Cell Line, Tumor , Gene Expression Regulation/genetics , Gene Knockdown Techniques , Inflammasomes/genetics , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , RNA Polymerase II/genetics , RNA Polymerase II/immunology
9.
Sci Rep ; 7(1): 7594, 2017 08 08.
Article in English | MEDLINE | ID: mdl-28790320

ABSTRACT

Activation-induced cytidine deaminase (AID) triggers antibody diversification in B cells by catalysing deamination and subsequently mutating immunoglobulin (Ig) genes. Association of AID with RNA Pol II and occurrence of epigenetic changes during Ig gene diversification suggest participation of AID in epigenetic regulation. AID is mutated in hyper-IgM type 2 (HIGM2) syndrome. Here, we investigated the potential role of AID in the acquisition of epigenetic changes. We discovered that AID binding to the IgH locus promotes an increase in H4K20me3. In 293F cells, we demonstrate interaction between co-transfected AID and the three SUV4-20 histone H4K20 methyltransferases, and that SUV4-20H1.2, bound to the IgH switch (S) mu site, is replaced by SUV4-20H2 upon AID binding. Analysis of HIGM2 mutants shows that the AID truncated form W68X is impaired to interact with SUV4-20H1.2 and SUV4-20H2 and is unable to bind and target H4K20me3 to the Smu site. We finally show in mouse primary B cells undergoing class-switch recombination (CSR) that AID deficiency associates with decreased H4K20me3 levels at the Smu site. Our results provide a novel link between SUV4-20 enzymes and CSR and offer a new aspect of the interplay between AID and histone modifications in setting the epigenetic status of CSR sites.


Subject(s)
Cytidine Deaminase/genetics , Epigenesis, Genetic/immunology , Histone-Lysine N-Methyltransferase/genetics , Histones/genetics , Hyper-IgM Immunodeficiency Syndrome/genetics , Immunoglobulin Class Switching/genetics , Animals , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Binding Sites , Cell Line, Tumor , Cytidine Deaminase/immunology , Gene Expression Regulation , HEK293 Cells , HeLa Cells , Histone-Lysine N-Methyltransferase/immunology , Histones/immunology , Humans , Hyper-IgM Immunodeficiency Syndrome/immunology , Hyper-IgM Immunodeficiency Syndrome/pathology , Immunoglobulin G/genetics , Lipopolysaccharides/pharmacology , Lymphocyte Activation/drug effects , Mice , Mice, Inbred C57BL , Models, Biological , Mutation , Protein Binding , RNA Polymerase II/genetics , RNA Polymerase II/immunology , Signal Transduction
10.
Exp Mol Med ; 48(11): e271, 2016 11 18.
Article in English | MEDLINE | ID: mdl-27857068

ABSTRACT

The C-terminal domain of RNA polymerase II is an unusual series of repeated residues appended to the C-terminus of the largest subunit and serves as a flexible binding scaffold for numerous nuclear factors. The binding of these factors is determined by the phosphorylation patterns on the repeats in the domain. In this study, we generated a synthetic antibody library by replacing the third heavy chain complementarity-determining region of an anti-HER2 (human epidermal growth factor receptor 2) antibody (trastuzumab) with artificial sequences of 7-18 amino-acid residues. From this library, antibodies were selected that were specific to serine phosphopeptides that represent typical phosphorylation patterns on the functional unit (YSPTSPS)2 of the RNA polymerase II C-terminal domain (CTD). Antibody clones pCTD-1stS2 and pCTD-2ndS2 showed specificity for peptides with phosphoserine at the second residues of the first or second heptamer repeat, respectively. Additional clones specifically reacted to peptides with phosphoserine at the fifth serine of the first repeat (pCTD-1stS5), the seventh residue of the first repeat and fifth residue of the second repeat (pCTD-S7S5) or the seventh residue of either the first or second repeat (pCTD-S7). All of these antibody clones successfully reacted to RNA polymerase II in immunoblot analysis. Interestingly, pCTD-2ndS2 precipitated predominately RNA polymerase II from the exonic regions of genes in genome-wide chromatin immunoprecipitation sequencing analysis, which suggests that the phosphoserine at the second residue of the second repeat of the functional unit (YSPTSPS)2 is a mediator of exon definition.


