Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Int J Biol Macromol ; 229: 515-528, 2023 Feb 28.
Article in English | MEDLINE | ID: mdl-36584781

ABSTRACT

Given the paucity of antiviral treatments for monkeypox disease, caused by the Monkeypox virus (MPXV), there is a pressing need for the development/identification of new drugs to treat the infection. MPXV possesses a linear dsDNA genome that is replicated by a DNA replication complex of which DNA polymerase (DPol) forms an important component. Owing to the importance of DPol in the viral life cycle, identifying/designing small molecules abolishing its function could yield new antivirals. In this study, we first used the AlphaFold artificial intelligence program to model the 3D structure of the MPXV DPol; like the fold of DPol from other organisms, the MPXV DPol structure has the characteristic exonuclease, thumb, palm, and fingers sub-domains arrangement. Subsequently, we have identified several inhibitors through virtual screening of ZINC and antiviral libraries. Molecules with phenyl scaffold along with alanine-based and tetrazole-based molecules showed the best docking score of -8 to -10 kcal/mol. These molecules bind in the palm and fingers sub-domains interface region, which partially overlaps with the DNA binding path. The delineation of DPol/inhibitor interactions showed that majorly active site residues ASP549, ASP753, TYR550, ASN551, SER552, and ASN665 interact with the inhibitors. These compounds exhibit good Absorption, Distribution, Metabolism and Excretion properties.


Subject(s)
Monkeypox virus , Mpox (monkeypox) , Humans , Monkeypox virus/genetics , Artificial Intelligence , Mpox (monkeypox)/diagnosis , DNA-Directed DNA Polymerase , Antiviral Agents/pharmacology
2.
J Immunol ; 208(4): 881-897, 2022 02 15.
Article in English | MEDLINE | ID: mdl-35101891

ABSTRACT

Diet plays an important role in lifestyle disorders associated with the disturbed immune system. During the study of methionine- and choline-deficient diet-induced nonalcoholic fatty liver disease, we observed a specific decrease in the plasmacytoid dendritic cell (pDC) fraction from murine spleens. While delineating the role for individual components, we identified that l-methionine supplementation correlates with representation of the pDC fraction. S-adenosylmethionine (SAM) is a key methyl donor, and we demonstrate that supplementation of methionine-deficient medium with SAM but not homocysteine reverses the defect in pDC development. l-Methionine has been implicated in maintenance of methylation status in the cell. Based on our observed effect of SAM and zebularine on DC subset development, we sought to clarify the role of DNA methylation in pDC biology. Whole-genome bisulfite sequencing analysis from the splenic DC subsets identified that pDCs display differentially hypermethylated regions in comparison with classical DC (cDC) subsets, whereas cDC1 and cDC2 exhibited comparable methylated regions, serving as a control in our study. We validated differentially methylated regions in the sorted pDC, CD8α+ cDC1, and CD4+ cDC2 subsets from spleens as well as FL-BMDC cultures. Upon analysis of genes linked with differentially methylated regions, we identified that differential DNA methylation is associated with the MAPK pathway such that its inhibition guides DC development toward the pDC subtype. Overall, our study identifies an important role for methionine in pDC biology.


Subject(s)
Choline/metabolism , DNA Methylation , Dendritic Cells/immunology , Dendritic Cells/metabolism , Diet , Methionine/metabolism , Animals , Biomarkers , Cell Differentiation/genetics , Cell Differentiation/immunology , Computational Biology/methods , Gene Expression Profiling , Gene Expression Regulation , Homeostasis , Immunity, Innate , Immunophenotyping , MAP Kinase Signaling System , Methionine/deficiency , Mice , Protein Interaction Mapping , Transcriptome
3.
Cell Immunol ; 371: 104468, 2022 01.
Article in English | MEDLINE | ID: mdl-34968772

ABSTRACT

Dendritic cells (DCs) play central role in innate as well as adaptive immune responses regulated by diverse DC subtypes that vary in terms of surface markers, transcriptional profile and functional responses. Generation of DC diversity from progenitor stage is tightly regulated by complex molecular inter-play between transcription factors. We earlier demonstrated that Batf3 and Id2 expression have a synergistic effect on the Irf8 directed classical cDC1 development. In present study, Bi-molecular fluorescence complementation assay suggested that IRF8 interacts with BATF3, and ID2 may aid cDC1 development independently. Genome wide recruitment analysis of IRF8 and BATF3 from different DC subtypes led to identification of the overlapping regions of occupancy by these two transcription factors. Further analysis of overlapping peaks of IRF8 and BATF3 occupancy in promoter region within the cDC1 subtype specific transcriptional pattern identified a metabolically important Pfkfb3 gene. Among various immune cell types; splenic cDC1 subtype displayed enhanced expression of Pfkfb3. Analysis of Irf8-/-, Irf8R294C and Batf3DCKO DC confirmed direct regulation of Pfkfb3 enhanced expression specifically in cDC1 subtype. Further we show that inhibition of PFKFB3 enzymatic activity by a chemical agent PFK15 led to reduction in cDC1 subtype in both in vitro FLDC cultures as well as in vivo mouse spleens. Together, our study identified the direct regulation of cDC1 specific enhanced expression of Pfkfb3 in glycolysis and cDC1 biology.


