Skip to main content

Main menu

  • Home
  • Articles
    • Current Issue
    • Next in The JI
    • Archive
    • Brief Reviews
      • Neuroimmunology: To Sense and Protect
    • Pillars of Immunology
    • Translating Immunology
    • Most Read
    • Top Downloads
    • Annual Meeting Abstracts
  • COVID-19/SARS/MERS Articles
  • Info
    • About the Journal
    • For Authors
    • Journal Policies
    • Influence Statement
    • For Advertisers
  • Editors
  • Submit
    • Submit a Manuscript
    • Instructions for Authors
    • Journal Policies
  • Subscribe
    • Journal Subscriptions
    • Email Alerts
    • RSS Feeds
    • ImmunoCasts
  • More
    • Most Read
    • Most Cited
    • ImmunoCasts
    • AAI Disclaimer
    • Feedback
    • Help
    • Accessibility Statement
  • Other Publications
    • American Association of Immunologists
    • ImmunoHorizons

User menu

  • Subscribe
  • Log in

Search

  • Advanced search
The Journal of Immunology
  • Other Publications
    • American Association of Immunologists
    • ImmunoHorizons
  • Subscribe
  • Log in
The Journal of Immunology

Advanced Search

  • Home
  • Articles
    • Current Issue
    • Next in The JI
    • Archive
    • Brief Reviews
    • Pillars of Immunology
    • Translating Immunology
    • Most Read
    • Top Downloads
    • Annual Meeting Abstracts
  • COVID-19/SARS/MERS Articles
  • Info
    • About the Journal
    • For Authors
    • Journal Policies
    • Influence Statement
    • For Advertisers
  • Editors
  • Submit
    • Submit a Manuscript
    • Instructions for Authors
    • Journal Policies
  • Subscribe
    • Journal Subscriptions
    • Email Alerts
    • RSS Feeds
    • ImmunoCasts
  • More
    • Most Read
    • Most Cited
    • ImmunoCasts
    • AAI Disclaimer
    • Feedback
    • Help
    • Accessibility Statement
  • Follow The Journal of Immunology on Twitter
  • Follow The Journal of Immunology on RSS

Bcl-3-Regulated Transcription from Major Immediate-Early Promoter of Human Cytomegalovirus in Monocyte-Derived Macrophages

Kashif Aziz Khan, Alain Coaquette, Christian Davrinche and Georges Herbein
J Immunol June 15, 2009, 182 (12) 7784-7794; DOI: https://doi.org/10.4049/jimmunol.0803800
Kashif Aziz Khan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Alain Coaquette
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Christian Davrinche
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Georges Herbein
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

Monocytes/macrophages are key cells in the pathogenesis of human CMV (HCMV) infection, but the in vitro rate of viral production in primary human monocyte-derived macrophages (MDM) is considerably lower than in fibroblasts. Considering that the NF-κB signaling pathway is potentially involved in the replication strategy of HCMV through efficient transactivation of the major immediate-early promoter (MIEP), efficient viral replication, and late gene expression, we investigated the composition of the NF-κB complex in HCMV-infected MDMs and fibroblasts. Preliminary studies showed that HCMV could grow in primary MDM culture but that the viral titer in culture supernatants was lower than that observed in the supernatants of more permissive MRC5 fibroblasts. EMSA and microwell colorimetric NF-κB assay demonstrated that HCMV infection of MDMs increased p52 binding activity without activating the canonical p50/p65 complex. Moreover, Bcl-3 was up-regulated and was demonstrated to associate with p52, indicating p52/Bcl-3 complexes as the major component of the NF-κB complex in MDMs. Luciferase assays in promonocytic U937 cells transfected with an MIEP-luciferase reporter construct demonstrated MIEP activation in response to p52 and Bcl-3 overexpression. Chromatin immunoprecipitation assay demonstrated that p52 and Bcl-3 bind the MIEP in acutely HCMV-infected MDMs. In contrast, HCMV infection of MRC5 fibroblasts resulted in activation of p50/p65 heterodimers. Thus, activation of p52/Bcl-3 complexes in MDMs and p50/p65 heterodimers in fibroblasts in response to HCMV infection might explain the low-level growth of the virus in MDMs vs efficient growth in fibroblasts.

Human CMV (HCMV)3 is an opportunistic, species-specific herpesvirus that infects a large part of the population worldwide and causes asymptomatic latent infection in healthy people. However, it can cause severe disease in the absence of an effective immune response, especially in patients with AIDS and in immunocompromised solid-organ and bone marrow allograft recipients. Furthermore, HCMV infection during pregnancy can cause permanent birth defects. Breast-feeding is the most common route of transmission from mother to child; however, HCMV can be transmitted via the placenta and during delivery. An association between HCMV infection and the development of cardiovascular disease, including atherosclerosis and arterial restenosis, has also been described (reviewed in Ref. 1).

Histological and immunohistochemical studies have demonstrated the presence of infected cells in virtually all organs and the virus targets a variety of cell types in vivo, including macrophages, endothelial cells, epithelial cells, fibroblasts, stromal cells, neuronal cells, smooth muscle cells, and hepatocytes (2). Blood monocytes and tissue macrophages are believed to serve as target cells in infected organs, acting as viral disseminators throughout the host or as sites of HCMV latency (3). The macrophage is the predominant infiltrating cell type found in HCMV-infected organs (4, 5). HCMV infects a large number of cell types in vitro as well, including skin or lung fibroblasts, vascular smooth muscle cells, retina pigment and kidney epithelial cells, placental trophoblast cells, hepatocytes, neuronal and glial brain cells, vascular endothelial cells, monocyte-derived dendritic cells, and monocyte-derived macrophages (MDM; Ref. 2). The cell type most widely used to grow HCMV is the fibroblast, which produces high titers of infectious virus after in vitro infection. Other primary cells, such as MDMs, are permissive for HCMV replication, but the rate of viral production in these cells is considerably lower than in fibroblasts (2, 6). The cellular and/or viral factors responsible for the low production of HCMV in primary human MDMs (4) remain unknown.

Interactions between viral and cellular proteins generate an intracellular signal transduction pathway that produces a diverse cascade of cellular responses leading to activation of transcription factors. HCMV has been reported to stimulate activation of a number of transcription factors including NF-κB, AP-1, and Sp1. NF-κB is a family of DNA-binding proteins with five members in mammals: Rel (c-Rel); RelA (p65); RelB; NF-κB1 (p50 and its precursor p105); and NF-κB2 (p52 and its precursor p100). These proteins exist in various homo- and heterodimeric complexes. NF-κB is normally sequestered in the cytoplasm in association with the members of the IκB family. Phosphorylation and ubiquitination of IκB by IκB kinase (IKK) free NF-κB to translocate to the nucleus to regulate the transcription of target genes. In mammals, the IκB family has seven known members: IκBα; IκBβ; IκBγ; IκBε; Bcl-3; and the precursor proteins p100 and p105 (7). Bcl-3 is an oncoprotein that, unlike other IκBs, is found in the nucleus and is not proteolytically degraded upon cell stimulation. It contains two transactivation domains upstream and downstream of ankyrin repeats and can activate (8, 9, 10, 11, 12, 13, 14, 15) or repress (16, 17, 18) transcription through NF-κB sites. This dual activity of Bcl-3 might be modulated by posttranslational modifications or the phosphorylation state of the protein (19). Bcl-3 preferentially binds with p50 and p52 homodimers and activates transcription through a ternary complex with nuclear coregulators such as JAB1, Tip60, and Bard1 (20).

Many viruses, including HCMV, have been reported to have NF-κB-binding sites in their transcriptional response elements and exploit cellular NF-κB to drive their own transcriptional events. HCMV infection activates the NF-κB signaling pathway in fibroblasts (21, 22) and monocytes (23). NF-κB and upstream IKK2 have been demonstrated as requirements for efficient transactivation of the major immediate-early promoter (MIEP), late gene expression, and viral replication of HCMV (24, 25, 26, 27). Conversely, others have reported inhibitory (28) or neutral (29) roles for NF-κB in HCMV replication.

Because distinct NF-κB complexes can be activated in different cell types in response to similar stimuli (30, 31), we hypothesized that this mechanistic versatility might be involved in the varied transcription of HCMV in different cell types. NF-κB proteins exist in dimmers, and at least 12 different dimers have been reported. Binding of different dimers to NF-κB binding sites at comparable levels of recruitment can support different levels of transcription (32, 33). Thus, the presence of distinct NF-κB dimers in different cell types might contribute to varied levels of viral replication in the different cell types. Because inhibitors of IKK/NF-κB have been proposed as therapeutic tools for controlling HMCV replication (27, 34), it is important to know which NF-κB pathway is activated and contributes to persistent replication of HCMV in macrophages. Here, we report differences in the composition of the NF-κB complex between HCMV-infected MDMs and fibroblasts, which might be responsible for the differences in virus production between these cell types.