Subject(s)
Antibodies/metabolism , Chromatin Immunoprecipitation/methods , Exons , RNA Polymerase II/metabolism , Antibodies/immunology , HEK293 Cells , HeLa Cells , Humans , Phosphorylation , Protein Binding , RNA Polymerase II/immunology
12.
PLoS One ; 11(6): e0157454, 2016.
Article in English | MEDLINE | ID: mdl-27310721

ABSTRACT

Neutrophils are essential innate immune cells whose responses are crucial in the clearance of invading pathogens. Neutrophils can respond to infection by releasing neutrophil extracellular traps (NETs). NETs are formed of chromatin and specific granular proteins and are released after execution of a poorly characterized cell death pathway. Here, we show that NET formation induced by PMA or Candida albicans is independent of RNA polymerase II and III-mediated transcription as well as of protein synthesis. Thus, neutrophils contain all the factors required for NET formation when they emerge from the bone marrow as differentiated cells.


Subject(s)
Extracellular Traps/immunology , Gene Expression Regulation/immunology , Neutrophils/immunology , Animals , Candida albicans/immunology , Chemokine CCL3/genetics , Chemokine CCL3/immunology , Cycloheximide/pharmacology , Dactinomycin/pharmacology , Extracellular Traps/chemistry , Extracellular Traps/drug effects , Extracellular Traps/genetics , Flavonoids/pharmacology , Gene Expression Regulation/drug effects , Humans , Interleukin-8/genetics , Interleukin-8/immunology , Lipopolysaccharides/pharmacology , Male , Mice , Neutrophils/drug effects , Neutrophils/metabolism , Neutrophils/microbiology , Piperidines/pharmacology , Primary Cell Culture , RNA Polymerase II/genetics , RNA Polymerase II/immunology , RNA Polymerase III/genetics , RNA Polymerase III/immunology , Salmonella typhimurium/immunology , Tetradecanoylphorbol Acetate/pharmacology
13.
Virus Res ; 220: 193-202, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27117321

ABSTRACT

Infectious hematopoietic necrosis virus is a highly contagious disease of juvenile salmonid species. From the IHNV HLJ-09 isolated in China, two recombinant viruses were generated by reverse genetics using the RNA polymerase II transcription system. The recombinant viruses were confirmed by RT-PCR, indirect immunofluorescence assay and electron microscopy. They were referred to as rIHNV HLJ-09 and rIHNV-EGFP. rIHNV HLJ-09 and rIHNV-EGFP could stably replicate in EPC cell lines and had the same cellular tropism as wtIHNV HLJ-09. But the titer of rIHNV-EGFP was significantly lower than rIHNV HLJ-09 and wtIHNV HLJ-09. rIHNV-EGFP strain could express EGFP stably at least in 20 passages, and the fluorescence could be observed clearly. To assess the virulence and pathogenicity of the recombinant viruses in vivo, juvenile rainbow trout were challenged by intraperitoneal injection with 20µl of rIHNV HLJ-09, rIHNV-EGFP or wtIHNV HLJ-09 (1×10(6)pfuml(-1)). Fish challenged with rIHNV HLJ-09 and wtIHNV HLJ-09 exhibited clinical signs typical of IHN disease and both produced 90% cumulative percent mortality, whlie rIHNV-EGFP produced only 5%. Pathological sectioning results showed that the tissues (liver, kidney, heart muscle, back muscle) of the fish infected with rIHNV HLJ-09 exhibited pathological changes, with the exception of cerebral neurons and the cheek. However, no lesions of liver, kidney, heart, muscle, brain in rainbow trout of rIHNV-EGFP or the control group were observed. Indirect ELISA results showed that a high level of serum antibody was detected in the experimental fish challenged with rIHNV HLJ-09, just as the same as wtIHNV HLJ-09, while a lower titer was detecred in the fish infected with rIHNV-EGFP. This indicated that the recombinant viruses could induce humoral immune response in the experimental fish. The recombinant viruses had unique genetic tags and could be used for genetic engineering, laying new ground for further investigation of IHNV pathopoiesis molecular mechanism, host tropism and the development of novel vaccines against IHN.