Subject(s)
Dendritic Cells/immunology , Interferon Regulatory Factors/metabolism , Phosphofructokinase-2/biosynthesis , Repressor Proteins/metabolism , Animals , Cell Line , Female , Gene Expression Regulation/genetics , Glycolysis/genetics , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Phosphofructokinase-2/antagonists & inhibitors , Phosphofructokinase-2/genetics , Promoter Regions, Genetic/genetics , Pyridines/pharmacology , Quinolines/pharmacology
4.
Front Immunol ; 12: 758190, 2021.
Article in English | MEDLINE | ID: mdl-34867997

ABSTRACT

Plasmacytoid dendritic cells (pDCs) are the key producers of type I interferons (IFNs), thus playing a central role in initiating antiviral immune response. Besides robust type I IFN production, pDCs also act as antigen presenting cells post immunogenic stimulation. Transcription factor Irf8 is indispensable for the development of both pDC and cDC1 subset. However, the mechanism underlying the differential regulation by IRF8 in cDC1- and pDC-specific genomic architecture of developmental pathways still remains to be fully elucidated. Previous studies indicated that the Irf8R294C mutation specifically abrogates development of cDC1 without affecting that of pDC. In the present study using RNA-seq based approach, we have found that though the point mutation Irf8R294C did not affect pDC development, it led to defective type I IFN production, thus resulting in inefficient antiviral response. This observation unraveled the distinctive roles of IRF8 in these two subpopulations-regulating the development of cDC1 whereas modulating the functionality of pDCs without affecting development. We have reported here that Irf8R294C mutation also caused defect in production of ISGs as well as defective upregulation of costimulatory molecules in pDCs in response to NDV infection (or CpG stimulation). Through in vivo studies, we demonstrated that abrogation of type I IFN production was concomitant with reduced upregulation of costimulatory molecules in pDCs and increased NDV burden in IRF8R294C mice in comparison with wild type, indicating inefficient viral clearance. Further, we have also shown that Irf8R294C mutation abolished the activation of type I IFN promoter by IRF8, justifying the low level of type I IFN production. Taken together, our study signifies that the single point mutation in Irf8, Irf8R294C severely compromised type I IFN-mediated immune response by murine pDCs, thereby causing impairment in antiviral immunity.


Subject(s)
Dendritic Cells/immunology , Interferon Regulatory Factors/genetics , Interferon Type I/immunology , Mutation, Missense , Newcastle Disease/immunology , Point Mutation , Animals , Bone Neoplasms/pathology , Cell Line, Tumor , CpG Islands/immunology , Dendritic Cells/metabolism , Female , Gene Expression Regulation , HEK293 Cells , Humans , Immunity, Innate , Interferon Regulatory Factors/immunology , Interferon Type I/biosynthesis , Male , Mice , Mice, Inbred C57BL , Newcastle disease virus , Osteosarcoma/pathology , Transcriptome
5.
Mol Pharm ; 18(10): 3832-3842, 2021 10 04.
Article in English | MEDLINE | ID: mdl-34499836

ABSTRACT

Use of tumor-associated antigens for cancer immunotherapy is limited due to their poor in vivo stability and low cellular uptake. Delivery of antigenic peptides using synthetic polymer-based nanostructures has been actively pursued but with limited success. Peptide-based nanostructures hold much promise as delivery vehicles due to their easy design and synthesis and inherent biocompatibility. Here, we report self-assembly of a dipeptide containing a non-natural amino acid, α,ß-dehydrophenylalanine (ΔF), into nanotubes, which efficiently entrapped a MAGE-3-derived peptide (M3). M3 entrapped in F-ΔF nanotubes was more stable to a nonspecific protease treatment and both F-ΔF and F-ΔF-M3 showed no cellular toxicity for four cancerous and noncancerous cell lines used. F-ΔF-M3 showed significantly higher cellular uptake in RAW 267.4 macrophage cells compared to M3 alone and also induced in vitro maturation of dendritic cells (DCs). Immunization of mice with F-ΔF-M3 selected a higher number of IFN-γ secreting CD8+ T cells and CD4+ T compared to M3 alone. On day 21, a tumor growth inhibition ratio (TGI, %) of 41% was observed in a murine melanoma model. These results indicate that F-ΔF nanotubes are highly biocompatible, efficiently delivered M3 to generate cytotoxic T lymphocytes responses, and able to protect M3 from degradation under in vivo conditions. The F-ΔF dipeptide-based nanotubes may be considered as a good platform for further development as delivery agents.


