Skip to main content
Journal of Virology logoLink to Journal of Virology
. 1998 Dec;72(12):9491–9502. doi: 10.1128/jvi.72.12.9491-9502.1998

Recombinant, Replication-Defective Adenovirus Gene Transfer Vectors Induce Cell Cycle Dysregulation and Inappropriate Expression of Cyclin Proteins

Robert P Wersto 1,*, Eugene R Rosenthal 2, Prem K Seth 3, N Tony Eissa 2, Robert E Donahue 1
PMCID: PMC110446  PMID: 9811682

Abstract

First-generation adenovirus (Ad) vectors that had been rendered replication defective by removal of the E1 region of the viral genome (ΔE1) or lacking the Ad E3 region in addition to E1 sequences (ΔE1ΔE3) induced G2 cell cycle arrest and inhibited traverse across G1/S in primary and immortalized human bronchial epithelial cells. Cell cycle arrest was independent of the cDNA contained in the expression cassette and was associated with the inappropriate expression and increase in cyclin A, cyclin B1, cyclin D, and cyclin-dependent kinase p34cdc2 protein levels. In some instances, infection with ΔE1 or ΔE1ΔE3 Ad vectors produced aneuploid DNA histogram patterns and induced polyploidization as a result of successive rounds of cell division without mitosis. Cell cycle arrest was absent in cells infected with a second-generation ΔE1Ad vector in which all of the early region E4 except the sixth open reading frame was also deleted. Consequently, E4 viral gene products present in ΔE1 or ΔE1ΔE3 Ad vectors induce G2 growth arrest, which may pose new and unintended consequences for human gene transfer and gene therapy.


Based on the tropism of wild-type adenovirus (Ad) for the respiratory epithelia (30) and its ability to infect nonreplicating cells (40), replication-defective Ad vectors were thought to be the ideal approach by gene therapy to correct the physiological defects in the airways of individuals having the inherited human disease cystic fibrosis (CF). Numerous studies have established the feasibility of Ad vectors to transfer the cDNA encoding the human cystic fibrosis transmembrane conductance regulator (CFTR) to cells in vitro and in vivo in animal models (4). Culminating in human clinical trials (4), these studies have become the prototype for other Ad-mediated gene therapy protocols targeting cancers, inherited metabolic deficiencies, and cardiovascular disease (1, 20, 73).

Although Ad vectors achieve high levels of transgene expression compared to other viral and nonviral gene transfer strategies (57), several obstacles have hindered the success of human trials for CF gene therapy. Gene transfer to differentiated columnar ciliated cells lining the human airway epithelium is poor (24); by contrast, the preferred targets for infection by Ad vectors are the underlying basal cells exposed to the airway lumen following mechanical abrasion (53) or regenerating epithelial cells (16). Ad-mediated gene transfer is epichromosomal, limiting the duration of the CFTR cDNA expression as the airway epithelium undergoes cellular turnover (49), thus requiring periodic exposure to Ad vectors (82). Moreover, contrary to early expectations, the inflammatory and host immune responses evoked by replication-defective Ad vectors (36, 39, 80) have led to concerns regarding their potential safety for gene therapy (4) and suggestions that immunosuppressive therapy be given during Ad-mediated human gene transfer (33, 37).

Little information is available regarding the impact of the transcription of viral genes remaining in ΔE1 and ΔE1ΔE3 Ad vectors on host cells (38). In the context of CF gene therapy, deletion of the E2a Ad gene along with E1 sequences has been reported to diminish deleterious cytotoxic T-lymphocyte responses (80). The Ad E4 region contains seven open reading frames (ORFs) encoding regulatory proteins involved in the viral life cycle (43). Deletion of most E4 ORF sequences from ΔE1 Ad vectors is thought to minimize the generation of replication-competent Ad and the induction of cellular immune responses and cytopathic effects (74). At the level of host cell DNA synthesis, infection with a ΔE1ΔE3 Ad vector has been reported to decrease cell proliferation (S phase) and induce apoptosis in human primary airway cells in vitro (70). An observation among adenovirologists has been the occurrence of large nonadherent cells present in Ad-infected cultures. Because cell size increases as cells traverse the cell cycle, we hypothesized that they might represent cells arrested in G2-M. This study demonstrates that first-generation ΔE1 and ΔE1ΔE3 Ad vectors perturb normal cell cycle progression and affect cyclin protein expression. Based on the comparison with a second-generation ΔE1 Ad vector containing only the E4 ORF6 region, we postulate that this mechanism involves proteins encoded by E4 sequences that remain in first-generation ΔE1 and ΔE1ΔE3 Ad vectors. Because ΔE1 and ΔE1ΔE3 Ad vectors lacking cDNA in the expression cassette or containing a marker transgene such as the bacterial lacZ gene are often used as control vectors, their effects on cell proliferation add new variables to gene transfer studies using first-generation Ad vectors.