Subject(s)
Antibodies, Viral/biosynthesis , Immunity, Humoral , Infectious hematopoietic necrosis virus/pathogenicity , Mutation , Oncorhynchus mykiss/virology , Rhabdoviridae Infections/veterinary , Animals , Fish Diseases/immunology , Fish Diseases/prevention & control , Fish Diseases/virology , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Heart/virology , Infectious hematopoietic necrosis virus/genetics , Infectious hematopoietic necrosis virus/immunology , Kidney/pathology , Kidney/virology , Liver/pathology , Liver/virology , Muscles/pathology , Muscles/virology , Plasmids/chemistry , Plasmids/immunology , RNA Polymerase II/genetics , RNA Polymerase II/immunology , Reverse Genetics , Rhabdoviridae Infections/immunology , Rhabdoviridae Infections/prevention & control , Rhabdoviridae Infections/virology , Viral Tropism , Viral Vaccines/biosynthesis , Viral Vaccines/genetics , Viral Vaccines/immunology , Virulence , Virus Replication
14.
J Immunol ; 195(3): 1312-9, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26116512

ABSTRACT

Primary lymphoma of the CNS (PCNSL) is a diffuse large B cell lymphoma confined to the CNS. To elucidate its peculiar organ tropism, we generated recombinant Abs (recAbs) identical to the BCR of 23 PCNSLs from immunocompetent patients. Although none of the recAbs showed self-reactivity upon testing with common autoantigens, they recognized 1547 proteins present on a large-scale protein microarray, indicating polyreactivity. Interestingly, proteins (GRINL1A, centaurin-α, BAIAP2) recognized by the recAbs are physiologically expressed by CNS neurons. Furthermore, 87% (20/23) of the recAbs, including all Abs derived from IGHV4-34 using PCNSL, recognized galectin-3, which was upregulated on microglia/macrophages, astrocytes, and cerebral endothelial cells upon CNS invasion by PCNSL. Thus, PCNSL Ig may recognize CNS proteins as self-Ags. Their interaction may contribute to BCR signaling with sustained NF-κB activation and, ultimately, may foster tumor cell proliferation and survival. These data may also explain, at least in part, the affinity of PCNSL cells for the CNS.


Subject(s)
Antibodies, Neoplasm/immunology , Central Nervous System Neoplasms/immunology , Lymphoma, B-Cell/immunology , Receptors, Antigen, B-Cell/immunology , Adaptor Proteins, Signal Transducing/immunology , Adult , Aged , Aged, 80 and over , Astrocytes/immunology , Base Sequence , Blood Proteins , Carcinoma, Large Cell/immunology , Cell Proliferation , Endothelial Cells/immunology , Enzyme Activation , Female , Galectin 3/immunology , Galectins , Gene Expression Profiling , Humans , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/immunology , Immunoglobulin Light Chains/genetics , Immunoglobulin Light Chains/immunology , Immunoglobulins/genetics , Immunoglobulins/immunology , Macrophages/immunology , Male , Microglia/immunology , Middle Aged , NF-kappa B/metabolism , Nerve Tissue Proteins/immunology , RNA Polymerase II/immunology , Sequence Analysis, DNA
15.
DNA Repair (Amst) ; 30: 46-52, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25869824

ABSTRACT

Activation-induced deaminase (AID) is a DNA cytosine deaminase that diversifies immunoglobulin genes in B cells. Recent work has shown that RNA polymerase II (Pol II) accumulation correlates with AID recruitment. However, a direct link between Pol II and AID abundance has not been tested. We used the DT40 B-cell line to manipulate levels of Pol II by decreasing topoisomerase I (Top1), which relaxes DNA supercoiling in front of the transcription complex. Top1 was decreased by stable transfection of a short hairpin RNA against Top1, which produced an accumulation of Pol II in transcribed genes, compared to cells transfected with sh-control RNA. The increased Pol II density enhanced AID recruitment to variable genes in the λ light chain locus, and resulted in higher levels of somatic hypermutation and gene conversion. It has been proposed by another lab that AID itself might directly suppress Top1 to increase somatic hypermutation. However, we found that in both AID(+/+) and AID(-/-) B cells from DT40 and mice, Top1 protein levels were identical, indicating that the presence or absence of AID did not decrease Top1 expression. Rather, our results suggest that the mechanism for increased diversity when Top1 is reduced is that Pol II accumulates and recruits AID to variable genes.