Subject(s)
Antigens, Neoplasm/administration & dosage , Nanoparticle Drug Delivery System/administration & dosage , Testis/immunology , Animals , Humans , Immunotherapy/methods , MCF-7 Cells , Male , Melanoma/immunology , Melanoma/therapy , Mice , Mice, Inbred BALB C , Microscopy, Electron, Transmission , Nanotubes, Peptide , Neoplasm Transplantation , RAW 264.7 Cells
6.
PLoS Comput Biol ; 17(9): e1009384, 2021 09.
Article in English | MEDLINE | ID: mdl-34516563

ABSTRACT

Apart from the canonical fingers, palm and thumb domains, the RNA dependent RNA polymerases (RdRp) from the viral order Nidovirales possess two additional domains. Of these, the function of the Nidovirus RdRp associated nucleotidyl transferase domain (NiRAN) remains unanswered. The elucidation of the 3D structure of RdRp from the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), provided the first ever insights into the domain organisation and possible functional characteristics of the NiRAN domain. Using in silico tools, we predict that the NiRAN domain assumes a kinase or phosphotransferase like fold and binds nucleoside triphosphates at its proposed active site. Additionally, using molecular docking we have predicted the binding of three widely used kinase inhibitors and five well characterized anti-microbial compounds at the NiRAN domain active site along with their drug-likeliness. For the first time ever, using basic biochemical tools, this study shows the presence of a kinase like activity exhibited by the SARS-CoV-2 RdRp. Interestingly, a well-known kinase inhibitor- Sorafenib showed a significant inhibition and dampened viral load in SARS-CoV-2 infected cells. In line with the current global COVID-19 pandemic urgency and the emergence of newer strains with significantly higher infectivity, this study provides a new anti-SARS-CoV-2 drug target and potential lead compounds for drug repurposing against SARS-CoV-2.


Subject(s)
Antiviral Agents/pharmacology , Coronavirus RNA-Dependent RNA Polymerase/antagonists & inhibitors , Protein Domains , SARS-CoV-2/drug effects , Catalytic Domain , Computer Simulation , Coronavirus RNA-Dependent RNA Polymerase/chemistry , Coronavirus RNA-Dependent RNA Polymerase/metabolism , Humans
7.
J Virol ; 95(21): e0040621, 2021 10 13.
Article in English | MEDLINE | ID: mdl-34379515

ABSTRACT

Interferon regulatory factor 8 (IRF8), a myeloid lineage transcription factor, emerges as an essential regulator for microglial activation. However, the precise role of IRF8 during Japanese encephalitis virus (JEV) infection in the brain remains elusive. Here, we report that JEV infection enhances IRF8 expression in the infected mouse brain. Comparative transcriptional profiling of whole-brain RNA analysis and validation by quantitative reverse transcription-PCR (qRT-PCR) reveals an impaired interferon gamma (IFN-γ) and related gene expression in Irf8 knockout (Irf8-/-)-infected mice. Further, Ifnγ knockout (Ifnγ-/-) mice exhibit a reduced level of Irf8. Both Ifnγ-/- and Irf8-/- mice exhibit significantly reduced levels of activated (CD11b+ CD45hi, CD11b+ CD45lo, Cd68, and CD86) and infiltrating immune cells (Ly6C+, CD4, and CD8) in the infected brain compared to those of wild-type (WT) mice. However, a higher level of granulocyte cell (Ly6G+) infiltration is evident in Irf8-/- mice as well as the increased concentration of tumor necrosis factor alpha (TNF-α), interleukin-6 (IL-6), monocyte chemoattractant protein 1 (MCP1) levels in the brain. Interestingly, neither the Irf8-/- nor the Ifnγ-/- conferred protection against lethal JEV challenge to mice and exhibit augmentation in JEV replication in the brain. The gain of function of Irf8 by overexpressing functional IRF8 in an IRF8-deficient cell line attenuates viral replication and enhances IFN-γ production. Overall, we summarize that in the murine model of JEV encephalitis, IRF8 modulation affects JEV replication. We also show that lack of Irf8 affects immune cell abundance in circulation and the infected brain, leading to a reduction in IFN-γ level and increased viral load in the brain. IMPORTANCE Microglial cells, the resident macrophages in the brain, play a vital role in Japanese encephalitis virus (JEV) pathogenesis. The deregulated activity of microglia can be lethal for the brain. Therefore, it is crucial to understand the regulators that drive microglia phenotype changes and induce inflammation in the brain. Interferon regulatory factor 8 (IRF8) is a myeloid lineage transcription factor involved in microglial activation. However, the impact of IRF8 modulation on JEV replication remains elusive. Moreover, the pathways regulated by IRF8 to initiate and amplify pathological neuroinflammation are not well understood. Here, we demonstrated the effect of IRF8 modulation on JEV replication, microglial activation, and immune cells infiltration in the brain.


