Skip to main content
  • ASM
    • Antimicrobial Agents and Chemotherapy
    • Applied and Environmental Microbiology
    • Clinical Microbiology Reviews
    • Clinical and Vaccine Immunology
    • EcoSal Plus
    • Eukaryotic Cell
    • Infection and Immunity
    • Journal of Bacteriology
    • Journal of Clinical Microbiology
    • Journal of Microbiology & Biology Education
    • Journal of Virology
    • mBio
    • Microbiology and Molecular Biology Reviews
    • Microbiology Resource Announcements
    • Microbiology Spectrum
    • Molecular and Cellular Biology
    • mSphere
    • mSystems
  • Log in
  • My alerts
  • My Cart

Main menu

  • Home
  • Articles
    • Current Issue
    • Accepted Manuscripts
    • COVID-19 Special Collection
    • Archive
    • Minireviews
  • For Authors
    • Submit a Manuscript
    • Scope
    • Editorial Policy
    • Submission, Review, & Publication Processes
    • Organization and Format
    • Errata, Author Corrections, Retractions
    • Illustrations and Tables
    • Nomenclature
    • Abbreviations and Conventions
    • Publication Fees
    • Ethics Resources and Policies
  • About the Journal
    • About AAC
    • Editor in Chief
    • Editorial Board
    • For Reviewers
    • For the Media
    • For Librarians
    • For Advertisers
    • Alerts
    • AAC Podcast
    • RSS
    • FAQ
  • Subscribe
    • Members
    • Institutions
  • ASM
    • Antimicrobial Agents and Chemotherapy
    • Applied and Environmental Microbiology
    • Clinical Microbiology Reviews
    • Clinical and Vaccine Immunology
    • EcoSal Plus
    • Eukaryotic Cell
    • Infection and Immunity
    • Journal of Bacteriology
    • Journal of Clinical Microbiology
    • Journal of Microbiology & Biology Education
    • Journal of Virology
    • mBio
    • Microbiology and Molecular Biology Reviews
    • Microbiology Resource Announcements
    • Microbiology Spectrum
    • Molecular and Cellular Biology
    • mSphere
    • mSystems

User menu

  • Log in
  • My alerts
  • My Cart

Search

  • Advanced search
Antimicrobial Agents and Chemotherapy
publisher-logosite-logo

Advanced Search

  • Home
  • Articles
    • Current Issue
    • Accepted Manuscripts
    • COVID-19 Special Collection
    • Archive
    • Minireviews
  • For Authors
    • Submit a Manuscript
    • Scope
    • Editorial Policy
    • Submission, Review, & Publication Processes
    • Organization and Format
    • Errata, Author Corrections, Retractions
    • Illustrations and Tables
    • Nomenclature
    • Abbreviations and Conventions
    • Publication Fees
    • Ethics Resources and Policies
  • About the Journal
    • About AAC
    • Editor in Chief
    • Editorial Board
    • For Reviewers
    • For the Media
    • For Librarians
    • For Advertisers
    • Alerts
    • AAC Podcast
    • RSS
    • FAQ
  • Subscribe
    • Members
    • Institutions
Antiviral Agents

Novel Real-Time Monitoring System for Human Cytomegalovirus- Infected Cells In Vitro That Uses a Green Fluorescent Protein-PML-Expressing Cell Line

T. Ueno, Y. Eizuru, H. Katano, T. Kurata, T. Sata, S. Irie, K. Ogawa-Goto
T. Ueno
1Nippi Research Institute of Biomatrix, Adachi, Tokyo 120-8601, Japan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Y. Eizuru
3Division of Persistent & Oncogenic Viruses, Center for Chronic Viral Diseases, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8520, Japan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
H. Katano
2Department of Pathology, National Institute of Infectious Diseases, Shinjuku, Tokyo 162-8640, Japan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
T. Kurata
2Department of Pathology, National Institute of Infectious Diseases, Shinjuku, Tokyo 162-8640, Japan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
T. Sata
2Department of Pathology, National Institute of Infectious Diseases, Shinjuku, Tokyo 162-8640, Japan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
S. Irie
1Nippi Research Institute of Biomatrix, Adachi, Tokyo 120-8601, Japan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
K. Ogawa-Goto
1Nippi Research Institute of Biomatrix, Adachi, Tokyo 120-8601, Japan
2Department of Pathology, National Institute of Infectious Diseases, Shinjuku, Tokyo 162-8640, Japan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • For correspondence: kgoto@nippi-inc.co.jp
DOI: 10.1128/AAC.01641-05
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

ABSTRACT

Promyelocytic leukemia (PML) bodies are discrete nuclear foci that are intimately associated with many DNA viruses. In human cytomegalovirus (HCMV) infection, the IE1 (for “immediate-early 1”) protein has a marked effect on PML bodies via de-SUMOylation of PML protein. Here, we report a novel real-time monitoring system for HCMV-infected cells using a newly established cell line (SE/15) that stably expresses green fluorescent protein (GFP)-PML protein. In SE/15 cells, HCMV infection causes specific and efficient dispersion of GFP-PML bodies in an IE1-dependent manner, allowing the infected cells to be monitored by fluorescence microscopy without immunostaining. Since a specific change in the detergent solubility of GFP-PML occurs upon infection, the infected cells can be quantified by GFP fluorescence measurement after extraction. With this assay, the inhibitory effects of heparin and neutralizing antibodies were determined in small-scale cultures, indicating its usefulness for screening inhibitory reagents for laboratory virus strains. Furthermore, we established a sensitive imaging assay by counting the number of nuclei containing dispersed GFP-PML, which is applicable for titration of slow-growing clinical isolates. In all strains tested, the virus titers estimated by the GFP-PML imaging assay were well correlated with the plaque-forming cell numbers determined in human embryonic lung cells. Coculture of SE/15 cells and HCMV-infected fibroblasts permitted a rapid and reliable method for estimating the 50% inhibitory concentration values of drugs for clinical isolates in susceptibility testing. Taken together, these results demonstrate the development of a rapid, sensitive, quantitative, and specific detection system for HCMV-infected cells involving a simple procedure that can be used for titration of low-titer clinical isolates.