Materials and Methods

Reagents

Anti-p50, anti-p65, anti-RelB, anti-c-Rel, anti-p52, anti-Bcl-3, and the single-stranded NF-κB oligonucleotide and mutated oligonucleotide were purchased from Santa Cruz Biotechnology. Anti-IκBα, anti-phospho-IκBα, anti-IKKα, anti-IKKβ, anti-IKKγ, and anti-p- IKKγ were purchased from Cell Signaling Technologies; anti-pIKKα was purchased from Abcam, anti-p-IKKβ was purchased from US Biological, and peroxidase-conjugated secondary anti-rabbit and anti-mouse Ig were obtained from Jackson ImmunoResearch Laboratories. A scrambled control, p52, and Bcl-3 small interfering RNA (siRNA) duplex were purchased from Santa Cruz Biotechnology. Plasmid constructs p52 (pUNO-hNFkBp52a), p50 (pUNO-hNFkBp50), and p65 (pUNO-hNFkBp65) were purchased from Invivogen. Bcl-3-expressing plasmid (pCMV6-XL4-Bcl-3) and pCMV-Luc were obtained from Origene and PlasmidFactory, respectively. All other reagents were obtained from Sigma-Aldrich unless stated otherwise. Peripheral blood and human AB serum of a healthy donor was provided by Établissement Français du Sang (Bourgogne Franche-Comté, France). MRC5 human fibroblasts were obtained from BioMérieux. Cell culture flasks and cell scrapers were purchased from Nunc.

Isolation and culture of MDMs

PBMCs were isolated by Ficoll gradient centrifugation, as previously reported (35). Blood from a healthy donor was diluted with equal amounts of PBS, overlaid on Ficoll medium (Eurobio) and centrifuged at 900 × g for 30 min at 25°C. The PBMC band was removed and washed twice with PBS. Cell count was determined by Malassez cytometer (Poly Labo), and the cells were resuspended in serum-free RPMI 1640. The cells were plated in plastic cell culture flasks and incubated at 37°C. After 2 h, nonadherent cells were removed to enrich the culture for monocytes. Adherent cells (>95% CD14+ by flow cytometric analysis) were washed with sterile PBS and cultured in RPMI 1640 supplemented with 10% (v/v) human AB serum, penicillin (100 IU/ml), and streptomycin (100 μg/ml) for 7–10 days to allow the monocytes to differentiate into macrophages.

HCMV infection of MDMs and MRC5 fibroblasts

Cell-free virus stock was prepared by propagating four strains of HCMV (high-passage laboratory strain AD169 (AD169); a clinical isolate HCMV-DB; TB40/E; and TB40/F) in MRC5 human fibroblasts as previously described (36). AD169 is a highly passaged laboratory strain of HCMV originally isolated from the adenoids of a child (37). The clinical isolate HCMV-DB used in this study was isolated from a cervical swab specimen from a 30-year-old pregnant woman. Strains TB40/E and TB40/F have been developed by 22 passages in endothelial cells and fibroblasts, respectively (38). MRC5 human fibroblasts were cultured in MEM with 10% FBS, penicillin (100 IU/ml), and streptomycin (100 μg/ml). Cells were infected with a viral isolate when the monolayer was confluent, and virus was collected when cytopathic effects were >90%. Supernatants were clarified by centrifugation and stored at −80°C until use. Virus titers were determined by plaque-forming assay in MRC5 human fibroblasts as previously described (39). The MDM cultures were infected at different multiplicities of infection (MOI) at days 7–10 postisolation, incubated for 12 h at 37°C, washed thoroughly, and covered with fresh medium.

Real-time PCR for quantification of viral titer

Quantification of viral titer in cell culture supernatants was performed by real-time PCR as previously described (36). DNA was extracted from 100 μl of supernatant using the KingFisher automatic instrument (Thermo Labsystems) and a QIAamp kit (Qiagen) according to the recommendations of the manufacturer. After elution in 100 μl of buffer, 5 μl of the DNA were used for PCR. HCMV DNA in the samples was quantified by real-time PCR using TaqMan technology on an ABI Prism 7000 Sequence Detection System (Applied Biosystems) according to the manufacturer’s recommendations. The sequences of the primers used were 5′-AACATAAGGACTTTTCACACTTTT and 5′-GAATACAGACACTTAGAGCTCGGGT. The sequence of the TaqMan PCR probes was FAM 5′-CTGGCCAGCACGTATCCCAACAGCA-3′ TAMRA. Reaction samples had a final volume of 25 μl and contained 5 μl of extracted DNA and 20 μl of the mixture containing 12.5 μl of 2× TaqMan Universal PCR Master Mix (Applied Biosystems), 800 nM concentrations of each primer, and 200 nM TaqMan probe. An internal positive control, TaqMan Exogenous Internal Positive Control Reagents VIC Probe (Applied Biosystems), was included in each run to distinguish target negatives from PCR inhibition. A distilled water sample and a positive control with 3000 copies of HCMV DNA were processed in parallel with samples. Amplification conditions were 50°C for 2 min and 95°C for 10 min followed by 45 cycles of 95°C for 15 s and 60°C for 1 min. To generate the HCMV external quantitative standard curve, PCR was performed on a plasmid containing one copy of the target sequence that had been serially diluted from 2 × 106 to 2 × 102 copies/ml. Final quantification was performed with the comparative threshold value (CT) method using the ABI Prim 7000 SDS software.

Isolation of nuclear and cytoplasmic extracts

Isolation of nuclear and cytoplasmic extracts was performed as previously described (40). Cells were scraped from the plastic surface of the culture dishes and washed with wash buffer (10 mM HEPES (pH 7.6), 10 mM KCl, 2 mM MgCl2, 1 mM EDTA). Cell pellets were then incubated on ice with cytoplasmic isolation buffer (10 mM HEPES (pH 7.6), 10 mM KCl, 2 mM MgCl2, 1 mM EDTA, 0.02% Nonidet P-40). Cytoplasmic extracts were collected by centrifugation, and the nuclear pellets were washed twice in wash buffer, spun, and incubated for 15 min on ice with nuclear isolation buffer (20 mM HEPES (pH 7.6), 420 mM NaCl, 1.5 mM MgCl2, 0.2 mM EDTA, 25% glycerol). Supernatants containing nuclear extracts were collected by centrifugation and stored at −80°C. Protease inhibitors (1 mM DTT, 1 mM PMSF, 1 μg/ml aprotinin, 1 μg/ml leupeptin, 1 μg/ml pepstatin) were added to all solutions. Protein concentration in nuclear and cytoplasmic extracts was determined by the Bradford method using a BioPhotometer (Eppendorf).

EMSA

To measure NF-κB activation, EMSA was conducted as previously described (41). Briefly, nuclear extracts prepared from HCMV-infected cells were incubated with 20 fmol of biotin-end-labeled 45-mer double-stranded NF-κB oligonucleotide, 5-TTGTTACAAGGGACTTTCCGCTGGGGACTTTCCAGGGAGGCGTGG-3 (bolded letters indicate NF-κB binding sites in the MIEP of HCMV) in the presence of binding buffer (10 mM Tris, 50 mM KCl, 1 mM DTT at pH 7.5 and polydeoxyinosinic-polydeoxycytidylic acid, 50 ng/μl). NF-κB oligonucleotide was labeled with biotin using the Biotin 3′ End DNA Labeling kit (Pierce), and complementary pairs were annealed by heating in boiling water for 5 min and then cooling slowly to room temperature. DNA-protein complexes were resolved from free oligonucleotide on a 6% native polyacrylamide gel in 1× Tris-borate-EDTA buffer using a MiniPROTEAN 3 Cell (Bio-Rad) and were transferred to a Biodyne precut nylon membrane (Pierce) using the Mini Trans-Blot Electrophoretic Transfer Cell (Bio-Rad). Biotin-end-labeled DNA was detected using the LightShift Chemiluminescent EMSA kit (Pierce). A control Epstein-Barr nuclear Ag (EBNA) system (Pierce) containing biotin-EBNA control DNA and EBNA extract were assayed in parallel with the sample to ensure that the kit components and the overall procedure were working properly.

Microwell colorimetric NF-κB assay

Microwell colorimetric NF-κB assay was performed as described previously (42) using the Trans-AM NF-κB family transcription factor assay kit (Active motif). Briefly, cell extracts were incubated in a 96-well plate coated with an oligonucleotide containing the NF-κB consensus binding site (5′-GGGACTTTCC-3′). Activated transcription factors from extracts that bound specifically to the respective immobilized oligonucleotide were detected using Abs to NF-κB p50, p52, p65, RelB, and c-Rel subunits followed by a secondary Ab conjugated to HRP in an ELISA-like assay. Optical density was read within 5 min on a Multilabel Reader (PerkinElmer) at 450 nm. The specificity of the assay was validated by including both the wild-type and mutated oligonucleotides in the reaction. Raji nuclear extract was used a positive control.

Western blot

Cellular extracts of HCMV-infected cells were used to examine different protein expression by Western blot according to previously described procedures (40). Cellular extracts were resolved by 10% SDS-PAGE using a MiniPROTEAN 3 Cell (Bio-Rad). The proteins were electrotransferred onto a PVDF membrane (Amersham Biosciences) using Mini Trans-Blot Electrophoretic Transfer Cell (Bio-Rad). The membranes were probed with primary Abs followed by HRP-conjugated secondary Ig raised against the appropriate species; bands were detected using the ECL Plus kit (Amersham).