Subject(s)
Brain/virology , Encephalitis Virus, Japanese/immunology , Encephalitis, Japanese/immunology , Interferon Regulatory Factors/genetics , Interferon-gamma/immunology , Virus Replication/immunology , Animals , Brain/immunology , Encephalitis Virus, Japanese/physiology , Female , Gene Expression Regulation/immunology , Interferon Regulatory Factors/immunology , Interferon-gamma/genetics , Male , Mice , Mice, Knockout , Microglia/immunology , Microglia/physiology , Microglia/virology , Signal Transduction
8.
iScience ; 23(7): 101322, 2020 Jul 24.
Article in English | MEDLINE | ID: mdl-32688283

ABSTRACT

Precise regulation of innate immunity is crucial for development of appropriate host immunity against microbial infections and maintenance of immune homeostasis. MicroRNAs are small non-coding RNAs, post-transcriptional regulator of multiple genes, and act as a rheostat for protein expression. Here, we identified microRNA-30e-5p induced by hepatitis B virus and other viruses that act as a master regulator for innate immunity. Moreover, pegylated interferons treatment of patients with HBV for viral reduction also reduces miRNA. Additionally, we have also shown the immuno-pathological effects of miR-30e in patients with systemic lupus erythematosus (SLE) and mouse model. Mechanistically, miR-30e targets multiple negative regulators of innate immune signaling and enhances immune responses. Furthermore, sequestering of miR-30e in patients with SLE and mouse model significantly reduces type-I interferon and pro-inflammatory cytokines. Collectively, our study demonstrates the novel role of miR-30e in innate immunity and its prognostic and therapeutic potential in infectious and autoimmune diseases.

9.
Cell Immunol ; 349: 104043, 2020 03.
Article in English | MEDLINE | ID: mdl-32044112

ABSTRACT

Type I Interferon (IFN) signaling plays a critical role in dendritic cell (DC) development and functions. Inhibition of hyper type I IFN signaling promotes cDC2 subtype development. Relb is essential to development of cDC2 subtype and here we analyzed its effect on type I IFN signaling in DCs. We show that Relb suppresses the homeostatic type I IFN signaling in cDC2 cultures. TLR stimulation of FL-DCs led to RelB induction coinciding with fall in IFN signatures; conforming with the observation Relb expression reduced TLR stimulated IFN induction along with decrease in ISGs. Towards understanding mechanism, we show that effects of RelB are mediated by increased levels of IκBα. We demonstrate that RelB dampened antiviral responses by lowering ISG levels and the defect in cDC2 development in RelB null mice can be rescued in Ifnar1-/- background. Overall, we propose a novel role of RelB as a negative regulator of the type I IFN signaling pathway; fine tuning development of cDC2 subtype.


Subject(s)
Dendritic Cells/immunology , Interferon Type I/immunology , NF-KappaB Inhibitor alpha/physiology , Transcription Factor RelB/physiology , Amino Acid Sequence , Animals , Cell Differentiation , Cells, Cultured , Crosses, Genetic , Dendritic Cells/classification , Dendritic Cells/cytology , Gene Expression Regulation/immunology , Mice , NIH 3T3 Cells , Newcastle disease virus/immunology , Peptides/pharmacology , Receptor, Interferon alpha-beta/deficiency , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/physiology , Signal Transduction/immunology , Spleen/cytology , Transcription Factor RelB/deficiency , Transcription Factor RelB/genetics , Viral Load
10.
Cell Death Dis ; 8(10): e3078, 2017 10 05.
Article in English | MEDLINE | ID: mdl-28981114

ABSTRACT

Cancer is a multifactorial disease and virus-mediated carcinogenesis is one of the crucial factors, which is poorly understood. Human cytomegalovirus (HCMV) is a herpesvirus and its components have been evidenced to be associated with cancer of different tissue origin. However, its role in cancer remains unknown. Here, we identified a conserved herpesviral tegument protein known as pUL48 of HCMV, encoding deubiquitinase enzyme, as having a key role in carcinogenesis. We show using deubiquitinase sufficient- and deficient-HCMV that HCMV deubiquitinase is a key in inducing enhanced cellular metabolic activity through upregulation of several anti-apoptotic genes and downregulation of several pro-apoptotic genes expression. Furthermore, HCMV deubiquitinase acquires pro-tumor functions by inhibiting PRR-mediated type I interferon via deubiquitination of TRAF6, TRAF3, IRAK1, IRF7 and STING. Taken together, our results suggest that HCMV infection may promote oncogenesis by inhibiting innate immunity of the host.


Subject(s)
Carcinogenesis/genetics , Deubiquitinating Enzymes/immunology , Neoplasms/virology , Viral Matrix Proteins/immunology , Cell Line, Tumor , Cytomegalovirus/genetics , Cytomegalovirus/pathogenicity , Deubiquitinating Enzymes/genetics , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Immunity, Innate/genetics , Neoplasms/genetics , Neoplasms/immunology , Transcriptional Activation , Viral Matrix Proteins/genetics , Virus Replication/genetics
11.
J Immunol ; 197(4): 1029-34, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27421479