MATERIALS AND METHODS

Ad vectors.

ΔE1 and ΔE1ΔE3 Ad vectors were propagated in 293 cells, purified by cesium chloride density centrifugation, and titrated by plaque assay as previously described (77). Recombinant virus was stored in vehicle buffer (10% glycerol, 10 mM Tris HCl, 1 mM MgCl2 [pH 7.4]) at −70°C. Viral infections are given as the multiplicity of infection (MOI) expressed as the number of PFU per cell. The AdCFTR, AdCL, Adα1AT, AdLacZ, and AV1Null vectors (described further in Results) are based on the Ad type 5 (Ad5) genome and lack all of the E1a, 69.5% of the left-hand portion of the E1b, and 66% of the middle section of the E3 regions. The absence of Ad E1a sequences was verified in aliquots of all ΔE1 and ΔE1ΔE3 vectors by using a PCR-based assay (17), and virus replication was assessed as previously described (65). The deletion mutant Addl312 lacks the E1a region (32) and Addl366 lacks the majority of the E4 region (26); both were gifts from T. Shenk (Princeton University, Princeton, N.J.). The Ad2/CMVβgal-5 vector, modified in the ΔE4 region to contain only ORF6 (ΔE1E4ORF6), uses a cytomegalovirus (CMV) promoter to drive the expression of the bacterial lacZ gene (2) and was obtained from Genzyme Corp. (Framingham, Mass.). In all experiments, uninfected control cells were treated with the largest volume of vehicle buffer used for virus dilution. Particle-to-infectious unit ratios (47) were below 50:1 for all vectors (8:1 for AdCFTR; 22:1 for AdCL; 32:1 for AV1Null; 35:1 for Ad2/CMVβgal-5; 40:1 for Adα1AT; 42:1 for AdLacZ; and 45:1 for Addl312).

Cells and cell culture.

IB3-1 cells, derived from the bronchial epithelium of a CF individual (84), were obtained from P. Zeitlin (Johns Hopkins University, Baltimore, Md.) and grown in modified LHC-8 medium. The normal human tracheal epithelial cell line HTE-80 (25), a gift from W. Guggino (Johns Hopkins University), was grown in Iscove’s modified Dulbecco’s medium supplemented with 20% fetal calf serum and 25 mM HEPES. Both IB3-1 and HTE-80 cells were propagated on fibronectin-coated substrates. CFPAC-1 cells, derived from a pancreatic adenocarcinoma of a CF individual (63), were a gift from R. Frizzell (University of Alabama, Birmingham, Ala.) and grown in Iscove’s modified Dulbecco’s medium with 10% fetal calf serum, as were the T84 and HT29 colonic adenocarcinoma cell lines (American Type Culture Collection, Rockville, Md.). Normal human bronchial epithelial (NHBE) cells were obtained from Clonetics (San Diego, Calif.) and propagated in SAGM medium according to the manufacturer’s directions. NHBE cells were used between four and six cell divisions after recovery from cryopreservation.

Flow cytometric analysis.

DNA cell cycle analysis was measured on propidium iodide (PI)-stained nuclei by using either a FACScan (Becton Dickinson Immunocytometry Systems, San Jose, Calif.) or EPICS Elite (Coulter, Hialeah, Fla.). Cell cycle compartments were deconvoluted from single-parameter DNA histograms of 10,000 cells by using Multicycle (Phoenix Flow Systems, San Diego, Calif.), and debris and doublets were removed via software algorithms (78). In all experiments, control and Ad-infected cells were harvested by trypsinization and pooled with the supernatant media from the corresponding culture to reflect accurately changes in the entire cell population.