Subject(s)
B-Lymphocytes/immunology , Cytidine Deaminase/metabolism , DNA Topoisomerases, Type I/metabolism , Genes, Immunoglobulin , Immunoglobulin Variable Region/genetics , RNA Polymerase II/metabolism , Animals , Cell Line , Chickens/genetics , Chickens/immunology , Cytidine Deaminase/genetics , Cytidine Deaminase/immunology , DNA/metabolism , DNA Topoisomerases, Type I/genetics , DNA Topoisomerases, Type I/immunology , Gene Knockout Techniques , Mice , RNA Polymerase II/genetics , RNA Polymerase II/immunology , Somatic Hypermutation, Immunoglobulin
16.
Clin J Am Soc Nephrol ; 10(3): 372-81, 2015 Mar 06.
Article in English | MEDLINE | ID: mdl-25542908

ABSTRACT

BACKGROUND AND OBJECTIVES: IgA plays a key role in IgA nephropathy (IgAN) by forming immune complexes and depositing in the glomeruli, leading to an inflammatory response. However, the antigenic targets and functional characterization of IgA have been incompletely defined in this disease. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS: This study was performed in sera from patients who were studied as part of a prospective, observational study of IgAN. These patients (n=22) all had biopsy-proven IgAN within 3 years of study initiation, complete clinical data, annual urinary inulin clearance for GFRs, and at least 5 years of follow-up. Progression was defined as loss of >5 ml/min per 1.73 m(2) per year of inulin clearance measured over at least 5 years. A protein microarray was used for detection of IgAN-specific IgA autoantibodies in blood across approximately 9000 human antigens to specifically identify the most immunogenic protein targets that drive IgA antibodies in IgAN (n=22), healthy controls (n=10), and non-IgAN glomerular diseases (n=17). Results were validated by ELISA assays in sera and by immunohistochemistry in IgAN kidney biopsies. IgA-specific antibodies were correlated with clinical and histologic variables to assess their effect on disease progression and prognosis. RESULTS: Fifty-four proteins mounted highly significant IgA antibody responses in patients with IgAN with a false discovery rate (q value) of ≤10%; 325 antibodies (P≤0.05) were increased overall. Antitissue transglutaminase IgA was significantly elevated in IgAN (P<0.001, q value of 0%). IgA antibodies to DDX4 (r=-0.55, P=0.01) and ZADH2 (r=-0.48, P=0.02) were significantly correlated with the decline of renal function. Specific IgA autoantibodies are elevated in IgAN compared with normal participants and those with other glomerular diseases. CONCLUSIONS: In this preliminary study, IgA autoantibodies target novel proteins, highly expressed in the kidney glomerulus and tubules. These IgA autoantibodies may play important roles in the pathogenesis of IgAN.


Subject(s)
Autoantibodies/blood , Epitope Mapping , Epitopes , Glomerulonephritis, IGA/immunology , Immunoglobulin A/blood , Adult , Antigens, Surface/immunology , Area Under Curve , Blood Proteins/immunology , Case-Control Studies , DEAD-box RNA Helicases/immunology , DNA-Binding Proteins/immunology , Disease Progression , Female , GTP-Binding Proteins/immunology , Glomerular Filtration Rate , Glomerulonephritis, IGA/pathology , Glomerulonephritis, IGA/physiopathology , Homeodomain Proteins/immunology , Humans , Male , Membrane Proteins/immunology , Middle Aged , Muscle Proteins/immunology , Nerve Tissue Proteins/immunology , Prospective Studies , Protein Array Analysis , Protein Glutamine gamma Glutamyltransferase 2 , RNA Polymerase II/immunology , ROC Curve , TEA Domain Transcription Factors , Transcription Factors/immunology , Transglutaminases/immunology , Ubiquitin-Protein Ligases/immunology
17.
Proc Natl Acad Sci U S A ; 111(35): 12829-34, 2014 Sep 02.
Article in English | MEDLINE | ID: mdl-25136117

ABSTRACT

Epigenetic modifications, such as posttranslational modifications of histones, play an important role in gene expression and regulation. These modifications are in part mediated by the Trithorax group (TrxG) complex and the Polycomb group (PcG) complex, which activate and repress transcription, respectively. We herein investigate the role of Menin, a component of the TrxG complex in T helper (Th) cell differentiation and show a critical role for Menin in differentiation and maintenance of Th17 cells. Menin(-/-) T cells do not efficiently differentiate into Th17 cells, leaving Th1 and Th2 cell differentiation intact in in vitro cultures. Menin deficiency resulted in the attenuation of Th17-induced airway inflammation. In differentiating Th17 cells, Menin directly bound to the Il17a gene locus and was required for the deposition of permissive histone modifications and recruitment of the RNA polymerase II transcriptional complex. Interestingly, although Menin bound to the Rorc locus, Menin was dispensable for the induction of Rorc expression and permissive histone modifications in differentiating Th17 cells. In contrast, Menin was required to maintain expression of Rorc in differentiated Th17 cells, indicating that Menin is essential to stabilize expression of the Rorc gene. Thus, Menin orchestrates Th17 cell differentiation and function by regulating both the induction and maintenance of target gene expression.