Viral entry assay

Viral entry into MDMs and MRC5 fibroblasts was assayed as described previously (43). Cells were incubated at 37°C with HCMV-DB at MOIs of 1 and 10 for 2 h and washed three times with PBS. Cells were treated with 0.25% trypsin for 10 min to release the virions that had adhered to the surface but had not entered the cell. The cells were pelleted and washed once with serum neutralization solution and three times with PBS. DNA was extracted from the cell pellet using the KingFisher automatic instrument (Thermo Labsystems) and a QIAamp kit (Qiagen) according to the recommendations of the manufacturers. Samples of eluted DNA were analyzed by PCR using primers specific for the MIEP of HCMV (sense, 5′-TGGGACTTTCCTACTTGG-3′; antisense, 5′-CCAGGCGATCTGACGGTT-3′). The β-globin PCR gene was used as an internal control (sense, 5′-TCCCCTCCTACCCCTACTTTCTA-3′; antisense, 5′-TGCCTGGACTAATCTGCAAGAG-3′). The amplification products were resolved by 2% agarose gel electrophoresis and visualized by ethidium bromide staining.

Coimmunoprecipitation test

Total cellular extracts were prepared from uninfected and HCMV-infected primary macrophages 5 days postinfection. Cellular extracts were precleared with 50 μl of protein A-Sepharose 50% suspension (Amersham) for 1 h at 4°C. Bcl-3 and control Abs were added to the cleared supernatant and the mixture was incubated overnight at 4°C. Immune complexes were precipitated with 50 μl of protein A-Sepharose suspension and washed, and bound proteins were eluted with 30 μl of sample buffer. SDS-PAGE and Western blot were performed as described above using anti-p52 primary Ab.

Chromatin immunoprecipitation assay (ChIP)

ChIP assay was performed on HCMV-infected MDMs using EZ-Magna ChIP A (Upstate) according to a previously described procedure (44). Briefly, MDMs (0.5 × 106 cells/reaction) were cross-linked with 1% formaldehyde and lysed, and nuclear extracts were sonicated to obtain DNA fragments ∼200–1000 bp long. After centrifugation, the nuclear extracts were diluted 10-fold with ChIP dilution buffer, and 1% of the material was saved as input. Nuclear extracts were incubated with 5 μg of control IgG, anti-Bcl-3, or anti-p52 Ab, and 20 μl of protein A magnetic beads slurry overnight at 4°C. Magnetic beads were separated and washed using a magnetic separator (Upstate). Immune complexes were eluted, and cross-linking was reversed by adding proteinase K and incubating at 62°C for 2 h. Immunoprecipitated DNA and input were analyzed by PCR using primers specific for the MIEP of HCMV as described above. The amplification product was run in a 2% agarose gel and visualized by ethidium bromide staining. Enrichment of MIEP was measured by real-time PCR using the ABI Prism 7000 Sequence Detection System (Applied Biosystems) according to the manufacturer’s recommendations.

Transfection and luciferase assay

U937 cells (2 × 106) were cotransfected with 20 μg of pCMV-Luc, which contains the luciferase reporter gene under control of MIEP, and a p52 plus Bcl3 or p50 plus p65 expression plasmid or the corresponding empty cloning vectors using the GenePulser XL electroporation system (Bio-Rad) according to manufacturer’s instructions. Luciferase activity in cell extracts was measured 24 h posttransfection using luciferase assay reagent (Promega) and a 20/20n Luminometer (Turner Biosystems). Luciferase activity was normalized to the total protein concentration and expressed as relative light units per microgram of protein. MRC5 fibroblasts were transfected with p52, Bcl-3, and p52 plus Bcl-3 expression plasmid using lipofectamine RNAiMAX (Invitrogen).

RNA interference

MDM cultures (0.5 × 106 cells) were transfected with a scrambled control or p52 plus Bcl-3 or p50 plus p65 siRNA duplex (Santa Cruz Biotechnology) using Lipofectamine RNAiMAX (Invitrogen). MDMs were infected with the clinical isolate HCMV-DB 24 h posttransfection, and viral titers in culture supernatants were determined at the indicated times postinfection by real-time PCR, as described above. For monitoring knockdown, total cellular extracts were prepared daily for 3 days posttransfection. Expression of p52, Bcl-3, p50, and p65 protein was analyzed by Western blot as described above. Transfection efficiency was monitored using a fluorescein-conjugated scrambled control duplex and exceeded 50% in MDMs and 90% in MRC5 fibroblasts (data not shown).

Statistical analysis

Values are the means and SDs of independent experiments. Statistical analysis was performed by Student’s t test, and differences were considered significant at a value of p < 0.05. Microsoft Excel was used to construct the plots.

Results

Low-level HCMV replication in primary MDMs vs efficient replication in fibroblasts

The ability of HCMV (AD169 and the clinical isolate HCMV-DB) to infect MDM and MRC5 fibroblast cultures was examined at different MOIs. MDM and MRC5 fibroblast cultures were infected as described in Materials and Methods, and supernatants were analyzed for production of extracellular virus by real-time PCR. The supernatants of the MDM culture infected with AD169 showed peak viral titer on day 8 postinfection at MOIs of 0.1 and 1. For the clinical isolate HCMV-DB, peak viral titer occurred at day 6 (MOI 0.1) and day 2 (MOI 1) postinfection (Fig. 1⇓, A–C). Supernatants of infected cultures assayed for 30 days showed similar growth patterns (data not shown). Moreover, no cytopathic effects were observed in the infected MDMs. In contrast, the viral titers in culture supernatants from MRC5 fibroblasts infected with the same viral isolates were much higher than those from MDM cultures (Fig. 1⇓, E–G). To assess the possibility that blocked viral entry influenced the differences in the viral titers, viral entry was assayed in both MDMs and MRC5 fibroblasts. As shown in Fig. 1⇓, D and H, viral entry was similar in both cell types. To rule out the possibility that cell death contributed to the low viral production by MDM cultures, infected MDMs were examined for viability at 5 and 10 days postinfection by trypan blue exclusion. There were no differences in trypan blue exclusion between the control and infected groups; <1% dead cells were observed in the control and infected groups (data not shown), indicating that the inability of the culture to support high viral replication was not due to MDM death. To assess the differential growth of HCMV in MDMs vs MRC5 fibroblasts, expression of HCMV pp65 protein was assayed 5 days postinfection in MDMs and MRC5 fibroblasts infected with HCMV-DB. HCMV pp65 protein was expressed in 1–2% of MDM cells, whereas most of the cells in the MRC5 fibroblast cultures expressed this protein (data not shown).

FIGURE 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 1.

Growth curve of HCMV in MDMs vs MRC5 fibroblasts. The MDM and the MRC5 cultures were left uninfected or infected at indicated MOIs with laboratory strain AD169 (A and E) and the clinical isolate HCMV-DB (B and F). Viral titers in culture supernatants were determined at the indicated times postinfection by real-time PCR. Peak viral titers in supernatants after infection of MDMs (C) and MRC5 fibroblasts (G) with laboratory strain AD169 and the clinical isolate HCMV-DB. For each point, mean values ± SD of duplicate samples are shown. Noninfected MDMs (D) and MRC5 fibroblasts (H) and cultures infected at the indicated MOI for 2 h were treated with trypsin for 10 min and then washed. Samples of extracted DNA were analyzed by PCR using primers specific for the MIEP of HCMV and for β-globin (internal loading control). The amplification products were resolved by 2% agarose gel electrophoresis and visualized by ethidium bromide staining. IE, Immediate-early Ag.

Subsequently, the growth of the clinical isolate HCMV-DB in MDMs was compared with that of the endothelial cell-propagated HCMV strain (TB40/E) and the fibroblast-propagated strains (AD169 and TB40/F). The endothelial cell-propagated strain TB40/E has been reported to replicate efficiently in MDMs (45). The clinical isolate HCMV-DB and the TB40/E strain grew better in MDMs than did the fibroblast-propagated strains (Fig. 2⇓).

FIGURE 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 2.

Growth of the clinical isolate HCMV-DB, endothelial cell-propagated HCMV strain (TB40/E), and fibroblast-propagated strains (TB40/F and AD169) in MDM cultures. The MDM cultures were left uninfected or infected at an MOI of 1 with different HCMV strains; viral titers in culture supernatants were determined at the indicated times postinfection by real-time PCR. For each point, mean values ± SD of duplicate samples are shown.

Activation of the noncanonical NF-κB pathway in HCMV-infected MDMs

Because HCMV infection of fibroblasts and monocytes results in activation of transcription factor NF-κB (22, 23) and because the NF-κB signaling pathway potentially contributes to the HCMV replication strategy (24, 25, 26, 27), we evaluated the activation and composition of NF-κB complexes in MDMs and fibroblasts infected with the clinical isolate HCMV-DB. At 5 days postinfection, EMSA demonstrated that infection of MDMs and MRC5 fibroblasts with HCMV resulted in activation of NF-κB (Fig. 3⇓, A and F). The gel shift bands were specific; formation of the complex was diminished by inclusion of an unlabeled NF-κB oligonucleotide but not by inclusion of a mutated NF-κB oligonucleotide (data not shown). To identify the subunits in the NF-κB complexes, nuclear extracts from HCMV-infected MDMs and MRC5 fibroblasts were incubated with Abs against different NF-κB subunits and against Bcl-3, and DNA-binding activity was assayed by EMSA. Interference of NF-κB activation by anti-p52 Ab and anti-Bcl-3 Ab (Fig. 3⇓B) suggested that p52 and Bcl-3 were the major components of the activated NF-κB complex in HCMV-infected MDMs. NF-κB subunit binding activity was also evaluated by microwell colorimetric NF-κB assay using Abs against NF-κB subunits (p50, p52, p65, RelB, and c-Rel). The results revealed an increase in the binding activity of p52 in HCMV-infected MDMs 5 days postinfection (Fig. 3⇓C). Overall, the results obtained by both techniques indicate that activated NF-κB complexes in HCMV-DB-infected MDMs involve the NF-κB subunit p52 (which is known to bind to NF-κB binding sites as a homodimer; see Ref. 7, 46) and Bcl-3 (which is known to bind with p52 and p50 homodimers; see Ref. 8), suggesting a p52/Bcl-3 complex. Recent studies have demonstrated the activation of the canonical p50/p65 NF-κB complex in monocytes early after HCMV binds to cell membrane receptors (47, 48). Therefore, we assayed the composition of NF-κB early after infection and found that the canonical p50/p65 NF-κB complex was activated during early phases of MDM infection by HCMV (Fig. 3⇓C), with a shift to the p52/Bcl-3 complex occurring later during infection.