Human cytomegalovirus (HCMV) is a major opportunistic pathogen in two main patient groups, namely, AIDS patients and transplant recipients. Since HCMV dissemination in blood has a high predictive value for HCMV disease in immunocompromised hosts, several methods have been developed to detect and quantify HCMV in blood. Viral parameters currently available for monitoring HCMV infection include viremia, antigenemia, DNAemia, RNAemia, and circulating cytomegalic endothelial cells (reviewed in references 3, 4, 6, and 30). Over the last two decades, traditional tissue culture methods have been replaced by the shell vial culture assay, which provides quantitative data within 16 to 48 h by immunodetection of immediate-early (IE) antigen (5, 8, 11, 28). Although viremia estimated by the shell vial assay is a widespread approach for the rapid detection of HCMV in cell culture, its practical significance has been limited, mainly due to its relatively low sensitivity. The antigenemia assay is a rapid quantitative method that detects pp65-positive polymorphonuclear leukocytes by using monoclonal antibodies, and it is widely used to monitor HCMV infection (7, 12, 31). However, antigenemia is often not correlated with virus replication in vivo, and it has been suggested that concomitant determination of DNAemia and viremia is necessary. For the purpose of correctly monitoring HCMV parameters, the development of alternative strategies for rapid and reliable determination of HCMV infection remains a high priority.

Promyelocytic leukemia (PML) bodies, also known as ND10 (for “nuclear domain 10”), PODs (for “PML oncogenic domains”), and Kr bodies, are present in the nuclei of most mammalian cells as 10 to 20 spherical structures of 0.3 to 0.5 μm in size (20). PML bodies are composed of multiprotein complexes, including the PML, Sp100, NDP55, and Daxx proteins (15). Infection with DNA viruses, such as herpes simplex virus type 1 (HSV-1), HCMV, and adenoviruses, has a marked effect on PML bodies. HCMV deposits its genome adjacent to PML bodies and starts IE transcription, causing dispersion of the PML bodies mainly through the function of the IE1 protein (1, 2, 18). Transfection experiments have revealed that the IE1 protein transiently colocalizes with PML bodies and that both IE1 and PML subsequently become diffusely distributed in the nucleus mainly via de-SUMOylation of PML protein (16, 23).

In order to generate a simple monitoring system for HCMV-infected cells in vitro, we exploited a unique event observed in HCMV-infected cells, i.e., dispersion of PML bodies upon IE1 expression. For this purpose, we established a new cell line (SE/15) that stably expresses green fluorescent protein (GFP)-PML protein and the human host cell factor p180 (27). In the present study, we developed a new assay system for real-time detection and titration of HCMV using this cell line, which is applicable for screening anti-virus adsorption reagents. Furthermore, we established a simple, sensitive, and quantitative protocol for drug susceptibility testing of low-titer clinical isolates, most of which are cell-associated virus.

MATERIALS AND METHODS

Virus, cell culture, and plasmids.HCMV strains (Towne, Davis, and other clinical isolates) were propagated and titrated in human embryonic lung (HEL) fibroblasts as described previously (26). For cell-associated clinical isolates, the number of plaque-forming cells (PFCs) present in an infected culture was estimated according to previously described methods (19). The following clinically isolated virus strains were used: strains 01, 03, and 05 were isolated from patients with congenital HCMV infection; strains 02 and 04 were from patients with bone marrow transplantation and kidney transplantation (21), respectively; strain 06 was from a patient with lung transplantation whose clinical course will be described elsewhere; strains 07 and 08 were isolated from a patient with CMV retinitis and encephalitis, as reported previously (13, 32); and strain 09 was from a patient with AIDS (14).

The full-length cDNA of human p180 (27) was inserted into the pcDNA4 HisMax vector (Invitrogen) to generate pcDNAp180-54R. The expression plasmid for IE1 was described previously (17).

Establishment of the SE/15 cell line.Chinese hamster ovary (CHO) cells have been reported to be as sensitive to HCMV as human fibroblasts regarding initial entry and IE1 expression (25). Since HCMV infection frequently caused detachment of CHO cells and this detachment was efficiently avoided by human p180 overexpression (unpublished observation), we generated new cell lines coexpressing GFP-PML and the human host cell factor p180 (27) by transfection of CHO/GFP-PML cells (17) with pcDNAp180-54R using Fugene 6. Cells were selected with 100 μg/ml zeocin and 100 μg/ml G418. Coexpression of GFP-PML and p180 was confirmed by immunofluorescence microscopy and Western blotting (not shown). Transfectants containing abnormally large GFP-PML structures appeared to exhibit difficulties in keeping the structures stably; thus, we carried out limited dilution to carefully select a suitable clone with respect to the stability and morphogenesis of the GFP-PML bodies, and we finally cloned a cell line, SE/15, and used these cells throughout this study.

Inoculation of SE/15 cells.SE/15 cells were seeded in 24- or 96-well tissue culture plates precoated with 10 μg/ml fibronectin (GIBCO/BRL) at a cell density of 1 × 105 or 0.2 × 105 cells/well, respectively. After 24 h of culture, HCMV was inoculated at a multiplicity of infection (MOI) of 3 and the cells were incubated in F12-Dulbecco modified Eagle medium containing 5% fetal bovine serum (FBS) for 18 h at 37°C unless otherwise described.

Quantification of infected SE/15 cells with an NPB fluorescence assay.At various times postinfection (p.i.), HCMV- or mock-infected SE/15 cells were washed with phosphate-buffered saline (PBS) and gently extracted with 250 or 30 μl of NPB (50 mM Tris-HCl buffer containing 0.15% NP-40, 150 mM NaCl, 15 mM MgCl2, and 5 mM EDTA) for 24- or 96-well plates, respectively, at 4°C for 15 to 20 min. If necessary, the solutions were centrifuged briefly and the supernatants were transferred to a μClear Fluorescence Black P 96-well plate (Greiner). The fluorescence intensity was measured with a CytoFluor multiwell plate reader (series 4000; Perseptive Biosystems) with an excitation wavelength of 485 nm and an emission wavelength of 530 nm. HCMV infectivity was expressed as the ratio of the net fluorescence intensity of the NPB extract to the fluorescence intensity of the total cell lysate prior to extraction, where the net fluorescence intensity was the NPB fluorescence intensity of infected cells after subtraction of the NPB fluorescence intensity of mock-infected cells. The total cell lysates prior to extraction were prepared in a parallel experiment by scraping of cells, followed by lysis with NPB in a tube.