Subject(s)
Asthma/immunology , Epigenesis, Genetic/immunology , Interleukin-17/immunology , Proto-Oncogene Proteins/immunology , Th17 Cells/immunology , Animals , Cell Differentiation/genetics , Cell Differentiation/immunology , Chromatin/immunology , Chromatin/metabolism , Epigenesis, Genetic/genetics , Gene Expression Regulation/immunology , Histone-Lysine N-Methyltransferase/immunology , Histone-Lysine N-Methyltransferase/metabolism , Interleukin-17/genetics , Interleukin-17/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Myeloid-Lymphoid Leukemia Protein/immunology , Myeloid-Lymphoid Leukemia Protein/metabolism , Neutrophils/drug effects , Neutrophils/immunology , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Ovalbumin/immunology , Ovalbumin/pharmacology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA Polymerase II/immunology , RNA Polymerase II/metabolism , Th17 Cells/metabolism
18.
J Immunol ; 193(1): 391-9, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24890719

ABSTRACT

The T cell-surface glycoprotein CD6 is a modulator of cellular responses and has been implicated in several autoimmune diseases such as multiple sclerosis, rheumatoid arthritis, and psoriasis. During Ag presentation, CD6 is targeted to the immunological synapse in a ligand binding-dependent manner, in which CD6 domain 3 directly contacts CD166, expressed on the APC. T cell activation results in the induction of CD6Δd3, an alternatively spliced isoform that lacks the ligand-binding domain and thus no longer localizes at the immunological synapse. In this study, we investigated the molecular mechanisms regulating the expression of CD6Δd3 upon human primary T cell activation. Using chromatin immunoprecipitation, we observed an increase in RNA polymerase II occupancy along the CD6 gene and augmented CD6 transcription. We showed that activation leads to transcription-related chromatin modifications, revealed by higher CD6 acetylation levels. Modulation of chromatin conformation using a histone deacetylase inhibitor that increases transcription rate causes an increase of exon 5 skipping. We further showed that the splicing factor SRSF1 binds to a regulatory element in CD6 intron 4, activating exon 5 splicing and promoting exon 5 inclusion. Concomitant with T cell activation-induced exon 5 skipping, we observed a downregulation of SRSF1. Using RNA immunoprecipitation, we showed that in activated T cells, SRSF1 recruitment to the CD6 transcript is impaired by increased chromatin acetylation levels. We propose that upon T cell activation, SRSF1 becomes limiting, and its function in CD6 exon 5 splicing is countered by an increase in CD6 transcription, dependent on chromatin acetylation.


Subject(s)
Alternative Splicing/physiology , Antigens, CD/immunology , Antigens, Differentiation, T-Lymphocyte/immunology , Lymphocyte Activation/physiology , Nuclear Proteins/immunology , RNA-Binding Proteins/immunology , T-Lymphocytes/immunology , Transcription, Genetic/immunology , Acetylation , Antigen-Presenting Cells/cytology , Antigen-Presenting Cells/immunology , Antigens, CD/genetics , Antigens, Differentiation, T-Lymphocyte/genetics , Cell Adhesion Molecules, Neuronal/genetics , Cell Adhesion Molecules, Neuronal/immunology , Chromatin/genetics , Chromatin/immunology , Female , Fetal Proteins/genetics , Fetal Proteins/immunology , Humans , Introns/immunology , Male , Nuclear Proteins/genetics , RNA Polymerase II/genetics , RNA Polymerase II/immunology , RNA-Binding Proteins/genetics , Serine-Arginine Splicing Factors , T-Lymphocytes/cytology , Transcription, Genetic/genetics
19.
J Immunol ; 192(10): 4821-32, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24711619