ABSTRACT

Dendritic cells (DCs) are a collection of different subtypes, each of which is characterized by specific surface markers, gene-expression patterns, and distinct functions. Members of the IFN regulatory factor family play critical roles in DC development and functions. Recently, Irf8 was shown to activate TGF-ß signaling, which led to exacerbated neuroinflammation in the experimental autoimmune encephalomyelitis mouse model. We analyzed the effect of Irf8 on TGF-ß/bone morphogenetic protein pathway-specific genes in DCs and identified Acvrl1, a type I TGF-ß superfamily receptor, as a gene strongly induced by Irf8 expression. Among various DC subtypes, Acvrl1 is differentially expressed in CD8α(+) DCs. ACVRL1 signaling augmented Irf8-directed classical CD8α(+) DC development. Irf8 expression is essential for plasmacytoid DC and CD8α(+) DC development, and this study demonstrates that ACVRL1 signaling plays a pivotal role whereby it suppresses plasmacytoid DC development while enhancing that of CD8α(+) DCs, thus contributing to DC diversity development.


Subject(s)
Activin Receptors, Type I/metabolism , Cell Differentiation/immunology , Dendritic Cells/cytology , Dendritic Cells/immunology , Signal Transduction/immunology , Activin Receptors, Type I/immunology , Activin Receptors, Type II , Animals , CD8 Antigens/immunology , Dendritic Cells/metabolism , Interferon Regulatory Factors/immunology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains
12.
PLoS One ; 10(3): e0118434, 2015.
Article in English | MEDLINE | ID: mdl-25785602

ABSTRACT

Ebolaviruses can cause severe hemorrhagic fever that is characterized by rapid viral replication, coagulopathy, inflammation, and high lethality rates. Although there is no clinically proven vaccine or treatment for Ebola virus infection, a virus-like particle (VLP) vaccine is effective in mice, guinea pigs, and non-human primates when given pre-infection. In this work, we report that VLPs protect Ebola virus-infected mice when given 24 hours post-infection. Analysis of cytokine expression in serum revealed a decrease in pro-inflammatory cytokine and chemokine levels in mice given VLPs post-exposure compared to infected, untreated mice. Using knockout mice, we show that VLP-mediated post-exposure protection requires perforin, B cells, macrophages, conventional dendritic cells (cDCs), and either CD4+ or CD8+ T cells. Protection was Ebola virus-specific, as marburgvirus VLPs did not protect Ebola virus-infected mice. Increased antibody production in VLP-treated mice correlated with protection, and macrophages were required for this increased production. However, NK cells, IFN-gamma, and TNF-alpha were not required for post-exposure-mediated protection. These data suggest that a non-replicating Ebola virus vaccine can provide post-exposure protection and that the mechanisms of immune protection in this setting require both increased antibody production and generation of cytotoxic T cells.


Subject(s)
Ebola Vaccines/immunology , Ebolavirus/immunology , Hemorrhagic Fever, Ebola/prevention & control , Post-Exposure Prophylaxis , Vaccination , Animals , Cytokines/blood , Hemorrhagic Fever, Ebola/immunology , Immunity , Mice , Mice, Knockout , Perforin/genetics
13.
J Immunol ; 191(12): 5993-6001, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24227775

ABSTRACT

Dendritic cells (DCs) are heterogeneous cell populations represented by different subtypes, each varying in terms of gene expression patterns and specific functions. Recent studies identified transcription factors essential for the development of different DC subtypes, yet molecular mechanisms for the developmental program and functions remain poorly understood. In this study, we developed and characterized a mouse DC progenitor-like cell line, designated DC9, from Irf8(-/-) bone marrow cells as a model for DC development and function. Expression of Irf8 in DC9 cells led to plasmacytoid DCs and CD8α(+) DC-like cells, with a concomitant increase in plasmacytoid DC- and CD8α(+) DC-specific gene transcripts and induction of type I IFNs and IL12p40 following TLR ligand stimulation. Irf8 expression in DC9 cells led to an increase in Id2 and Batf3 transcript levels, transcription factors shown to be important for the development of CD8α(+) DCs. We show that, without Irf8, expression of Id2 and Batf3 was not sufficient for directing classical CD8α(+) DC development. When coexpressed with Irf8, Batf3 and Id2 had a synergistic effect on classical CD8α(+) DC development. We demonstrate that Irf8 is upstream of Batf3 and Id2 in the classical CD8α(+) DC developmental program and define the hierarchical relationship of transcription factors important for classical CD8α(+) DC development.