FIGURE 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 3.

Activation and composition of NF-κB complexes in MDMs and in MRC5 fibroblasts after HCMV infection. MDM cultures (A) and MRC5 fibroblasts (F) were infected with the clinical isolate HCMV-DB at an MOI of 1. Nuclear extracts were prepared at 5 days postinfection and then assayed for NF-κB activation by EMSA. B and G, Interference of NF-κB activation in response to HCMV-DB by Abs against NF-κB subunits: Nuclear extracts from infected MDMs (B) and MRC5 fibroblasts (G) were incubated for 20 min with anti-p50, anti-p52, anti-p65, anti-c-Rel, anti-RelB, and anti-Bcl-3 Abs and then assayed for NF-κB binding activity by EMSA. C and H, Nuclear extracts from HCMV-DB-infected MDMs (C) and MRC5 fibroblasts (H) were assayed at 6 h and 5 days postinfection for NF-κB binding subunits by Microwell colorimetric NF-κB assay. NF-κB activation is shown in fold increase relative to NF-κB binding activity in uninfected cells (set equal to 1). Mean values ± SD of two independent experiments are shown. D and I, IκBα phosphorylation and degradation in HCMV-DB-infected MDMs and MRC5 fibroblasts. Cytoplasmic extracts were prepared from infected MDMs (D) and MRC5 fibroblasts (I) at 5 days postinfection and then assayed for the expression of phospho-IκBα and IκBα by Western blot. β-Actin was used as a loading control. E and J, IKKα, IKKβ, and IKKγ expression and phosphorylation in HCMV-DB-infected MDMs and MRC5 fibroblasts. Cytoplasmic extracts were prepared from infected MDMs (E) and MRC5 fibroblasts (J) at 5 days postinfection and then assayed for the corresponding protein by Western blot. β-Actin was used as a loading control.

In contrast to the NF-κB complexes observed in MDMs, anti-p50, anti-p65, and the combination of both Abs diminished the binding activity of NF-κB in HCMV-infected MRC5 fibroblasts (Fig. 3⇑G), suggesting the presence of canonical p50/p65 heterodimers in the activated NF-κB complex in HCMV-infected fibroblasts. Thus, NF-κB subunit binding activity was evaluated by microwell colorimetric NF-κB assay. Nuclear extracts from infected fibroblasts showed increased binding activity of p50 and p65 (Fig. 3⇑H), confirming the results obtained by EMSA.

In addition, expression of IκBα and a phosphorylated form of IκBα, which is required for IκBα degradation, was monitored by Western blot. We observed decreased phosphorylation of IκBα in infected MDMs, whereas the expression of IκBα increased (Fig. 3⇑D). In contrast, HCMV infection of fibroblasts resulted in phosphorylation and degradation of IκBα, as typically occurs during activation of the canonical NF-κB pathway (Fig. 3⇑I). Moreover, expression and phosphorylation of upstream IKKα, IKKβ, and IKKγ were monitored by Western blot.

Although there were no differences in the expression levels of IKKα (Fig. 3⇑, E and J), IKKα was highly phosphorylated in HCMV-infected MDMs (Fig. 3⇑E) as compared with MRC5 fibroblasts (Fig. 3⇑J). In contrast, phosphorylation of IKKβ was observed only in infected MRC5 fibroblasts (Fig. 3⇑, E and J). Phosphorylation of IKKγ was not observed in either cell type.

Bcl-3 expression in HCMV-infected MDMs and activation of MIEP by the p52/Bcl-3 complex

Bcl-3 has the potential to activate transcription after association with p52 and p50 (8, 9, 11); therefore, we wanted to evaluate the presence of p52/Bcl-3 complexes after HCMV infection of MDMs. We assayed Bcl-3 expression by MDMs and MRC5 fibroblasts after HCMV-DB and AD169 infection by Western blot and found that the amount of Bcl-3 was increased in nuclear extracts of infected MDMs but not in those of MRC5 fibroblasts (Fig. 4⇓A). In addition, p65 expression was observed early after HCMV infection of MDMs but not later on (Fig. 4⇓A), indicating a shift from p65 to Bcl-3 in MDMs. On the other hand, p65 was expressed both early and late during HCMV infection of MRC5 fibroblasts (Fig. 4⇓A). Quantification of Bcl-3 and p65 protein expression by densitometry showed that Bcl-3 expression and the Bcl-3/p65 ratio was much higher in infected MDMs than in infected MRC5 fibroblasts (Fig. 4⇓B). Next, we analyzed the interaction between Bcl-3 and p52 by coimmunoprecipitation assay. Total cellular extracts from uninfected and HCMV-DB-infected MDMs were immunoprecipitated with control IgG and anti-Bcl-3 Ab, followed by detection with anti-p52 Ab. The results revealed the specific interaction of Bcl-3 with p52 in HCMV-infected MDMs (Fig. 4⇓C).

FIGURE 4.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 4.

Bcl-3 up-regulation and binding with p52 in HCMV-infected MDMs. A, MDM and MRC5 fibroblast cultures were left uninfected or infected with the clinical isolate HCMV-DB and the laboratory strain AD169 at an MOI of 1. Nuclear extracts were prepared at different days postinfection and then assayed for the expression of Bcl-3 and p65 by Western blot. β-Actin was used as a loading control. B, Activation of Bcl-3 and p65 was quantified using ImageJ 1.40 software (National Institutes of Health) in uninfected and HCMV-DB-infected MDMs. Results are shown as relative intensities of Bcl-3 and the Bcl-3:p65 ratio. Means ± SD of two independent experiments are shown. C, Bcl-3 interacts with p52 in HCMV-infected MDM. Total cellular extracts from uninfected and HCMV-DB-infected MDMs were immunoprecipitated with anti-Bcl-3 and control Abs. Immunoprecipitated material was analyzed by Western blot using an anti-p52 Ab. The input control represents 1% of the material used for immunoprecipitation.

Next, we examined the ability of p52 and Bcl-3 to regulate transcription from HCMV MIEP by luciferase reporter assay using pCMV-Luc, in which the luciferase reporter gene is under the control of the MIEP of HCMV (Fig. 5⇓A). Promonocytic U937 cells were transfected with p52 plus Bcl3 or p50 plus p65-expressing plasmids along with pCMV-Luc; luciferase activity in cell extracts was measured 24 h posttransfection. We found that p52/Bcl-3 could activate transcription from MIEP (Fig. 5⇓B) but that the ability of p65 or the p50/p65 complex to drive transcription of the HCMV MIEP-luciferase reporter construct was many fold greater than that of p52/Bcl-3 (Fig. 5⇓C), which is in line with the low-level growth of the virus in MDMs vs efficient growth in fibroblasts. Next, we analyzed whether the MIEP was occupied by p52 and Bcl-3. The presence of p52 and Bcl-3 on the MIEP of HCMV-DB in acutely infected MDMs was demonstrated by ChIP using Abs specific for p52 and Bcl-3 (Fig. 6⇓, A and B). In addition, we assayed the binding of p50 and p65 with MIEP on days 1 and 5 postinfection. The results demonstrated that the canonical p50/p65 complex was activated early but not late after infection (Fig. 6⇓C). Finally, MDM and MRC5 fibroblast cultures were transfected with p52 plus Bcl-3 and p50 plus p65-specific siRNA and infected with HCMV-DB. Knockdown of p52, Bcl-3, p50, and p65 proteins in MDMs (Fig. 7⇓A) and fibroblasts (Fig. 7⇓B) was monitored by Western blot. Band density was quantified using ImageJ 1.40 software (National Institutes of Health), and the results are shown as relative intensities (Fig. 7⇓C). Knockdown of p50/p65 resulted in decreased growth of HCMV-DB in MDM cultures on day 1, whereas p52/Bcl-3 ablation resulted in decreased viral growth on days 4–6 postinfection (Fig. 8⇓A). In addition, p52/Bcl-3 knockdown did not block viral replication in MRC5 fibroblasts whereas p50/p65 ablation resulted in decreased growth of HCMV-DB in fibroblasts (Fig. 8⇓B). Cultures infected with AD169 showed similar results (data not shown). Moreover, overexpression of p52 and Bcl-3 in HCMV-infected MRC5 fibroblasts resulted in reduced viral replication (Fig. 8⇓C). Altogether, our results indicate that in HCMV-infected MDMs, HCMV MIEP is preferentially activated by a p52/Bcl-3 complex.