ABSTRACT

The four Toll/IL-1R domain-containing adaptor proteins MyD88, MAL, TRIF, and TRAM are well established as essential mediators of TLR signaling and gene induction following microbial detection. In contrast, the function of the fifth, most evolutionarily conserved Toll/IL-1R adaptor, sterile α and HEAT/Armadillo motif-containing protein (SARM), has remained more elusive. Recent studies of Sarm(-/-) mice have highlighted a role for SARM in stress-induced neuronal cell death and immune responses in the CNS. However, whether SARM has a role in immune responses in peripheral myeloid immune cells is less clear. Thus, we characterized TLR-induced cytokine responses in SARM-deficient murine macrophages and discovered a requirement for SARM in CCL5 production, whereas gene induction of TNF, IL-1ß, CCL2, and CXCL10 were SARM-independent. SARM was not required for TLR-induced activation of MAPKs or of transcription factors implicated in CCL5 induction, namely NF-κB and IFN regulatory factors, nor for Ccl5 mRNA stability or splicing. However, SARM was critical for the recruitment of transcription factors and of RNA polymerase II to the Ccl5 promoter. Strikingly, the requirement of SARM for CCL5 induction was not restricted to TLR pathways, as it was also apparent in cytosolic RNA and DNA responses. Thus, this study identifies a new role for SARM in CCL5 expression in macrophages.


Subject(s)
Armadillo Domain Proteins/immunology , Chemokine CCL5/immunology , Cytoskeletal Proteins/immunology , Interferon Regulatory Factors/immunology , Macrophages, Peritoneal/immunology , NF-kappa B/immunology , Promoter Regions, Genetic/immunology , RNA Polymerase II/immunology , Animals , Armadillo Domain Proteins/genetics , Armadillo Domain Proteins/metabolism , Chemokine CCL2/genetics , Chemokine CCL2/immunology , Chemokine CCL2/metabolism , Chemokine CCL5/biosynthesis , Chemokine CCL5/genetics , Chemokine CXCL10/genetics , Chemokine CXCL10/immunology , Chemokine CXCL10/metabolism , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Gene Expression Regulation/physiology , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Macrophages, Peritoneal/cytology , Macrophages, Peritoneal/metabolism , Mice , Mice, Knockout , NF-kappa B/genetics , NF-kappa B/metabolism , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
20.
PLoS Pathog ; 10(1): e1003854, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24391504

ABSTRACT

Histone modifications are important regulators of gene expression in all eukaryotes. In Plasmodium falciparum, these epigenetic marks regulate expression of genes involved in several aspects of host-parasite interactions, including antigenic variation. While the identities and genomic positions of many histone modifications have now been cataloged, how they are targeted to defined genomic regions remains poorly understood. For example, how variant antigen encoding loci (var) are targeted for deposition of unique histone marks is a mystery that continues to perplex the field. Here we describe the recruitment of an ortholog of the histone modifier SET2 to var genes through direct interactions with the C-terminal domain (CTD) of RNA polymerase II. In higher eukaryotes, SET2 is a histone methyltransferase recruited by RNA pol II during mRNA transcription; however, the ortholog in P. falciparum (PfSET2) has an atypical architecture and its role in regulating transcription is unknown. Here we show that PfSET2 binds to the unphosphorylated form of the CTD, a property inconsistent with its recruitment during mRNA synthesis. Further, we show that H3K36me3, the epigenetic mark deposited by PfSET2, is enriched at both active and silent var gene loci, providing additional evidence that its recruitment is not associated with mRNA production. Over-expression of a dominant negative form of PfSET2 designed to disrupt binding to RNA pol II induced rapid var gene expression switching, confirming both the importance of PfSET2 in var gene regulation and a role for RNA pol II in its recruitment. RNA pol II is known to transcribe non-coding RNAs from both active and silent var genes, providing a possible mechanism by which it could recruit PfSET2 to var loci. This work unifies previous reports of histone modifications, the production of ncRNAs, and the promoter activity of var introns into a mechanism that contributes to antigenic variation by malaria parasites.


Subject(s)
Antigenic Variation/physiology , Antigens, Protozoan/immunology , Histone-Lysine N-Methyltransferase/immunology , Plasmodium falciparum/immunology , Protozoan Proteins/immunology , RNA Polymerase II/immunology , Antigens, Protozoan/genetics , Epigenesis, Genetic/immunology , Histone Methyltransferases , Histone-Lysine N-Methyltransferase/genetics , Humans , Plasmodium falciparum/genetics , Protozoan Proteins/genetics , RNA Polymerase II/genetics , RNA, Messenger/genetics , RNA, Messenger/immunology , RNA, Protozoan/genetics , RNA, Protozoan/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...