Quantification of infected cell numbers by a GFP-PML imaging assay.For measurement of infected cell numbers, fluorescence signals were captured with an Olympus fluorescence microscope (model IX71) equipped with a cooled charge-coupled-device camera (DP50; Olympus) using a fixed data collection time of 0.2 s. Imaging analysis was performed with the public-domain software Image J (http://rsb.info.nih.gov/ij ) to obtain a series of data sets of particle numbers of each particle size. To evaluate the total cell numbers of uninfected SE/15 cells, a correlation coefficient was calculated from the numbers of GFP-PML dots and DAPI (4′,6′-diamidino-2-phenylindole)-stained nuclei. Under these conditions, GFP-PML dots that were too small were not counted, to avoid capturing signals that were unable to be distinguished from electrical noise, and hence the mean number of GFP-PML dots/nucleus was estimated to be less than 2. In the present study, we used the following formula for evaluating the total number of cells bearing GFP-PML bodies: total number of cells = 0.632 × total number of GFP-PML dots. HCMV infectivity was expressed as the ratio of the number of nuclei possessing diffuse GFP-PML to the total number of SE/15 cells harboring GFP-PML.

Susceptibility tests against antiviral agents.The susceptibilities to antiviral compounds were measured by the following methods.

(i) NPB fluorescence assay.For heparin inhibition assays, HCMV (Towne) was pretreated with heparin (0 to 200 μg/ml) for 1 h at 37°C and inoculated into SE/15 cells. For antibody neutralization assays, HCMV (Towne) was pretreated with monoclonal antibodies (MAbs) against HCMV gB or gH (Goodwin Institute), or a control mouse immunoglobulin G (IgG), at a final concentration of 25 or 100 μg/ml in the absence of complement for 1 h at 37°C. Quantification of the NPB fluorescence intensity was performed according to the standard protocol described above. Infectivity was expressed as the percentage of infectivity at each concentration of a drug to that in the absence of the same drug.

(ii) Coculture of clinical isolates and GFP-PML imaging assays.Since SE/15 cells do not support a full replication cycle of HCMV, we performed coculturing of SE/15 cells with infected HEL cells for susceptibility tests. HCMV clinical isolates were cultured in HEL cells in 24-well plates (1 × 105 cells/well) in the presence of various concentrations of antiviral drugs for 48 h. The infected cells were trypsinized, briefly centrifuged, and mixed with a suspension of SE/15 cells. The ratio of cocultured, infected HEL cells to SE/15 cells was determined to be 1:2 through our preliminary study to define the optimum results. Next, the cells were seeded in triplicate onto fibronectin-precoated μClear Fluorescence Black P 96-well plates. After another 48 h of incubation in the presence of various concentrations of antiviral drugs, infectivity was measured with a GFP-PML imaging assay. (For a schematic diagram of the protocol, see Fig. 6A.) Curve fitting and 50% inhibitory concentration (IC50) calculations were performed with the PRISM4 program (GraphPad Inc.).

(iii) Plaque reduction assay.HEL monolayers in 60-mm dishes were infected with HCMV by inoculating approximately 100 to 200 PFU of each virus inoculum. The cells were then overlaid with medium containing 1% methylcellulose, 2% FBS, and various concentrations of drugs. Assay cultures were incubated for 10 to 14 days, and the plaques were counted after fixation with 5% formalin in PBS and staining with 0.03% methylene blue. Triplicate samples were prepared for each drug concentration.

Antibody and immunofluorescence microscopy.The following MAbs were used: anti-HCMV IE1 (MAB810; Chemicon); anti-PML (PG-M3; Santa Cruz Biotechnology); and anti-calnexin (Transduction Laboratories). Normal mouse IgG was obtained from Santa Cruz Biotechnology. The cells were fixed with paraformaldehyde, permeabilized with 0.1% Triton X-100, and then incubated with the antibodies as described previously (27). Alexa Fluor 568-conjugated goat anti-mouse IgG was used as secondary antibody. Cells were imaged with a Zeiss LSM410 confocal microscopy system.

RESULTS

SE/15 cells have functional PML bodies labeled with GFP.To generate a simple monitoring system for HCMV-infected cells in vitro, we have cloned a CHO cell line, SE/15, stably expressing both GFP-PML protein and p180 (see Materials and Methods). SE/15 cells contained a number of small nuclear foci labeled with GFP (Fig. 1A) that were also costained for PML (data not shown). Upon HCMV infection, infected cells positive for IE1 exhibited a diffuse GFP pattern throughout the nucleus (Fig. 1B), the intensity of which appeared to be higher than that of IE1 (compare panels B1 and B2). The unique appearance of nuclei containing dispersed GFP-PML, which was never observed in mock-infected cells, began at 3 to 4 h p.i., and the number of these nuclei continued to increase until 10 to 16 h p.i. These data indicate that SE/15 cells have functional GFP-PML bodies whose morphology is specifically changed upon HCMV infection. Thus, visual assessment under fluorescence microscopy is sufficient for sensitive detection of infected cells without the necessity for any further procedures such as fixation and staining. To verify the specificity of HCMV-induced GFP-PML dispersion, HSV-1 (strain F) was inoculated onto SE/15 cells. No morphological change of GFP-PML caused by the virus was observed, mainly because SE/15 cells do not support initial entry of HSV-1 (data not shown). To establish a quantification method for infected SE/15 cells, we applied two strategies, namely, measurement of the GFP fluorescence intensity and direct counting of the infected nuclei by imaging analysis.

FIG. 1.
  • Open in new tab
  • Download powerpoint
FIG. 1.