FIGURE 5.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 5.

Activation of gene expression from MIEP by NF-κB subunits. A, Schematic presentation of the luciferase reporter plasmid. Luciferase expression is under control of MIEP, which contains four NF-κB binding sites. B–C, U937 cells (2 × 106) were transfected with p52 plus Bcl3 (B) and p50 plus p65 (C) expression plasmids along with pCMV-Luc, which contains the luciferase gene under the control of MIEP of HCMV. Luciferase activity was measured in cell extracts at 24 h posttransfection. Data are expressed as relative light units (RLU) per microgram of protein. Controls were transfected with the corresponding empty vectors. Mean values ± SD of two independent experiments are shown; ∗, p < 0.05.

FIGURE 6.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 6.

Bcl-3 and p52 binding to MIEP in HCMV-infected MDMs. A, ChIP assay was performed with HCMV-DB-infected MDM cultures at 5 days postinfection. Briefly, cross-linked cell extracts from MDMs infected with HCMV-DB at an MOI of 1 were immunoprecipitated with control IgG, Bcl-3, and p52 Abs and assayed for the presence of MIEP by PCR. Amplification products were resolved by 2% agarose gel electrophoresis. Normal rabbit IgG was used as a nonspecific binding control. Input represents 1% of material used for immunoprecipitation. B, Enrichment of MIEP with p52 and Bcl-3 was measured by real-time PCR. Results represent mean values ± SD of two independent experiments. C, Enrichment of MIEP by p50 and p65 was measured by real-time PCR at day 1 and day 5 postinfection. Results represent mean values ± SD of two independent experiments.

FIGURE 7.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 7.

Knockdown of p52, Bcl-3, p50, and p65 proteins by siRNA. MDM (A) and MRC5 fibroblast (B) cultures were transfected with scrambled control, p52, Bcl-3, p50, or p65 siRNA, and total cellular extracts were prepared up to 3 days posttransfection. Protein expression was analyzed by Western blot; β-actin was used as a loading control. C, Protein levels after siRNA transfection were quantified by densitometry using ImageJ 1.40 software (National Institutes of Health), and the results are presented as relative intensities. Means ± SD of two independent experiments are shown.

FIGURE 8.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 8.

Effect of p52 plus Bcl-3 and p50 plus p65 siRNA on HCMV replication. MDM (A) and MRC5 fibroblast (B) cultures (0.5 × 106 cells) were transfected with a scrambled control, p52 plus Bcl-3 (top), or p50 plus p65 (bottom) siRNAs and infected with HCMV-DB at an MOI of 0.1. C, Inhibition of HCMV replication in p52 plus Bcl-3-transfected fibroblasts. p52, Bcl-3, and p52 plus Bcl-3 were overexpressed in MRC5 fibroblasts by transfection with the corresponding expression plasmid and infected with HCMV-DB at an MOI of 0.1. Viral titers in culture supernatants were determined at the indicated times postinfection by real-time PCR. An uninfected culture was tested in parallel. For each point, means ± SD of duplicate samples are shown; ∗, p < 0.05.

Discussion

The goal of this study was to assess the role of NF-κB in the transcriptional activity of the MIEP of HCMV in primary MDMs and fibroblasts. We observed low-level sustained growth of the virus and activation of p52/Bcl-3 complexes in MDMs. In contrast, the canonical p50/p65 heterodimer was the major component of activated NF-κB complexes in highly permissive MRC5 fibroblasts.

We observed that infection of MDMs with the clinical isolate HCMV-DB and the laboratory strain AD169 resulted in low-level sustained growth and concluded that HCMV was able to infect MDM cultures but that the viral titers in the culture supernatants were much lower than the viral titers of infected fibroblasts. Others have also reported that macrophages produce only low levels of HCMV (2, 6) and murine CMV (49). The efficiency of viral entry might be a contributing factor in HCMV cell tropism, but we observed that viral entry into MDMs and MRC5 fibroblasts was similar. Other reports have also demonstrated that the difference in viral growth kinetics between MDMs and fibroblasts was not due to differential attachment and adsorption of the virus (6, 50). Restricted replication in some cell types is believed to depend on a postentry block to viral gene expression (51). The inability of the macrophage culture to support high rates of viral replication might be dependent in part on the ability of the virus to inhibit macrophage differentiation, which is itself required for viral replication (52).

Moreover, our study demonstrates that significant signal transduction events occur after infection of MDMs by HCMV. There was an increase in NF-κB binding activity in nuclear extracts of MDMs and fibroblasts in response to HCMV infection. This finding is in line with findings from previous studies reporting activation of NF-κB in response to HCMV infection in fibroblasts (21, 22) and monocytes (23). The HCMV UL55 (gB) glycoprotein ligands have been reported to be initiators of the rapid activation of NF-κB in fibroblasts and monocytes (23). Our data support these findings but indicate differences in the composition of the NF-κB complexes between MDMs and fibroblasts.

Most importantly, our data show activation of p52/Bcl-3 complexes in MDMs after HCMV infection and activation of MIEP. Because p52 (NF-κB2) lacks a transactivation domain and the intrinsic ability to activate transcription, homodimers of p52 are poor transcriptional activators (7, 46) unless associated with Bcl-3 (9). Although this is the first report of regulation of the MIEP of HCMV by p52/Bcl-3 complexes, others have reported activation of a number of human genes by Bcl-3, including P-selectin (10), cyclin D1 (12, 53, 54, 55), Bcl-2 (11, 14, 56), inducible NO synthase (13), and epidermal growth factor receptor (15, 57) through NF-κB sites. Bcl-3 preferentially binds with p50 and p52 homodimers and activates transcription through ternary complex with nuclear coregulators such as JAB1, Tip60, and Bard1 (20). Therefore, the presence of p52/Bcl-3 complexes in infected MDMs represents differential regulation of NF-κB during HCMV infection. Although not fully characterized, histone deacetylases (HDAC) may be recruited by p52 (54) and Bcl-3 (16, 17, 18), indicating a possible role for HDACs in HCMV latency in macrophages. HDACs have been reported to mediate inhibition of MIEP activity in nonpermissive cells (58).

In contrast to the events that occur during activation of the canonical NF-κB pathway, we did not observe phosphorylation and degradation of IκBα in infected MDMs; instead, we found increased expression of IκBα in HCMV-infected MDMs. In fact, IκBα does not target p52 or p50; thus, p52 homodimers remain uninhibited (59). Our results agree with those from a previous study reporting that noncanonical NF-κB pathways are independent of IκBα degradation (56). In addition, increased IκBα mRNA expression in monocytes in response to HCMV infection has been reported (23), although this study did not report increased protein expression. Increased IκBα expression suggests a second mechanism for inhibiting p65-containing dimers in tandem with competition by p52 homodimers, indicating a double checkpoint to limit HCMV replication in primary MDMs. IKKα was more highly activated in infected MDMs than in infected MRC5 fibroblasts. Moreover, IKKβ activation was observed in infected MRC5 fibroblasts but not in infected MDMs, indicating activation of the canonical NF-κB pathway in MRC5 fibroblasts and the noncanonical NF-κB pathway in HCMV-infected MDMs in response to HCMV infection. In fact, activation of IKKβ is sufficient for phosphorylation of IκBα (60), whereas IKKα is required for p100 processing to p52 in an IκB-independent noncanonical NF-κB manner (61).

NF-κB activation in response to HCMV infection could consist of two steps: the early phase represents release of preformed stores of NF-κB in response to the binding of viral glycoproteins gB and gH with cell membrane receptors; whereas the second phase represents de novo synthesis of NF-κB proteins (25). Our results demonstrate the activation of p50/p65, expression of p65, and binding of p50 and p65 to MIEPs early after infection but not later in infection, underscoring a scenario in which the first phase of NF-κB activation in MDMs involves canonical p50/p65 complexes, leading to initiation of HCMV gene expression, as reported by others (23, 47, 48). Later during infection, there is a shift toward the involvement of p52 homodimer/Bcl-3 complexes, leading to sustained low-level growth of HCMV in MDMs. This hypothesis fits well with the kinetics of the growth curve of HCMV-DB in MDMs (Fig. 1⇑B) and with the inhibition of viral replication that occurred in the RNA interference experiments (Fig. 8⇑, A and B). Because NF-κB2 (62) and Bcl-3 (63) genes are regulated by NF-κB, the first phase of p50/p65 heterodimer NF-κB activation could contribute to the activation of p52/Bcl-3 complexes later during HCMV infection. It has already been reported that p52-containing complexes appear at later phases after an NF-κB-activating stimulus (64). Because IKKα has been reported to repress NF-κB activity by enhancing nuclear degradation of p65 in macrophages (65), the shift in the composition of NF-κB dimers during HCMV infection of macrophages might be dependent on IKKα. Another likely player is IL-10, because it is produced after HCMV infection of MDMs (unpublished observation) and can deactivate macrophages, thereby reducing viral growth. Some clues come from the ability of IL-10 to induce Bcl-3 (66) and the ability of CMV IL-10 to repress NF-κB activity through reduced degradation of IκBα (67).