Subject(s)
Basic-Leucine Zipper Transcription Factors/physiology , Dendritic Cells/cytology , Gene Expression Regulation/immunology , Inhibitor of Differentiation Protein 2/physiology , Interferon Regulatory Factors/physiology , Repressor Proteins/physiology , Animals , Basic-Leucine Zipper Transcription Factors/biosynthesis , Basic-Leucine Zipper Transcription Factors/genetics , CD8 Antigens/analysis , Cell Differentiation/drug effects , Cell Line , Dendrites/ultrastructure , Dendritic Cells/chemistry , Dendritic Cells/classification , Dendritic Cells/ultrastructure , Hematopoietic Stem Cells/cytology , Inhibitor of Differentiation Protein 2/biosynthesis , Inhibitor of Differentiation Protein 2/genetics , Interferon Regulatory Factors/biosynthesis , Interferon Regulatory Factors/genetics , Interferon-alpha/biosynthesis , Interferon-alpha/genetics , Interleukin-12 Subunit p40/biosynthesis , Interleukin-12 Subunit p40/genetics , Membrane Proteins/pharmacology , Mice , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Recombinant Fusion Proteins/metabolism , Repressor Proteins/biosynthesis , Repressor Proteins/genetics , Transduction, Genetic
14.
Virus Res ; 173(2): 280-5, 2013 May.
Article in English | MEDLINE | ID: mdl-23454489

ABSTRACT

Human herpesvirus 6B (HHV-6B) primary infections occur in early childhood and establish a life-long latency in the most healthy adults. HHV-6B was detectable in the peripheral blood mononuclear cells (PBMC) and granulocytes by serial genomic DNA dilution PCR till 10 pg of template DNA, in a healthy adult. Epstein Barr virus (EBV) mediated transformation of the PBMC resulted in establishment of a B-cell line. Southern hybridization with the PBMC as well as the cell line DNA showed distinct signals for high copy viral genomes and Gardella gel analysis indicated chromosomal integration of the HHV-6B. Integration site analysis in the PBMC and the cell line indicated an atypical viral integration in non-telomeric region of chromosome 12. Cell free culture medium of the cell line could infect different mononuclear cell lines, naïve or mitogen stimulated PBMC and was found to impart productive infection in a recipient T cell line. An HIV-1 LTR driven luciferase based reporter cell line was made and a single step assay was developed for estimating HHV-6B relative concentration in the culture supernatants. This study thus reports a new infectious HHV-6B isolate with uncommon integration site, spontaneous production from a cell line and also development of a simple relative HHV-6B titer assay.


Subject(s)
Herpesvirus 6, Human/genetics , Herpesvirus 6, Human/isolation & purification , Proviruses/genetics , Proviruses/isolation & purification , Roseolovirus Infections/virology , Adult , Asymptomatic Diseases , Chromosomes/virology , DNA, Viral/chemistry , DNA, Viral/genetics , Granulocytes/virology , Humans , Leukocytes, Mononuclear/virology , Male , Molecular Sequence Data , Sequence Analysis, DNA , Viral Load/methods
15.
J Immunol ; 189(7): 3548-56, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-22942423

ABSTRACT

Macrophages, when activated by IFN-γ and TLR signaling, elicit innate immune responses. IFN regulatory factor 8 (IRF8) is a transcription factor that facilitates macrophage activation and innate immunity. We show that, in resting macrophages, some IRF8 is conjugated to small ubiquitin-like modifiers (SUMO) 2/3 through the lysine residue 310. SUMO3-conjugated IRF8 failed to induce IL12p40 and other IRF8 target genes, consistent with SUMO-mediated transcriptional repression reported for other transcription factors. SUMO3-conjugated IRF8 showed reduced mobility in live nuclei and bound poorly to the IL12p40 gene. However, macrophage activation caused a sharp reduction in the amount of SUMOylated IRF8. This reduction coincided with the induction of a deSUMOylating enzyme, sentrin-specific peptidase 1 (SENP1), in activated macrophages. In transfection analysis, SENP1 removed SUMO3 from IRF8 and enhanced expression of IL12p40 and other target genes. Conversely, SENP1 knockdown repressed IRF8 target gene expression. In parallel with IRF8 deSUMOylation, macrophage activation led to the induction of proteins active in the SUMO pathway and caused a global shift in nuclear protein SUMOylation patterns. Together, the IRF8 SUMO conjugation/deconjugation switch is part of a larger transition in SUMO modifications that takes place upon macrophage activation, serving as a mechanism to trigger innate immune responses.


Subject(s)
Endopeptidases/physiology , Interferon Regulatory Factors/metabolism , Macrophage Activation/immunology , Animals , Cells, Cultured , Cysteine Endopeptidases , HEK293 Cells , Humans , Interferon Regulatory Factors/physiology , Interleukin-12 Subunit p40/metabolism , Lysine/metabolism , Macrophages/cytology , Macrophages/enzymology , Macrophages/immunology , Mice , NIH 3T3 Cells , Protein Binding/immunology , Repressor Proteins/physiology , Resting Phase, Cell Cycle/immunology , Small Ubiquitin-Related Modifier Proteins/metabolism , Sumoylation/immunology , Ubiquitins/metabolism
16.
FEBS Lett ; 585(9): 1331-7, 2011 May 06.
Article in English | MEDLINE | ID: mdl-21492741

ABSTRACT

We investigated mRNA expression of 49 nuclear hormone receptors (NRs) and 35 transcriptional coregulators in mouse bone marrow-derived dendritic cells (DCs) upon infection with Newcastle Disease virus (NDV) or murine cytomegalovirus (MCMV). These viruses regulated mRNA expression of some NRs among which NOR1 and LXRα were highly induced at mRNA and protein levels. Exogenous expression of the latter NRs repressed IRF3- or IRF7-induced transactivation of the interferon ß promoter and NDV infection further potentiated their repressive effect. The viral infection also significantly regulated mRNA expression of some coregulators, including HDAC1. Toll-like receptor ligands regulated NR and coregulator mRNA expression similar to the viruses. Thus, NRs and coregulators are integral components of DC-organizing anti-viral response wherein NOR1 and LXRα participate in regulating interferon production.