Correlated measurements of CFTR protein expression across the cell cycle were obtained by indirect immunofluorescence of acetone-fixed (15 min at −20°C) cells incubated (1 h) with a goat anti-human antibody to the C terminus of the CFTR protein (Genzyme) followed by fluorescein isothiocyanate (FITC)-labeled goat anti-mouse immunoglobulin G (IgG) (Southern Biotech, Birmingham, Ala.). Labeled cells were stained with PI (10 μg/ml) and treated with RNAse (100 μg/ml) prior to flow cytometric analysis. Protease inhibitors (46) were included in all buffers, as was 10% goat serum. Isotype-matched mouse IgG was used in place of the CFTR antibody for negative controls.

5-Bromodeoxyuridine (BrdU) incorporation was measured in cells pulsed (30 min) with 10 μM BrdU (Sigma). BrdU content was analyzed as described by Schutte et al. (64), using an FITC-labeled monoclonal antibody to BrdU (clone B44; Becton Dickinson). Labeled cells were washed and resuspended in phosphate-buffered saline (PBS) containing 10 μg of PI per ml for 30 min prior to flow cytometric analysis.

For evaluation of cyclin protein, p34cdc2 protein kinase, and MPM-2 antibody expression across the cell cycle, trypsinized cells were fixed for 30 min at −20°C in a 1:1 acetone-methanol mixture (cyclins A and B1) or 10 min with 1% paraformaldehyde in PBS (cyclin D), followed by washing in 70% ethanol and PBS, permeabilized for 5 min with 0.25% Triton X-100 in PBS containing 1% bovine serum albumin, and incubated overnight (4°C) with monoclonal antibodies to cyclin A, B1, or D (Pharmingen, San Diego, Calif.) (13) or p34cdc2 (clone HCDC1; ICN Biomedicals, Costa Mesa, Calif.) (3). Cells were subsequently incubated (90 min) with FITC-labeled goat-anti-mouse Ig antibody (Caltag Laboratories, Burlingame, Calif.) and resuspended in PBS containing PI (10 μg/ml) and RNase (1 mg/ml) for 30 min prior to analysis. Mouse IgG (cyclin B1, cyclin D, and p34) or IgE (cyclin A) was used at the same antibody concentrations as negative controls. In all dual-parameter flow cytometric measurements, green FITC fluorescence was measured at 525 nm and red PI fluorescence was measured at >650 nm. When measurements were obtained with a FACScan, photomultiplier FL3 was reconfigured for doublet discrimination. All bivariate distributions were gated on PI fluorescence area versus fluorescence height to exclude G0/1 and G2 doublets from the analyses. Each experiment was repeated two to three times unless indicated otherwise.

Additional procedures.

Cell volumes were measured with a Coulter Counter (model ZBI) connected to a C256 channel analyzer (Coulter). Cyclin kinase activity was measured by immunoprecipitation with either an anti-Cdk2 or an anti-cyclin B1 antibody (Santa Cruz Biotechnology, Santa Cruz, Calif.); [32P]ATP (Amersham, Arlington Heights, Ill.) and histone H1 (Gibco BRL, Gaithersburg, Md.) were used as substrates (11). lacZ activity was quantitated by using C12 fluorescein di-β-d-galactopyranoside as the substrate (ImaGene Green lacZ kit; Molecular Probes, Eugene, Oreg.) as specified by the manufacturer. Endogenous β-d-galactosidase activity was inhibited by treatment with chloroquine.

RESULTS

Ad vectors induce G2 cell cycle arrest.

In epithelial cell lines commonly used to assess the efficiency of Ad-mediated gene transfer for CF gene therapy (CFPAC-1, IB3-1, and HTE-80), infection with ΔE1ΔE3 Ad vectors conveying the human CFTR (AdCFTR), the human catalase (AdCL), or human α-1-antitrypsin (Adα1AT) cDNA, or control vectors lacking human transgenic cDNA (AV1Null or the ΔE1 vector Addl312), caused an Ad dose-dependent increase in the number of nonadherent cells in the culture medium. Microscopically, these rounded cells were large and upon staining with a DNA fluorochrome (Hoechst 33342) possessed none of the morphologic attributes of apoptotic cells (i.e., nuclear condensation or fragmentation and decreased area).