The switch in NF-κB family members after HCMV infection of MDMs could result in at least two distinct phenomena. First, the NF-κB switch regulates the viral gene expression in infected MDMs, resulting in sustained low levels of viral replication and in viral persistence. In addition, HCMV-infected MDMs could fuel the progression of the disease by allowing the spread of the infection to more permissive cells such as fibroblasts and/or epithelial cells present in the vicinity of infected MDMs. Second, the NF-κB switch could modulate the expression of cellular genes in HCMV-infected MDMs resulting in an alternative cellular phenotype, e.g., an M2 phenotype. Altogether, the switch in NF-κB family members after HCMV infection of MDMs might influence the outcome of HCMV infection by modulating the expression of both viral and cellular genes.

The presence of different NF-κB complexes in primary MDMs and fibroblasts in response to HCMV infection led us to conclude that the p52 homodimer/Bcl-3 complexes in MDMs and p50/p65 heterodimers in fibroblasts could be responsible for the different levels of viral growth in MDMs vs fibroblasts (Fig. 9⇓). Moreover, our finding of differential regulation of NF-κB during HCMV infection of different cell types suggests further complexity and diversity in the role of NF-κB during HCMV infection. A better understanding of the mechanisms that underlie HCMV replication in monocytes/macrophages is likely to pave the way for new therapeutic approaches to control HCMV infection in this cell type.

FIGURE 9.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 9.

Model of HCMV MIEP transcription by NF-κB in macrophages and fibroblasts. HCMV infection of fibroblasts leads to activation of classical p50/p65 heterodimers, which are strong activators of MIEP transcription (A). HCMV infection of MDMs leads to activation of classical p50/p65 heterodimers, which are activators of MIEP transcription, early after infection (B). Later during infection, Bcl-3 is activated, and processing of p100 into p52 by IKKα results in the activation of p52/Bcl-3 complexes (C) but not of p50/p65 heterodimers (D). The association of Bcl-3 with p52 homodimers activates MIEP transcription at low levels. The presence of p52 homodimer/Bcl-3 complexes in MDMs vs p50/p65 heterodimers in fibroblasts could be responsible for the different levels of growth of HCMV in MDMs (low level) vs fibroblasts (efficient).

Acknowledgments

Viral strains TB40/E and TB40/F were provided by Dr. C. Sinzger, University of Tuebingen (Tuebingen, Germany).

Disclosures

The authors have no financial conflict of interest.

Footnotes

  • The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

  • ↵1 This work was supported by grants from Franche-Comté University (to G.H.). K.A.K. is a recipient of a doctoral scholarship from the Higher Education Commission, Pakistan. Results of these studies were presented in part at the 11th International CMV and Beta Herpesvirus Workshop, Toulouse France, May 13–17, 2007.

  • ↵2 Address correspondence and reprint requests to Dr. Georges Herbein, Department of Virology, Franche-Comté University, Hôpital Saint-Jacques, 2, place Saint-Jacques, F-25030 Besançon cedex, France. E-mail address: gherbein{at}chu-besancon.fr

  • ↵3 Abbreviations used in this paper: HCMV, human CMV; IKK, IκB kinase; AD169, high-passage laboratory strain AD169; ChIP, chromatin immunoprecipitation; MDM, primary human monocyte-derived macrophage; MIEP, major immediate-early promoter; MOI, multiplicity of infection; siRNA, small interfering RNA; EBNA, Epstein-Barr nuclear Ag; HDAC, histone deacetylase.

  • Received November 13, 2008.
  • Accepted April 2, 2009.
  • Copyright © 2009 by The American Association of Immunologists, Inc.