Fluorescence microscopic images of SE/15 cells showing aberrant localization of GFP-PML in HCMV-infected cells. SE/15 cells were mock-infected (A1 to A3) or infected with Towne (MOI = 3) (B1 to B3), followed by staining for IE1 at 18 h p.i. (A1 and B1) GFP signals. (A2 and B2) IE1 staining. (A3 and B3) Merged images. Bars, 25 μm.

Diffuse GFP-PML in HCMV-infected cells is efficiently extracted with NPB.In normal cells, it has been reported that PML protein is present as an insoluble form in the nucleus, probably as multiprotein complexes anchored to the nuclear matrix, and that SUMOylation of PML protein is required for this distribution (23, 34). Since de-SUMOylation of PML protein occurs in HCMV-infected cells (22), it is supposed that the diffuse PML in the infected nuclei would be soluble in NP-40 buffer. To address this idea, the cells were gently treated with NPB. As expected, most of the GFP signals in the HCMV-infected nuclei disappeared after NPB treatment under fluorescence microscopy, while the small GFP-PML dots in mock-infected cells remained unchanged (Fig. 2A). Western blotting analyses confirmed effective extraction of GFP-PML into NPB from the infected cells but not from mock-infected cells (Fig. 2B). Thus, GFP-PML protein in SE/15 cells retains characteristics similar to PML protein expressed in HCMV-infected fibroblasts. Identical results were obtained after IE1 transfection, instead of HCMV infection, confirming that expression of IE1 alone is sufficient to induce this change (data not shown). These data indicate that NPB extraction-based fluorescence measurements in SE/15 cells allow quantification of HCMV-infected cells.

FIG. 2.
  • Open in new tab
  • Download powerpoint
FIG. 2.

GFP-PML is efficiently extracted into NPB following HCMV infection. SE/15 cells were mock-infected or infected with HCMV as described in the legend to Fig. 1, followed by mild extraction by NPB, which permeabilized the cells but did not lyse them. (A) Fluorescence microscopic images of GFP before (a and b) and after (c and d) NPB treatment are shown for mock-infected (a and c) and HCMV-infected (b and d) cells. (B) Western blotting data of total cell lysates (lanes 1 and 2) and NPB extracts (lanes 3 and 4) from HCMV-infected (lanes 1 and 3) and mock-infected (lanes 2 and 4) cells are shown, respectively. Calnexin was used as a loading control.

Quantification of HCMV-infected cells by GFP fluorescence after NPB extraction.To establish a sensitive assay, we first optimized the conditions for extraction. Through systematic screening of various conditions, the optimum conditions were determined to be as follows. At various times p.i., mock- and HCMV-infected cells were washed with PBS and gently extracted with 50 mM Tris-HCl buffer (pH 7.4) containing 0.15% NP-40, 150 mM NaCl, 15 mM MgCl2, and 5 mM EDTA at 4°C for 20 min. The HCMV infectivity was expressed as the ratio of the net fluorescence intensity of the NPB extract to the fluorescence intensity of the total cell lysate, where the net fluorescence intensity was the NPB fluorescence intensity of infected cells after subtraction of the NPB fluorescence intensity of mock-infected cells. Due to the distinct background fluorescence of mock-infected cells, which probably arises through cell debris and non-GFP fluorescent background, subtraction was necessary for accurate estimations. By this protocol, infected cells at various times p.i. were estimated as shown in Fig. 3A. Consistent with the microscopic observations, the net NPB fluorescence was detectable as early as 4 h p.i., increased until 10 to 12 h p.i., and then almost reached a plateau. The correlation with the virus titer was tested with serially diluted HCMV inoculums in 12- or 96-well plates (Fig. 3B), and the results revealed a good correlation between the NPB fluorescence and the input virus over a diverse range. On the 12-well plate scale, HCMV-infected cells at an MOI of 0.4 were measurable. The NPB fluorescence was well correlated with IE1 expression levels in blotting analyses (Fig. 3C).

FIG. 3.
  • Open in new tab
  • Download powerpoint
FIG. 3.

Correlation of the NPB fluorescence assay with IE1 expression levels. (A) SE/15 cells were infected with Towne (MOI = 3) in a 24-well plate. At various times p.i., the standard NPB fluorescence assay was performed as described in the text. (B) SE/15 cells were infected, as described for panel A, in a 96- or 12-well plate at a MOI of 0.001 to 10. At 18 h p.i., the standard NPB fluorescence assay was performed. (C) Western blot analysis of total cell lysates from the experiment shown in panel B. Data are means ± standard deviations from three experiments.

Application of the NPB fluorescence assay to inhibition experiments with antiviral agents.Next, we tested whether our assay, termed the NPB fluorescence assay, can be applied to determination of the IC50 values of heparin in inhibition experiments. As shown in Fig. 4A, HCMV infectivity estimated by the NPB fluorescence assay was well fitted with a sigmoid curve, and the IC50 was estimated to be about 10 μg/ml, similar to previously reported values (24). Again Western blotting analysis revealed that the net fluorescence intensity of NPB extracts was well correlated with the expression levels of IE1 (Fig. 4B). Furthermore, the neutralizing abilities of MAbs against HCMV gB or gH were evaluated in the absence of complement (Fig. 4C). The assay was also feasible in 96-well plates (data not shown). These data suggest that our NPB fluorescence assay provides a simple and rapid procedure for evaluating inhibitors of the initial replication phase without the use of any immunological reagents within 10 to 16 h.

FIG. 4.
  • Open in new tab
  • Download powerpoint
FIG. 4.

The NPB fluorescence assay is applicable for determining the effects of infectivity inhibitors. (A and B) HCMV was preincubated for 1 h at 37°C with various concentrations of heparin, an entry process inhibitor of the virion into the cells. SE/15 cells were infected (MOI = 1) in a 24-well plate for 18 h, followed by the standard NPB fluorescence assay as described in the text. (C) HCMV was preincubated with a MAb against gB or gH for 1 h at 37°C and assayed as described for panel A. Data are means ± standard deviations from three experiments. Asterisks indicate a statistically significant difference between HCMV- and mock-infected cells by Student's t test (**, P < 0.01; *,P< 0.05).