Because the cell volume of the nonadherent cells was roughly double that of the attached cells, we hypothesized that Ad infection might induce G2-M arrest. DNA cell cycle analysis verified a correlation between Ad dose and an increase in the G2-M fraction (Table 1). Flow cytometric features of apoptosis (12), such as the presence of a subdiploid peak to the left of the G0/1 peak in the DNA histograms (Fig. 1, for example), a decrease in forward angle light scatter, a decrease in the integrity of the plasma membrane as assessed by supravital staining with PI or Hoechst 33342, or detection of apoptotic cells by using BrdU to label DNA strand breaks (44), were absent in Ad-infected cells (data not shown). Although CFTR protein overexpression has been reported to cause G2-M arrest in nonhuman primate kidney cells (62), cells infected with non-CFTR-containing vectors (AdCL and Adα1AT) or lacking human transgene cDNA (AV1Null and Addl312) became G2-M arrested (for example, a >150% increase above control levels for cells infected with AV1Null [Table 1]), indicating that this effect was independent of the cDNA contained in the expression cassettes.

TABLE 1.

Replication-defective Ad vectors increase the G2-M fraction

Cell type Mean % of G2-M cells ± SEMa
AdCFTR dose MOI of:
0 5 20 50 100 200 400 800
IB3-1 9.4 ± 0.7 10.3 ± 1.5 15.8 ± 3.4 (b) 22.0 ± 1.8 (b) 32.2 ± 3.1 (b) 35.2 ± 3.4 (b) 43.8 ± 5.1 (b) 50.3 ± 18.1 (b)
CFPAC 11.3 ± 0.9 10.4 ± 2.4 13.6 ± 1.0 (a) 15.5 ± 0.4 (a) 27.3 ± 2.0 (b)
HTE-80 18.3 ± 2.6 22.8 ± 2.0 30.4 ± 1.8 (b) 42.3 ± 1.6 (b) 47.1 ± 0.9 (b)
NHBE 8.6 ± 0.8 15.2 ± 2 (b) 24.1 ± 1.6 (b) 36.9 ± 1.6 (b) 44.7 ± 2.5 (b)
Mean % of G2-M cells ± SEMa
Infection withb:
No vector AdCFTR AdCL Adα1AT AV1Null Addl312
8.2 ± 0.5 (14)c 37.5 ± 3.5 (11) 34.8 ± 1.5 (6) 43.5 ± 0.6 (2) 18.3 ± 0.3 (2) 23.2 ± 1.25 (3)
10.5 ± 0.6 (9) 16.6 ± 1.7 (5) 15.2 ± 1.4 (5)
18.2 ± 1.8 (6) 30.4 ± 1.8 (3) 28.1 ± 4.7 (3)
8.6 ± 0.8 (13) 36.9 ± 1.6 (7) 28.2 ± 1.5 (7) 66.5 ± 4.3 (2)
a

Derived from single-parameter DNA histograms of cells infected with Ad vectors for 72 h. The coefficient of variation of the G0/1 peak, a measure of resolution of DNA cell cycle analysis, was <2.5%. The ratio of the G2-M to the G0/1 peak was 2.0 (range, 1.92 to 2.02), indicating stoichiometric binding of PI. —, data not obtained. t-test P values for comparisons of uninfected and AdCFTR-infected cells are <0.05 (a) and <0.01 (b) (n = 3 to 12). 

b

Cells were infected with Ad vectors at an MOI of 200. Uninfected cells were treated with virus vehicle. P values are <0.01 for cells infected with different Ad vectors. Infection with laboratory-grade and clinical-grade AdCFTR vectors (IB3-1; 5 days) produced essentially equivalent increases in the percentage of G2-M cells (56.3% ± 2.7% [n = 6] versus 62.5% ± 6.4% [n = 3], respectively). DNA histograms from Ad vectors (AdCFTR and Addl312) inactivated by UV irradiation (254 nm at 6 cm for 2 min [66]) were superimposable on histograms from cells treated with vehicle alone (control). 

c

Number of determinations. 

FIG. 1.

FIG. 1

Ad vectors induce G2 cell cycle arrest. IB3-1 cells were exposed to either 5 μM SKF 96365, a reversible and specific inhibitor that arrests cells in M phase (50), for 24 h, virus vehicle (control), or AdCFTR (200 PFU/cell) for 72 h, fixed, and incubated with MPM-2 antibody. The trapezoidal window represents the level of immunofluorescence staining of cells stained with isotype IgG control, indicating that only M-phase cells reacted with this antibody. The corresponding DNA histogram from each bivariate display is shown projected above the dual-parameter dot plots. The G2- and M-phase populations are indicated by solid boxes. Data represent measurements from 25,000 cells. An arrow represents the position of G2-M cells in single-parameter DNA histograms. There is no peak to the left of the G0/1 peak, denoting the absence of apoptotic cells.