References

  1. ↵
    Landolfo, S., M. Gariglio, G. Gribaudo, D. Lembo. 2003. The human cytomegalovirus. Pharmacol. Ther. 98: 269-297.
    OpenUrlCrossRefPubMed
  2. ↵
    Sinzger, C., M. Digel, G. Jahn. 2008. Cytomegalovirus cell tropism. T. E. Shenk, and M. F. Stinski, eds. Human Cytomegalovirus 63-83. Springer-Verlag, Berlin-Heidelberg.
  3. ↵
    Michelson, S.. 1997. Interaction of human cytomegalovirus with monocytes/macrophages: a love-hate relationship. Pathol. Biol. (Paris) 45: 146-158.
    OpenUrlPubMed
  4. ↵
    Larsson, S., C. Soderberg-Naucler, F. Z. Wang, E. Moller. 1998. Cytomegalovirus DNA can be detected in peripheral blood mononuclear cells from all seropositive and most seronegative healthy blood donors over time. Transfusion 38: 271-278.
    OpenUrlCrossRefPubMed
  5. ↵
    Sinzger, C., B. Plachter, A. Grefte, T. H. The, G. Jahn. 1996. Tissue macrophages are infected by human cytomegalovirus in vivo. J. Infect. Dis. 173: 240-245.
    OpenUrlAbstract/FREE Full Text
  6. ↵
    Fish, K. N., A. S. Depto, A. V. Moses, W. Britt, J. A. Nelson. 1995. Growth kinetics of human cytomegalovirus are altered in monocyte-derived macrophages. J. Virol. 69: 3737-3743.
    OpenUrlAbstract/FREE Full Text
  7. ↵
    Hayden, M. S., S. Ghosh. 2004. Signaling to NF-κB. Genes Dev. 18: 2195-2224.
    OpenUrlAbstract/FREE Full Text
  8. ↵
    Bours, V., G. Franzoso, V. Azarenko, S. Park, T. Kanno, K. Brown, U. Siebenlist. 1993. The oncoprotein Bcl-3 directly transactivates through κB motifs via association with DNA-binding p50B homodimers. Cell 72: 729-739.
    OpenUrlCrossRefPubMed
  9. ↵
    Nolan, G. P., T. Fujita, K. Bhatia, C. Huppi, H. C. Liou, M. L. Scott, D. Baltimore. 1993. The bcl-3 proto-oncogene encodes a nuclear IκB-like molecule that preferentially interacts with NF-κB p50 and p52 in a phosphorylation-dependent manner. Mol. Cell Biol. 13: 3557-3566.
    OpenUrlAbstract/FREE Full Text
  10. ↵
    Pan, J., R. P. McEver. 1995. Regulation of the human P-selectin promoter by Bcl-3 and specific homodimeric members of the NF-κB/Rel family. J. Biol. Chem. 270: 23077-23083.
    OpenUrlAbstract/FREE Full Text
  11. ↵
    Viatour, P., M. Bentires-Alj, A. Chariot, V. Deregowski, L. de Leval, M. P. Merville, V. Bours. 2003. NF-κB2/p100 induces Bcl-2 expression. Leukemia 17: 1349-1356.
    OpenUrlCrossRefPubMed
  12. ↵
    Massoumi, R., K. Chmielarska, K. Hennecke, A. Pfeifer, R. Fassler. 2006. Cyld inhibits tumor cell proliferation by blocking Bcl-3-dependent NF-κB signaling. Cell 125: 665-677.
    OpenUrlCrossRefPubMed
  13. ↵
    Dai, R., R. A. Phillips, S. A. Ahmed. 2007. Despite inhibition of nuclear localization of NF-κB p65, c-Rel, and RelB, 17β-estradiol up-regulates NF-κB signaling in mouse splenocytes: the potential role of Bcl-3. J. Immunol. 179: 1776-1783.
    OpenUrlAbstract/FREE Full Text
  14. ↵
    Hovelmeyer, N., F. T. Wunderlich, R. Massoumi, C. G. Jakobsen, J. Song, M. A. Worns, C. Merkwirth, A. Kovalenko, M. Aumailley, D. Strand, et al 2007. Regulation of B cell homeostasis and activation by the tumor suppressor gene CYLD. J. Exp. Med. 204: 2615-2627.
    OpenUrlAbstract/FREE Full Text
  15. ↵
    Thornburg, N. J., N. Raab-Traub. 2007. Induction of epidermal growth factor receptor expression by Epstein-Barr virus latent membrane protein 1 C-terminal-activating region 1 is mediated by NF-κB p50 homodimer/Bcl-3 complexes. J. Virol. 81: 12954-12961.
    OpenUrlAbstract/FREE Full Text
  16. ↵
    Jamaluddin, M., S. Choudhary, S. Wang, A. Casola, R. Huda, R. P. Garofalo, S. Ray, A. R. Brasier. 2005. Respiratory syncytial virus-inducible BCL-3 expression antagonizes the STAT/IRF and NF-κB signaling pathways by inducing histone deacetylase 1 recruitment to the interleukin-8 promoter. J. Virol. 79: 15302-15313.
    OpenUrlAbstract/FREE Full Text
  17. ↵
    Hishiki, T., T. Ohshima, T. Ego, K. Shimotohno. 2007. BCL3 acts as a negative regulator of transcription from the human T-cell leukemia virus type 1 long terminal repeat through interactions with TORC3. J. Biol. Chem. 282: 28335-28343.
    OpenUrlAbstract/FREE Full Text
  18. ↵
    Wessells, J., M. Baer, H. A. Young, E. Claudio, K. Brown, U. Siebenlist, P. F. Johnson. 2004. BCL-3 and NF-κB p50 attenuate lipopolysaccharide-induced inflammatory responses in macrophages. J. Biol. Chem. 279: 49995-50003.
    OpenUrlAbstract/FREE Full Text
  19. ↵
    Viatour, P., M. P. Merville, V. Bours, A. Chariot. 2004. Protein phosphorylation as a key mechanism for the regulation of BCL-3 activity. Cell Cycle 3: 1498-1501.
    OpenUrlPubMed
  20. ↵
    Dechend, R., F. Hirano, K. Lehmann, V. Heissmeyer, S. Ansieau, F. G. Wulczyn, C. Scheidereit, A. Leutz. 1999. The Bcl-3 oncoprotein acts as a bridging factor between NF-κB/Rel and nuclear co-regulators. Oncogene 18: 3316-3323.
    OpenUrlCrossRefPubMed
  21. ↵
    Kowalik, T. F., B. Wing, J. S. Haskill, J. C. Azizkhan, A. S. Baldwin, Jr, E. S. Huang. 1993. Multiple mechanisms are implicated in the regulation of NF-κB activity during human cytomegalovirus infection. Proc. Natl. Acad. Sci. USA 90: 1107-1111.
    OpenUrlAbstract/FREE Full Text
  22. ↵
    Yurochko, A. D., T. F. Kowalik, S. M. Huong, E. S. Huang. 1995. Human cytomegalovirus upregulates NF-κB activity by transactivating the NF-κB p105/p50 and p65 promoters. J. Virol. 69: 5391-5400.
    OpenUrlAbstract/FREE Full Text
  23. ↵
    Yurochko, A. D., E. S. Huang. 1999. Human cytomegalovirus binding to human monocytes induces immunoregulatory gene expression. J. Immunol. 162: 4806-4816.
    OpenUrlAbstract/FREE Full Text
  24. ↵
    DeMeritt, I. B., L. E. Milford, A. D. Yurochko. 2004. Activation of the NF-κB pathway in human cytomegalovirus-infected cells is necessary for efficient transactivation of the major immediate-early promoter. J. Virol. 78: 4498-4507.
    OpenUrlAbstract/FREE Full Text
  25. ↵
    DeMeritt, I. B., J. P. Podduturi, A. M. Tilley, M. T. Nogalski, A. D. Yurochko. 2006. Prolonged activation of NF-κB by human cytomegalovirus promotes efficient viral replication and late gene expression. Virology 346: 15-31.
    OpenUrlCrossRefPubMed
  26. ↵
    Caposio, P., A. Luganini, G. Hahn, S. Landolfo, G. Gribaudo. 2007. Activation of the virus-induced IKK/NF-κB signalling axis is critical for the replication of human cytomegalovirus in quiescent cells. Cell Microbiol. 9: 2040-2054.
    OpenUrlCrossRefPubMed
  27. ↵
    Caposio, P., T. Musso, A. Luganini, H. Inoue, M. Gariglio, S. Landolfo, G. Gribaudo. 2007. Targeting the NF-κB pathway through pharmacological inhibition of IKK2 prevents human cytomegalovirus replication and virus-induced inflammatory response in infected endothelial cells. Antiviral Res. 73: 175-184.
    OpenUrlCrossRefPubMed
  28. ↵
    Eickhoff, J., M. Hanke, M. Stein-Gerlach, T. P. Kiang, K. Herzberger, P. Habenberger, S. Muller, B. Klebl, M. Marschall, T. Stamminger, M. Cotten. 2004. RICK activates a NF-κB-dependent anti-human cytomegalovirus response. J. Biol. Chem. 279: 9642-9652.
    OpenUrlAbstract/FREE Full Text
  29. ↵
    Gustems, M., E. Borst, C. A. Benedict, C. Perez, M. Messerle, P. Ghazal, A. Angulo. 2006. Regulation of the transcription and replication cycle of human cytomegalovirus is insensitive to genetic elimination of the cognate NF-κB binding sites in the enhancer. J. Virol. 80: 9899-9904.
    OpenUrlAbstract/FREE Full Text
  30. ↵
    Lernbecher, T., U. Muller, T. Wirth. 1993. Distinct NF-κB/Rel transcription factors are responsible for tissue-specific and inducible gene activation. Nature 365: 767-770.
    OpenUrlCrossRefPubMed
  31. ↵
    Beg, A. A., A. S. Baldwin, Jr. 1994. Activation of multiple NF-κB/Rel DNA-binding complexes by tumor necrosis factor. Oncogene 9: 1487-1492.
    OpenUrlPubMed
  32. ↵
    Lin, R., D. Gewert, J. Hiscott. 1995. Differential transcriptional activation in vitro by NF-κB/Rel proteins. J. Biol. Chem. 270: 3123-3131.
    OpenUrlAbstract/FREE Full Text
  33. ↵
    Saccani, S., S. Pantano, G. Natoli. 2003. Modulation of NF-κB activity by exchange of dimers. Mol Cell 11: 1563-1574.
    OpenUrlCrossRefPubMed
  34. ↵
    Prosch, S., R. Wuttke, D. H. Kruger, H. D. Volk. 2002. NF-κB: a potential therapeutic target for inhibition of human cytomegalovirus (re)activation?. Biol Chem. 383: 1601-1609.
    OpenUrlCrossRefPubMed
  35. ↵
    Herbein, G., U. Mahlknecht, F. Batliwalla, P. Gregersen, T. Pappas, J. Butler, W. A. O'Brien, E. Verdin. 1998. Apoptosis of CD8+ T cells is mediated by macrophages through interaction of HIV gp120 with chemokine receptor CXCR4. Nature 395: 189-194.
    OpenUrlCrossRefPubMed
  36. ↵
    Coaquette, A., A. Bourgeois, C. Dirand, A. Varin, W. Chen, G. Herbein. 2004. Mixed cytomegalovirus glycoprotein B genotypes in immunocompromised patients. Clin. Infect. Dis. 39: 155-161.
    OpenUrlAbstract/FREE Full Text
  37. ↵
    Murphy, E., D. Yu, J. Grimwood, J. Schmutz, M. Dickson, M. A. Jarvis, G. Hahn, J. A. Nelson, R. M. Myers, T. E. Shenk. 2003. Coding potential of laboratory and clinical strains of human cytomegalovirus. Proc. Natl. Acad. Sci. USA 100: 14976-14981.
    OpenUrlAbstract/FREE Full Text
  38. ↵
    Sinzger, C., K. Schmidt, J. Knapp, M. Kahl, R. Beck, J. Waldman, H. Hebart, H. Einsele, G. Jahn. 1999. Modification of human cytomegalovirus tropism through propagation in vitro is associated with changes in the viral genome. J. Gen. Virol. 80: (Pt. 11):2867-2877.
    OpenUrlAbstract/FREE Full Text
  39. ↵
    Arrode, G., C. Boccaccio, J. P. Abastado, C. Davrinche. 2002. Cross-presentation of human cytomegalovirus pp65 (UL83) to CD8+ T cells is regulated by virus-induced, soluble-mediator-dependent maturation of dendritic cells. J. Virol. 76: 142-150.
    OpenUrlAbstract/FREE Full Text
  40. ↵
    Varin, A., S. K. Manna, V. Quivy, A. Z. Decrion, C. Van Lint, G. Herbein, B. B. Aggarwal. 2003. Exogenous Nef protein activates NF-κB, AP-1, and c-Jun N-terminal kinase and stimulates HIV transcription in promonocytic cells: role in AIDS pathogenesis. J. Biol. Chem. 278: 2219-2227.
    OpenUrlAbstract/FREE Full Text
  41. ↵
    Davis, M. E., I. M. Grumbach, T. Fukai, A. Cutchins, D. G. Harrison. 2004. Shear stress regulates endothelial nitric-oxide synthase promoter activity through nuclear factor κB binding. J. Biol. Chem. 279: 163-168.
    OpenUrlAbstract/FREE Full Text
  42. ↵
    Renard, P., I. Ernest, A. Houbion, M. Art, H. Le Calvez, M. Raes, J. Remacle. 2001. Development of a sensitive multi-well colorimetric assay for active NFκB. Nucleic Acids Res. 29: E21
    OpenUrlCrossRefPubMed
  43. ↵
    Sainz, B., Jr, H. L. LaMarca, R. F. Garry, C. A. Morris. 2005. Synergistic inhibition of human cytomegalovirus replication by interferon-α/β and interferon-γ. Virol J. 2: 14
    OpenUrlCrossRefPubMed
  44. ↵
    Mahlknecht, U., J. Will, A. Varin, D. Hoelzer, G. Herbein. 2004. Histone deacetylase 3, a class I histone deacetylase, suppresses MAPK11-mediated activating transcription factor-2 activation and represses TNF gene expression. J. Immunol. 173: 3979-3990.
    OpenUrlAbstract/FREE Full Text
  45. ↵
    Sinzger, C., K. Eberhardt, Y. Cavignac, C. Weinstock, T. Kessler, G. Jahn, J. L. Davignon. 2006. Macrophage cultures are susceptible to lytic productive infection by endothelial-cell-propagated human cytomegalovirus strains and present viral IE1 protein to CD4+ T cells despite late downregulation of MHC class II molecules. J. Gen. Virol. 87: 1853-1862.
    OpenUrlAbstract/FREE Full Text
  46. ↵
    Chen, L. F., W. C. Greene. 2004. Shaping the nuclear action of NF-κB. Nat. Rev. Mol. Cell Biol. 5: 392-401.
    OpenUrlCrossRefPubMed
  47. ↵
    Chan, G., E. R. Bivins-Smith, M. S. Smith, A. D. Yurochko. 2008. Transcriptome analysis of NF-κB- and phosphatidylinositol 3-kinase-regulated genes in human cytomegalovirus-infected monocytes. J. Virol. 82: 1040-1046.
    OpenUrlAbstract/FREE Full Text
  48. ↵
    Smith, M. S., E. R. Bivins-Smith, A. M. Tilley, G. L. Bentz, G. Chan, J. Minard, A. D. Yurochko. 2007. Roles of phosphatidylinositol 3-kinase and NF-κB in human cytomegalovirus-mediated monocyte diapedesis and adhesion: strategy for viral persistence. J. Virol. 81: 7683-7694.
    OpenUrlAbstract/FREE Full Text
  49. ↵
    Hanson, L. K., A. N. Campbell. 2006. Determinants of macrophage tropism. M. J. Reddehase, Jr, ed. Cytomegaloviruses: Molecular Biology and Immunology 419-443. Caister Academic Press, Wymondham, Norfolk, U.K.
  50. ↵
    Ibanez, C. E., R. Schrier, P. Ghazal, C. Wiley, J. A. Nelson. 1991. Human cytomegalovirus productively infects primary differentiated macrophages. J. Virol. 65: 6581-6588.
    OpenUrlAbstract/FREE Full Text
  51. ↵
    Sinzger, C., M. Kahl, K. Laib, K. Klingel, P. Rieger, B. Plachter, G. Jahn. 2000. Tropism of human cytomegalovirus for endothelial cells is determined by a post-entry step dependent on efficient translocation to the nucleus. J. Gen. Virol. 81: 3021-3035.
    OpenUrlAbstract/FREE Full Text
  52. ↵
    Gredmark, S., T. Tilburgs, C. Soderberg-Naucler. 2004. Human cytomegalovirus inhibits cytokine-induced macrophage differentiation. J. Virol. 78: 10378-10389.
    OpenUrlAbstract/FREE Full Text
  53. ↵
    Westerheide, S. D., M. W. Mayo, V. Anest, J. L. Hanson, A. S. Baldwin, Jr. 2001. The putative oncoprotein Bcl-3 induces cyclin D1 to stimulate G(1) transition. Mol. Cell. Biol. 21: 8428-8436.
    OpenUrlAbstract/FREE Full Text
  54. ↵
    Rocha, S., A. M. Martin, D. W. Meek, N. D. Perkins. 2003. p53 represses cyclin D1 transcription through down regulation of Bcl-3 and inducing increased association of the p52 NF-κB subunit with histone deacetylase 1. Mol. Cell Biol. 23: 4713-4727.
    OpenUrlAbstract/FREE Full Text
  55. ↵
    Park, S. G., C. Chung, H. Kang, J. Y. Kim, G. Jung. 2006. Up-regulation of cyclin D1 by HBx is mediated by NF-κB2/BCL3 complex through κB site of cyclin D1 promoter. J. Biol. Chem. 281: 31770-31777.
    OpenUrlAbstract/FREE Full Text
  56. ↵
    Cristofanon, S., F. Morceau, A. I. Scovassi, M. Dicato, L. Ghibelli, M. Diederich. 2009. Oxidative, multistep activation of the noncanonical NF-κB pathway via disulfide Bcl-3/p50 complex. FASEB J. 23: 45-57.
    OpenUrlAbstract/FREE Full Text
  57. ↵
    Kung, C. P., N. Raab-Traub. 2008. Epstein-Barr virus latent membrane protein 1 induces expression of the epidermal growth factor receptor through effects on Bcl-3 and STAT3. J. Virol. 82: 5486-5493.
    OpenUrlAbstract/FREE Full Text
  58. ↵
    Murphy, J. C., W. Fischle, E. Verdin, J. H. Sinclair. 2002. Control of cytomegalovirus lytic gene expression by histone acetylation. EMBO J. 21: 1112-1120.
    OpenUrlAbstract
  59. ↵
    Fu, D., M. Kobayashi, L. Lin. 2004. A p105-based inhibitor broadly represses NF-κB activities. J. Biol. Chem. 279: 12819-12826.
    OpenUrlAbstract/FREE Full Text
  60. ↵
    Ghosh, S., M. S. Hayden. 2008. New regulators of NF-κB in inflammation. Nat. Rev. Immunol. 8: 837-848.
    OpenUrlCrossRefPubMed
  61. ↵
    Senftleben, U., Y. Cao, G. Xiao, F. R. Greten, G. Krahn, G. Bonizzi, Y. Chen, Y. Hu, A. Fong, S. C. Sun, M. Karin. 2001. Activation by IKKalpha of a second, evolutionary conserved, NF-κB signaling pathway. Science 293: 1495-1499.
    OpenUrlAbstract/FREE Full Text
  62. ↵
    Liptay, S., R. M. Schmid, E. G. Nabel, G. J. Nabel. 1994. Transcriptional regulation of NF-κB2: evidence for κB-mediated positive and negative autoregulation. Mol. Cell Biol. 14: 7695-7703.
    OpenUrlAbstract/FREE Full Text
  63. ↵
    Brasier, A. R., M. Lu, T. Hai, Y. Lu, I. Boldogh. 2001. NF-κB-inducible BCL-3 expression is an autoregulatory loop controlling nuclear p50/NF-κB1 residence. J. Biol. Chem. 276: 32080-32093.
    OpenUrlAbstract/FREE Full Text
  64. ↵
    Perkins, N. D.. 2003. Oncogenes, tumor suppressors and p52 NF-κB. Oncogene 22: 7553-7556.
    OpenUrlCrossRefPubMed
  65. ↵
    Lawrence, T., M. Bebien, G. Y. Liu, V. Nizet, M. Karin. 2005. IKKα limits macrophage NF-κB activation and contributes to the resolution of inflammation. Nature 434: 1138-1143.
    OpenUrlCrossRefPubMed
  66. ↵
    Kuwata, H., Y. Watanabe, H. Miyoshi, M. Yamamoto, T. Kaisho, K. Takeda, S. Akira. 2003. IL-10-inducible Bcl-3 negatively regulates LPS-induced TNF-α production in macrophages. Blood 102: 4123-4129.
    OpenUrlAbstract/FREE Full Text
  67. ↵
    Nachtwey, J., J. V. Spencer. 2008. HCMV IL-10 suppresses cytokine expression in monocytes through inhibition of nuclear factor-κB. Viral Immunol. 21: 477-482.
    OpenUrlCrossRefPubMed
PreviousNext
Back to top