Quantification of HCMV-infected cells with a GFP-PML imaging assay.Next, we investigated the application of our system to drug susceptibility testing of low-titer HCMV clinical isolates. Since our preliminary study showed that a higher sensitivity was required for assaying clinical isolates, we developed an imaging assay for this purpose rather than using the NPB fluorescence assay.

Since GFP-PML is dispersed throughout the nuclei of HCMV-infected SE/15 cells and the GFP-PML areas are markedly larger than the GFP-PML dots of uninfected cells (Fig. 1 or 2), imaging analysis of fluorescence micrographs enables direct counting of the number of infected nuclei. A typical profiling pattern by particle size of mock-infected cells gave one peak representing the small GFP-PML foci (Fig. 5A, left). The total number of SE/15 cell nuclei was evaluated from the total number of GFP dots by using this pattern, as described in Materials and Methods. In contrast, a second broad peak appeared in the profiling pattern of HCMV-infected cells, corresponding to the sizes of nuclei bearing a diffuse GFP signal (Fig. 5A, right). Therefore, HCMV infectivity was expressed as the ratio of the number of nuclei possessing a diffuse GFP signal to the total number of SE/15 cell nuclei. Using this procedure, termed the GFP-PML imaging assay, we titrated various HCMV strains, including clinical isolates, and compared the results with the number of PFCs determined in HEL cells. Figure 5B shows representative graphs of the well-correlated relationship observed between the infectivity estimated by the GFP-PML imaging assay and the numbers of PFCs, thereby confirming the feasibility of the GFP-PML imaging assay. For all of the strains tested, the correlation coefficients were >0.90.

FIG. 5.
  • Open in new tab
  • Download powerpoint
FIG. 5.

HCMV infectivity estimated by the GFP-PML imaging assay is correlated with the number of PFCs. (A) Profile patterns of the particle sizes of mock-infected (left) and Towne-infected (right) cells. (B) Typical graphs showing the correlation between the HCMV infectivity of clinical isolates estimated by the GFP-PML imaging assay and the number of PFCs determined in HEL cells. Data are means ± standard deviations from three experiments.

Application of the GFP-PML imaging assay to drug susceptibility testing of clinical isolates.We performed drug susceptibility tests of various HCMV strains by using the GFP-PML imaging assay. Since SE/15 cells do not support a full replication cycle, we cocultured SE/15 cells with infected HEL cells which had been cultured in the presence or absence of antiviral agents for 48 h prior to coculturing (Fig. 6A). At 96 h p.i., the HCMV infectivities at various concentrations of drugs were measured to evaluate the IC50 values from their inhibition curves. Figure 6B shows representative graphs of a cell-associated laboratory strain (Davis) and a clinical isolate (01) for ganciclovir (GCV) or foscarnet (FOS). Subsequently, we compared the IC50 values obtained for the nine clinical isolates (Table 1), including four GCV-resistant strains, with the values obtained with a conventional plaque reduction assay. With the GFP-PML imaging assay, the IC50 values of GCV for the four resistant viruses were estimated to be >9 μM, which were higher than those of the sensitive strains (<5 μM). In contrast, the IC50 values of FOS for all the viruses were estimated to be <200 μM. The IC50 values determined by the GFP-PML imaging assay were relatively higher but still remained comparable to those obtained with the plaque reduction assay. These results suggest that the GFP-PML imaging assay is sensitive enough to distinguish GCV-resistant viruses from GCV-sensitive viruses. The IC50 values of these isolates were also determined by a standard plaque reduction assay using cell-free viruses, and the results correlated well with the data from the GFP-PML imaging assay (Table 1).

FIG. 6.
  • Open in new tab
  • Download powerpoint
FIG. 6.

Drug susceptibility testing of clinical isolates by the GFP-PML imaging assay. (A) Schematic representation of the protocol for drug susceptibility testing of clinical isolates with the coculture system. (B) Data for drug susceptibility testing of the Davis strain and clinical isolate #01 for GCV and FOS using cell-associated viruses. Data are means ± standard deviations from three experiments.

View this table:
  • View inline
  • View popup
TABLE 1.

Clinical diagnoses of patients and results of drug susceptibility testing

DISCUSSION

Despite recent advances in antiviral therapies, CMV infection remains an important opportunistic infection that causes serious life-threatening problems, especially in patients undergoing bone marrow and solid organ transplantation. The extensive use of antiviral compounds for prophylaxis and treatment of HCMV infection has led to the emergence of drug-resistant HCMV strains (10). Although rapid and accurate determination of drug susceptibility is important and necessary for patient management, conventional phenotypic drug susceptibility tests for clinical HCMV isolates are usually very laborious and can require long periods of time. In contrast, genetic assays of drug resistance-associated mutations in the UL97 and UL54 genes can greatly shorten the time required for the identification of drug-resistant strains. However, such genotypic analyses are limited to known mutations that confer resistance, and identification of HCMV strains with specific mutations in target genes provides only indirect evidence of drug resistance. Therefore, phenotypic determination of drug susceptibility remains important and necessary, and the development of rapid determination methods for HCMV infection is required. Many groups have reported improvements in phenotypic testing for drug resistance on the basis of plaque reduction assays coupled with detection by MAbs against IE1 (9, 19, 29, 33).

Here, we have presented a novel real-time detection system for HCMV-infected cells in vitro using SE/15 cells that stably express GFP-PML. In the nuclei of these cells, HCMV infection causes specific and efficient dispersion of the GFP-PML bodies in an IE1-dependent manner, thereby enabling real-time monitoring of infected cells without immunostaining. We have further demonstrated quantitative protocols for HCMV detection using either an NPB fluorescence assay or a GFP-PML imaging assay. The former assay is rapid, sensitive, and sufficiently simple for the performance of multiple or high-throughput screening of inhibitory reagents, particularly inhibitory drugs or neutralizing antibodies, using laboratory strains from which cell-free viruses are easily prepared. Indeed, the inhibitory effects of heparin and a neutralizing antibody for Towne were measurable within 16 h in cultures at the 96-well microplate level. The infectivities determined by the assays were well correlated with the IE1 expression levels, suggesting that these assays are based on indirect evaluation of IE1 without immunostaining. On the other hand, the GFP-PML imaging assay coupled with a coculture system permits highly sensitive and specific detection of infected cells and is applicable to drug susceptibility testing of slow-growing cell-associated clinical isolates, most of which are cell-associated viruses.