Subject(s)
Dendritic Cells/metabolism , Dendritic Cells/virology , Histone Deacetylase 1/metabolism , Muromegalovirus/physiology , Newcastle disease virus/physiology , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Cells, Cultured , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Profiling , HCT116 Cells , Histone Deacetylase 1/genetics , Host-Pathogen Interactions , Humans , Immunoblotting , Interferon-alpha/genetics , Interferon-alpha/metabolism , Liver X Receptors , Male , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Orphan Nuclear Receptors/genetics , Orphan Nuclear Receptors/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Steroid/genetics , Receptors, Steroid/metabolism , Receptors, Thyroid Hormone/genetics , Receptors, Thyroid Hormone/metabolism , Reverse Transcriptase Polymerase Chain Reaction
17.
J Biol Chem ; 286(10): 7893-7904, 2011 Mar 11.
Article in English | MEDLINE | ID: mdl-21216962

ABSTRACT

Dendritic cells (DCs) are key mediators of immune function through robust and tightly regulated presentation of antigen in the context of the MHC Class II. MHC Class II expression is controlled by the transactivator CIITA. CIITA expression in conventional DCs is uniquely dependent on an uncharacterized myeloid cell-specific promoter, CIITApI. We now identify in vivo the promoter structure and factors regulating CIITApI. In immature DCs transcription requires binding of PU.1, IRF8, NFκB, and Sp1 to the promoter. PU.1 binds independently at one site and in a required heterodimer with IRF8 at a composite element. DCs from IRF8-null mice have an unoccupied CIITApI promoter that can be rescued by reconstitution with IRF8 in vitro. Furthermore, mutation of either PU.1 site or the IFR8 site inhibits transcriptional activation. In vivo footprinting and chromatin immunoprecipitation reveals that DC maturation induces complete disassociation of the bound activators paralleled by recruitment of PRDM1/Blimp-1 to the promoter. PRDM1 is a transcriptional repressor with essential roles in B cells, T cells, NK cells, and DCs. We show that PRDM1 co-repressors, G9a and HDAC2, are recruited to CIITApI, leading to a loss of histone acetylation and acquisition of histone H3K9 dimethylation and heterochromatin protein 1γ (HP1γ). PRDM1 binding also blocks IRF8-mediated activation dependent on the PU.1/IRF composite element. Together these findings reveal the mechanisms regulating CIITA and, thus, antigen presentation in DCs, demonstrating that PRDM1 and IRF8/PU.1 counter-regulate expression. The activity of PRDM1 in silencing all three cell type-specific CIITA promoters places it as a central regulator of antigen presentation.


Subject(s)
Antigen Presentation/physiology , Dendritic Cells/metabolism , Interferon Regulatory Factors/metabolism , Nuclear Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Repressor Proteins/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism , Acetylation , Animals , Dendritic Cells/cytology , Dendritic Cells/immunology , Histocompatibility Antigens/genetics , Histocompatibility Antigens/immunology , Histocompatibility Antigens/metabolism , Histone Deacetylase 2/genetics , Histone Deacetylase 2/immunology , Histone Deacetylase 2/metabolism , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/immunology , Histone-Lysine N-Methyltransferase/metabolism , Histones/genetics , Histones/immunology , Histones/metabolism , Humans , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/immunology , Mice , Mice, Knockout , Nuclear Proteins/genetics , Nuclear Proteins/immunology , Positive Regulatory Domain I-Binding Factor 1 , Protein Structure, Tertiary , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/immunology , Repressor Proteins/genetics , Repressor Proteins/immunology , Response Elements/physiology , Trans-Activators/genetics , Trans-Activators/immunology , Transcription Factors/genetics , Transcription Factors/immunology , Transcription, Genetic/physiology
18.
Immunity ; 31(6): 941-52, 2009 Dec 18.
Article in English | MEDLINE | ID: mdl-20064451

ABSTRACT

Interleukin-21 (IL-21) is a pleiotropic cytokine that induces expression of transcription factor BLIMP1 (encoded by Prdm1), which regulates plasma cell differentiation and T cell homeostasis. We identified an IL-21 response element downstream of Prdm1 that binds the transcription factors STAT3 and IRF4, which are required for optimal Prdm1 expression. Genome-wide ChIP-Seq mapping of STAT3- and IRF4-binding sites showed that most regions with IL-21-induced STAT3 binding also bound IRF4 in vivo and furthermore revealed that the noncanonical TTCnnnTAA GAS motif critical in Prdm1 was broadly used for STAT3 binding. Comparing genome-wide expression array data to binding sites revealed that most IL-21-regulated genes were associated with combined STAT3-IRF4 sites rather than pure STAT3 sites. Correspondingly, ChIP-Seq analysis of Irf4(-/-) T cells showed greatly diminished STAT3 binding after IL-21 treatment, and Irf4(-/-) mice showed impaired IL-21-induced Tfh cell differentiation in vivo. These results reveal broad cooperative gene regulation by STAT3 and IRF4.