In this issue

The Journal of Immunology: 182 (12)
The Journal of Immunology
Vol. 182, Issue 12
15 Jun 2009
  • Table of Contents
  • Table of Contents (PDF)
  • About the Cover
  • Advertising (PDF)
  • Back Matter (PDF)
  • Editorial Board (PDF)
  • Front Matter (PDF)
Print
Download PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for your interest in spreading the word about The Journal of Immunology.

NOTE: We only request your email address so that the person you are recommending the page to knows that you wanted them to see it, and that it is not junk mail. We do not capture any email address.

Enter multiple addresses on separate lines or separate them with commas.
Bcl-3-Regulated Transcription from Major Immediate-Early Promoter of Human Cytomegalovirus in Monocyte-Derived Macrophages
(Your Name) has forwarded a page to you from The Journal of Immunology
(Your Name) thought you would like to see this page from the The Journal of Immunology web site.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Citation Tools
Bcl-3-Regulated Transcription from Major Immediate-Early Promoter of Human Cytomegalovirus in Monocyte-Derived Macrophages
Kashif Aziz Khan, Alain Coaquette, Christian Davrinche, Georges Herbein
The Journal of Immunology June 15, 2009, 182 (12) 7784-7794; DOI: 10.4049/jimmunol.0803800

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Share
Bcl-3-Regulated Transcription from Major Immediate-Early Promoter of Human Cytomegalovirus in Monocyte-Derived Macrophages
Kashif Aziz Khan, Alain Coaquette, Christian Davrinche, Georges Herbein
The Journal of Immunology June 15, 2009, 182 (12) 7784-7794; DOI: 10.4049/jimmunol.0803800
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like

Jump to section

  • Article
    • Abstract
    • Materials and Methods
    • Results
    • Discussion
    • Acknowledgments
    • Disclosures
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • PDF

Related Articles

Cited By...

More in this TOC Section

  • Early Self-Regulatory Mechanisms Control the Magnitude of CD8+ T Cell Responses Against Liver Stages of Murine Malaria
  • Sublethal Hyperoxia Impairs Pulmonary Innate Immunity
  • Dependence of IL-4, IL-13, and Nematode-Induced Alterations in Murine Small Intestinal Smooth Muscle Contractility on Stat6 and Enteric Nerves
Show more HOST DEFENSE

Similar Articles

Navigate

  • Home
  • Current Issue
  • Next in The JI
  • Archive
  • Brief Reviews
  • Pillars of Immunology
  • Translating Immunology

For Authors

  • Submit a Manuscript
  • Instructions for Authors
  • About the Journal
  • Journal Policies
  • Editors

General Information

  • Advertisers
  • Subscribers
  • Rights and Permissions
  • Accessibility Statement
  • Public Access
  • Privacy Policy
  • Disclaimer

Journal Services

  • Email Alerts
  • RSS Feeds
  • ImmunoCasts
  • Twitter

Copyright © 2021 by The American Association of Immunologists, Inc.

Print ISSN 0022-1767        Online ISSN 1550-6606