In all of the strains tested, the virus titers estimated by the GFP-PML imaging assay were well correlated with the numbers of PFCs determined in HEL cells. Although the IC50 cutoff values in our assay of individual drugs remain to be determined with a large number of virus strains, the value for distinguishing GCV-resistant viruses from GCV-sensitive viruses was tentatively assumed to be about 5 μM, which is comparable to the reported value for plaque reduction assays (19). The IC50 values determined by our assay are slightly higher than those obtained with plaque reduction assays. This may arise through a potential underestimation of the infectivity introduced by our protocol, thereby resulting in higher IC50 values. Since it is assumed that a few HEL cells can infect a large number of SE/15 cells during cocultivation, a moderate but still significant reduction in virus replication mediated by the drug in HEL cells may not be detected in SE/15 cells. If rapidly growing strains are used, this potential underestimation becomes more prominent. Indeed, such a phenomenon was observed in strain #08, which grows very rapidly in vitro.

HCMV viremia detection by the shell vial assay is a widespread approach to rapid virus identification in cell cultures based on the use of a MAb against IE1. However, its practical significance has been limited by its relatively low sensitivity and somewhat cumbersome procedure. These disadvantages of the shell vial assay may be improved by our monitoring system using SE/15 cells, and a trial assessing its feasibility is now being undertaken.

One of the inherent difficulties in working with HCMV is visual assessment of its cytopathic effects in HEL cells when virus titers are being determined. Regarding this point, our system has a strong advantage because the infected cells are clearly distinguishable from uninfected cells by fluorescence microscopy without immunostaining. Future improvements to our system will include the generation (or subcloning) of suitable cell lines that retain GFP-PML protein under more stable conditions with a higher incidence, although SE/15 cells can be utilized for over 25 passages for the purpose of GFP-PML imaging assays (unpublished data). The generation of a similar cell line harboring full permissiveness to HCMV should be attempted, although our preliminary trials seeking such cells have repeatedly failed, probably due to the growth disadvantage of these transfectants.

In conclusion, we have demonstrated a rapid, sensitive, quantitative, and specific detection system for HCMV-infected cells involving a simple procedure that is applicable for titrating low-titer clinical isolates. Our established procedure, including coculturing of SE/15 and HCMV-infected HEL cells, should also provide a rapid and reliable tool for drug susceptibility testing.

ACKNOWLEDGMENTS

This study was supported in part by a Grant-in-Aid for Research on Sensory and Communicative Disorders from the Ministry of Health and Welfare, Japan.

FOOTNOTES

    • Received 27 December 2005.
    • Returned for modification 2 March 2006.
    • Accepted 4 May 2006.
  • Copyright © 2006 American Society for Microbiology