Subject(s)
Gene Expression Regulation , Interferon Regulatory Factors/metabolism , Interleukins/metabolism , STAT3 Transcription Factor/metabolism , Transcription Factors/genetics , Animals , B-Lymphocytes/immunology , Base Sequence , Binding Sites , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation , Genome-Wide Association Study , Interferon Regulatory Factors/genetics , Introns , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Positive Regulatory Domain I-Binding Factor 1 , STAT3 Transcription Factor/genetics
19.
Blood ; 112(10): 4028-38, 2008 Nov 15.
Article in English | MEDLINE | ID: mdl-18799728

ABSTRACT

PU.1, IKAROS, E2A, EBF, and PAX5 comprise a transcriptional network that orchestrates B-cell lineage specification, commitment, and differentiation. Here we identify interferon regulatory factor 8 (IRF8) as another component of this complex, and show that it also modulates lineage choice by hematopoietic stem cells (HSCs). IRF8 binds directly to an IRF8/Ets consensus sequence located in promoter regions of Sfpi1 and Ebf1, which encode PU.1 and EBF, respectively, and is associated with transcriptional repression of Sfpi1 and transcriptional activation of Ebf1. Bone marrows of IRF8 knockout mice (IRF8(-/-)) had significantly reduced numbers of pre-pro-B cells and increased numbers of myeloid cells. Although HSCs of IRF8(-/-) mice failed to differentiate to B220(+) B-lineage cells in vitro, the defect could be rescued by transfecting HSCs with wild-type but not with a signaling-deficient IRF8 mutant. In contrast, overexpression of IRF8 in HSC-differentiated progenitor cells resulted in growth inhibition and apoptosis. We also found that IRF8 was expressed at higher levels in pre-pro-B cells than more mature B cells in wild-type mice. Together, these results indicate that IRF8 modulates lineage choice by HSCs and is part of the transcriptional network governing B-cell lineage specification, commitment, and differentiation.


Subject(s)
B-Lymphocytes/metabolism , Cell Differentiation/physiology , Hematopoietic Stem Cells/metabolism , Interferon Regulatory Factors/metabolism , Response Elements/physiology , Signal Transduction/physiology , Animals , B-Lymphocytes/cytology , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Hematopoietic Stem Cells/chemistry , Ikaros Transcription Factor/genetics , Ikaros Transcription Factor/metabolism , Interferon Regulatory Factors/genetics , Leukocyte Common Antigens/genetics , Leukocyte Common Antigens/metabolism , Mice , Mice, Knockout , PAX5 Transcription Factor/genetics , PAX5 Transcription Factor/metabolism , Protein Binding/physiology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism
20.
J Biol Chem ; 283(37): 25660-25670, 2008 Sep 12.
Article in English | MEDLINE | ID: mdl-18635538

ABSTRACT

Viral infection activates Toll-like receptor and RIG-I (retinoic acid-inducible gene I) signaling pathways, leading to phosphorylation of IRF3 (interferon regulatory factor 3) and IRF7 and stimulation of type I interferon (IFN) transcription, a process important for innate immunity. We show that upon vesicular stomatitis virus infection, IRF3 and IRF7 are modified not only by phosphorylation but by the small ubiquitin-related modifiers SUMO1, SUMO2, and SUMO3. SUMOylation of IRF3 and IRF7 was dependent on the activation of Toll-like receptor and RIG-I pathways but not on the IFN-stimulated pathway. However, SUMOylation of IRF3 and IRF7 was not dependent on their phosphorylation, and vice versa. We identified Lys(152) of IRF3 and Lys(406) of IRF7 to be their sole small ubiquitin-related modifier (SUMO) conjugation site. IRF3 and IRF7 mutants defective in SUMOylation led to higher levels of IFN mRNA induction after viral infection, relative to the wild type IRFs, indicating a negative role for SUMOylation in IFN transcription. Together, SUMO modification is an integral part of IRF3 and IRF7 activity that contributes to postactivation attenuation of IFN production.


Subject(s)
Gene Expression Regulation , Interferon Regulatory Factor-3/metabolism , Interferon Regulatory Factor-7/metabolism , Interferon Type I/metabolism , Virus Diseases/metabolism , Animals , Humans , Lysine/chemistry , Membrane Proteins/metabolism , Mice , Models, Biological , Mutation , NIH 3T3 Cells , Receptors, Retinoic Acid/metabolism , Toll-Like Receptors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...