REFERENCES

  1. 1.↵
    Ahn, J. H., E. J. Brignole III, and G. S. Hayward. 1998. Disruption of PML subnuclear domains by the acidic IE1 protein of human cytomegalovirus is mediated through interaction with PML and may modulate a RING finger-dependent cryptic transactivator function of PML. Mol. Cell. Biol.18:4899-4913.
    OpenUrlAbstract/FREE Full Text
  2. 2.↵
    Ahn, J. H., and G. S. Hayward. 1997. The major immediate-early proteins IE1 and IE2 of human cytomegalovirus colocalize with and disrupt PML-associated nuclear bodies at very early times in infected permissive cells. J. Virol.71:4599-4613.
    OpenUrlAbstract/FREE Full Text
  3. 3.↵
    Baldanti, F., and G. Gerna. 2003. Human cytomegalovirus resistance to antiviral drugs: diagnosis, monitoring and clinical impact. J. Antimicrob. Chemother.52:324-330.
    OpenUrlCrossRefPubMedWeb of Science
  4. 4.↵
    Boeckh, M., and G. Boivin. 1998. Quantitation of cytomegalovirus: methodologic aspects and clinical applications. Clin. Microbiol. Rev.11:533-554.
    OpenUrlAbstract/FREE Full Text
  5. 5.↵
    Chou, S. W., and K. M. Scott. 1988. Rapid quantitation of cytomegalovirus and assay of neutralizing antibody by using monoclonal antibody to the major immediate-early viral protein. J. Clin. Microbiol.26:504-507.
    OpenUrlAbstract/FREE Full Text
  6. 6.↵
    Drew, W. L., C. V. Paya, and V. Emery. 2001. Cytomegalovirus (CMV) resistance to antivirals. Am. J. Transplant.1:307-312.
    OpenUrlCrossRefPubMedWeb of Science
  7. 7.↵
    Gerna, G., M. G. Revello, E. Percivalle, and F. Morini. 1992. Comparison of different immunostaining techniques and monoclonal antibodies to the lower matrix phosphoprotein (pp65) for optimal quantitation of human cytomegalovirus antigenemia. J. Clin. Microbiol.30:1232-1237.
    OpenUrlAbstract/FREE Full Text
  8. 8.↵
    Gerna, G., M. G. Revello, E. Percivalle, M. Zavattoni, M. Parea, and M. Battaglia. 1990. Quantification of human cytomegalovirus viremia by using monoclonal antibodies to different viral proteins. J. Clin. Microbiol.28:2681-2688.
    OpenUrlAbstract/FREE Full Text
  9. 9.↵
    Gerna, G., A. Sarasini, E. Percivalle, M. Zavattoni, F. Baldanti, and M. G. Revello. 1995. Rapid screening for resistance to ganciclovir and foscarnet of primary isolates of human cytomegalovirus from culture-positive blood samples. J. Clin. Microbiol.33:738-741.
    OpenUrlAbstract/FREE Full Text
  10. 10.↵
    Gilbert, C., and G. Boivin. 2005. Human cytomegalovirus resistance to antiviral drugs. Antimicrob. Agents Chemother.49:873-883.
    OpenUrlFREE Full Text
  11. 11.↵
    Gleaves, C. A., T. F. Smith, E. A. Shuster, and G. R. Pearson. 1984. Rapid detection of cytomegalovirus in MRC-5 cells inoculated with urine specimens by using low-speed centrifugation and monoclonal antibody to an early antigen. J. Clin. Microbiol.19:917-919.
    OpenUrlAbstract/FREE Full Text
  12. 12.↵
    Grefte, J. M., B. T. van der Gun, S. Schmolke, M. van der Giessen, W. J. van Son, B. Plachter, G. Jahn, and T. H. The. 1992. The lower matrix protein pp65 is the principal viral antigen present in peripheral blood leukocytes during an active cytomegalovirus infection. J. Gen. Virol.73:2923-2932.
    OpenUrlPubMedWeb of Science
  13. 13.↵
    Harada, K., Y. Eizuru, Y. Isashiki, S. Ihara, and Y. Minamishima. 1997. Genetic analysis of a clinical isolate of human cytomegalovirus exhibiting resistance against both ganciclovir and cidofovir. Arch. Virol.142:215-225.
    OpenUrlCrossRefPubMedWeb of Science
  14. 14.↵
    Ijichi, O., D. Michel, T. Mertens, K. Miyata, and Y. Eizuru. 2002. GCV resistance due to the mutation A594P in the cytomegalovirus protein UL97 is partially reconstituted by a second mutation at D605E. Antivir. Res.53:135-142.
    OpenUrlCrossRefPubMed
  15. 15.↵
    Ishov, A. M., A. G. Sotnikov, D. Negorev, O. V. Vladimirova, N. Neff, T. Kamitani, E. T. Yeh, J. F. Strauss III, and G. G. Maul. 1999. PML is critical for ND10 formation and recruits the PML-interacting protein daxx to this nuclear structure when modified by SUMO-1. J. Cell Biol.147:221-234.
    OpenUrlAbstract/FREE Full Text
  16. 16.↵
    Ishov, A. M., R. M. Stenberg, and G. G. Maul. 1997. Human cytomegalovirus immediate early interaction with host nuclear structures: definition of an immediate transcript environment. J. Cell Biol.138:5-16.
    OpenUrlAbstract/FREE Full Text
  17. 17.↵
    Katano, H., K. Ogawa-Goto, H. Hasegawa, T. Kurata, and T. Sata. 2001. Human-herpesvirus-8-encoded K8 protein colocalizes with the promyelocytic leukemia protein (PML) bodies and recruits p53 to the PML bodies. Virology286:446-455.
    OpenUrlCrossRefPubMed
  18. 18.↵
    Korioth, F., G. G. Maul, B. Plachter, T. Stamminger, and J. Frey. 1996. The nuclear domain 10 (ND10) is disrupted by the human cytomegalovirus gene product IE1. Exp. Cell Res.229:155-158.
    OpenUrlCrossRefPubMedWeb of Science
  19. 19.↵
    Landry, M. L., S. Stanat, K. Biron, D. Brambilla, W. Britt, J. Jokela, S. Chou, W. L. Drew, A. Erice, B. Gilliam, N. Lurain, J. Manischewitz, R. Miner, M. Nokta, P. Reichelderfer, S. Spector, A. Weinberg, B. Yen-Lieberman, and C. Crumpacker. 2000. A standardized plaque reduction assay for determination of drug susceptibilities of cytomegalovirus clinical isolates. Antimicrob. Agents Chemother.44:688-692.
    OpenUrlAbstract/FREE Full Text
  20. 20.↵
    Maul, G. G., D. Negorev, P. Bell, and A. M. Ishov. 2000. Review: properties and assembly mechanisms of ND10, PML bodies, or PODs. J. Struct. Biol.129:278-287.
    OpenUrlCrossRefPubMedWeb of Science
  21. 21.↵
    Meigata, K., R. Hondo, A. Fujima, M. Shinkai-Shibata, S. Itoh, K. Kikuchi, Y. Ando, N. Ichikawa, Y. Nomura, K. Watanabe, H. Degawa, Y. Beck, S. Tomikawa, T. Nagao, and H. Uchida. 1996. Titration of human cytomegalovirus (HCMV) DNA in urine by combined use of PCR and microplate hybridization in a renal transplant patient with HCMV pneumonitis. Jpn. J. Med. Sci. Biol.49:121-127.
    OpenUrlPubMed
  22. 22.↵
    Muller, S., and A. Dejean. 1999. Viral immediate-early proteins abrogate the modification by SUMO-1 of PML and Sp100 proteins, correlating with nuclear body disruption. J. Virol.73:5137-5143.
    OpenUrlAbstract/FREE Full Text
  23. 23.↵
    Muller, S., M. J. Matunis, and A. Dejean. 1998. Conjugation with the ubiquitin-related modifier SUMO-1 regulates the partitioning of PML within the nucleus. EMBO J.17:61-70.
    OpenUrlAbstract
  24. 24.↵
    Neyts, J., R. Snoeck, D. Schols, J. Balzarini, J. D. Esko, A. Van Schepdael, and E. De Clercq. 1992. Sulfated polymers inhibit the interaction of human cytomegalovirus with cell surface heparan sulfate. Virology189:48-58.
    OpenUrlCrossRefPubMedWeb of Science
  25. 25.↵
    Nowlin, D. M., N. R. Cooper, and T. Compton. 1991. Expression of a human cytomegalovirus receptor correlates with infectibility of cells. J. Virol.65:3114-3121.
    OpenUrlAbstract/FREE Full Text
  26. 26.↵
    Ogawa-Goto, K., Y. Arao, Y. Ito, T. Ogawa, T. Abe, T. Kurata, S. Irie, and H. Akanuma. 1998. Binding of human cytomegalovirus to sulfated glucuronyl glycosphingolipids and their inhibitory effects on the infection. J. Gen. Virol.79:2533-2541.
    OpenUrlCrossRefPubMedWeb of Science
  27. 27.↵
    Ogawa-Goto, K., S. Irie, A. Omori, Y. Miura, H. Katano, H. Hasegawa, T. Kurata, T. Sata, and Y. Arao. 2002. An endoplasmic reticulum protein, p180, is highly expressed in human cytomegalovirus-permissive cells and interacts with the tegument protein encoded by UL48. J. Virol.76:2350-2362.
    OpenUrlAbstract/FREE Full Text
  28. 28.↵
    Paya, C. V., A. D. Wold, and T. F. Smith. 1987. Detection of cytomegalovirus infections in specimens other than urine by the shell vial assay and conventional tube cell cultures. J. Clin. Microbiol.25:755-757.
    OpenUrlAbstract/FREE Full Text
  29. 29.↵
    Prix, L., J. Maierl, G. Jahn, and K. Hamprecht. 1998. A simplified assay for screening of drug resistance of cell-associated cytomegalovirus strains. J. Clin. Virol.11:29-37.
    OpenUrlCrossRefPubMedWeb of Science
  30. 30.↵
    Razonable, R. R., C. V. Paya, and T. F. Smith. 2002. Role of the laboratory in diagnosis and management of cytomegalovirus infection in hematopoietic stem cell and solid-organ transplant recipients. J. Clin. Microbiol.40:746-752.
    OpenUrlFREE Full Text
  31. 31.↵
    Revello, M. G., E. Percivalle, A. Di Matteo, F. Morini, and G. Gerna. 1992. Nuclear expression of the lower matrix protein of human cytomegalovirus in peripheral blood leukocytes of immunocompromised viraemic patients. J. Gen. Virol.73:437-442.
    OpenUrlPubMedWeb of Science
  32. 32.↵
    Sasaki, T., A. Okayama, Y. Eizuru, K. Murai, H. Matsuoka, H. Uno, Y. Minamishima, H. Kataoka, M. Koono, Y. Matsuura, N. Nao-i, A. Sawada, N. Tachibana, and H. Tsubouchi. 1997. Progressive retinitis-encephalitis due to ganciclovir-resistant cytomegalovirus associated with aplastic anemia. Intern. Med.36:375-379.
    OpenUrlCrossRefPubMed
  33. 33.↵
    Stanat, S. C., J. E. Reardon, A. Erice, M. C. Jordan, W. L. Drew, and K. K. Biron. 1991. Ganciclovir-resistant cytomegalovirus clinical isolates: mode of resistance to ganciclovir. Antimicrob. Agents Chemother.35:2191-2197.
    OpenUrlAbstract/FREE Full Text
  34. 34.↵
    Zhong, S., S. Muller, S. Ronchetti, P. S. Freemont, A. Dejean, and P. P. Pandolfi. 2000. Role of SUMO-1-modified PML in nuclear body formation. Blood95:2748-2752.
    OpenUrlAbstract/FREE Full Text
PreviousNext
Back to top
Download PDF
Citation Tools
Novel Real-Time Monitoring System for Human Cytomegalovirus- Infected Cells In Vitro That Uses a Green Fluorescent Protein-PML-Expressing Cell Line
T. Ueno, Y. Eizuru, H. Katano, T. Kurata, T. Sata, S. Irie, K. Ogawa-Goto
Antimicrobial Agents and Chemotherapy Jul 2006, 50 (8) 2806-2813; DOI: 10.1128/AAC.01641-05

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Print

Alerts
Sign In to Email Alerts with your Email Address
Email

Thank you for sharing this Antimicrobial Agents and Chemotherapy article.

NOTE: We request your email address only to inform the recipient that it was you who recommended this article, and that it is not junk mail. We do not retain these email addresses.

Enter multiple addresses on separate lines or separate them with commas.
Novel Real-Time Monitoring System for Human Cytomegalovirus- Infected Cells In Vitro That Uses a Green Fluorescent Protein-PML-Expressing Cell Line
(Your Name) has forwarded a page to you from Antimicrobial Agents and Chemotherapy
(Your Name) thought you would be interested in this article in Antimicrobial Agents and Chemotherapy.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Share
Novel Real-Time Monitoring System for Human Cytomegalovirus- Infected Cells In Vitro That Uses a Green Fluorescent Protein-PML-Expressing Cell Line
T. Ueno, Y. Eizuru, H. Katano, T. Kurata, T. Sata, S. Irie, K. Ogawa-Goto
Antimicrobial Agents and Chemotherapy Jul 2006, 50 (8) 2806-2813; DOI: 10.1128/AAC.01641-05
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Top
  • Article
    • ABSTRACT
    • MATERIALS AND METHODS
    • RESULTS
    • DISCUSSION
    • ACKNOWLEDGMENTS
    • FOOTNOTES
    • REFERENCES
  • Figures & Data
  • Info & Metrics
  • PDF

KEYWORDS

Antigens, Viral
cytomegalovirus
Cytomegalovirus Infections
Green Fluorescent Proteins
Immediate-Early Proteins
Neoplasm Proteins
Nuclear Proteins
transcription factors
Tumor Suppressor Proteins

Related Articles

Cited By...

About

  • About AAC
  • Editor in Chief
  • Editorial Board
  • Policies
  • For Reviewers
  • For the Media
  • For Librarians
  • For Advertisers
  • Alerts
  • AAC Podcast
  • RSS
  • FAQ
  • Permissions
  • Journal Announcements

Authors

  • ASM Author Center
  • Submit a Manuscript
  • Article Types
  • Ethics
  • Contact Us

Follow #AACJournal

@ASMicrobiology

       

ASM Journals

ASM journals are the most prominent publications in the field, delivering up-to-date and authoritative coverage of both basic and clinical microbiology.

About ASM | Contact Us | Press Room

 

ASM is a member of

Scientific Society Publisher Alliance

 

American Society for Microbiology
1752 N St. NW
Washington, DC 20036
Phone: (202) 737-3600

Copyright © 2021 American Society for Microbiology | Privacy Policy | Website feedback

Print ISSN: 0066-4804; Online ISSN: 1098-6596