To exclude the possibility that Ad-induced G2-M arrest was specifically restricted to human bronchial epithelial cells immortalized by simian virus 40 (IB3-1 and HTE-80) or epithelial cells derived from tumors (CFPAC-1), primary cultures of NHBE cells were infected with Ad vectors and subjected to DNA cell cycle analysis. NHBE cells arrested in G2-M in an Ad dose-dependent manner (Table 1), comparable to the levels observed in Ad-infected IB3-1 cells. Infection at low MOIs (5 PFU/cell in NHBE cells and 20 PFU/cell in IB3-1 cells for 72 h [Table 1]) nearly doubled the percentage of G2-M cells. Generally, the magnitude of G2-M growth arrest was proportional to Ad dose, culture time after infection, and S-phase transit rate. In cells with slow rates of growth (HTE-80 or normal human endothelial vein cells) infected with Addl312, arrest occurred at either higher Ad dose (200 PFU/cell) or at an MOI of 25 following increased culture time after infection (6 to 8 days [data not shown]).

Because single-parameter DNA histograms cannot discriminate G2 from M-phase cells, immunofluorescence staining of M-phase-specific proteins identified by the MPM-2 antibody (14, 23) was used to delineate which of these compartments was increased in ΔE1 or ΔE1ΔE3 Ad-infected cells. Ad infection specifically arrested cells in the G2 phase of the cell cycle (Fig. 1). Similar results were obtained when total nuclear protein was correlated with DNA content (54), another flow cytometric technique to distinguish G2 from M cells; in these experiments, M cells constituted <5% of the total number of cells in Ad-infected cultures.

Ad vectors affect S-phase entry.

ΔE1ΔE3 Ad vectors reportedly decrease cell proliferation (70). To assess directly their effect on cellular DNA synthesis, we exposed AdCFTR-infected cells to BrdU and quantitated incorporation of this thymidine analog by flow cytometry (30-min pulse) (Fig. 2A). The percentages of BrdU-labeled cells decreased slightly from 73 ± 1 (control) to 67 ± 1 (MOI of 25) and 55 ± 2 (MOI of 200; P < .05). Diminished BrdU incorporation was also observed with other vectors (AdCL and Addl312 [data not shown]).

FIG. 2.

FIG. 2

(A) Ad vectors inhibit progression from G2 and delay the entry of cells into S phase. IB3-1 cells were treated with virus vehicle or AdCFTR (25 and 200 PFU/cell) for 72 h, pulse-labeled with BrdU, and monitored for an additional 24 or 48 h. Data represent measurements from 25,000 cells. (B) Cartoon of the positions of the BrdU+ and BrdU populations across the cell cycle.

These observations suggested that infection with the ΔE1 or ΔE1ΔE3 Ad vector may affect multiple cell cycle checkpoints. To confirm this hypothesis, the progression of control and Ad-infected cells through the cell cycle was analyzed by BrdU pulse-chase analysis (Fig. 2A). Following a 30-min BrdU pulse, cells were chased for 24 or 48 h in fresh growth medium. During the first cell division cycle after the BrdU pulse (24-h chase), BrdU-labeled S-phase cells traverse the cell cycle into G2, divide, and give rise to BrdU-expressing (BrdU+) G1 cells. Cells in G2-M during the BrdU pulse are unlabeled and after mitosis result in G1 cells lacking BrdU incorporation (BrdU G1 cells). Likewise, cells in G1 during the BrdU pulse are also unlabeled and appear as BrdU S-phase cells after the first division cycle. Following a second cell division (48-h chase), BrdU+ S-phase cells subsequently reappear. In control, uninfected IB3-1 cells, BrdU+ G1 and BrdU+ S-phase cells were present at the 24- and 48-h chase time points, respectively (Fig. 2A, vehicle). In contrast, by the 24-h chase, the majority of IB3-1 cells infected with AdCFTR at an MOI of 200 PFU/cell appeared as a BrdU+ G2-arrested population. Only BrdU+ G1 cells from IB3-1 cells infected with AdCFTR at a low MOI (25 PFU/cell) partially reinitiated DNA synthesis (BrdU+ S-phase cells [Fig. 2A, 48-h chase). At an MOI of 200 PFU/cell, BrdU+ G1 cells failed to reenter S phase at 48 h.

Some cells escape from Ad vector-induced cell cycle arrest.

In Ad-infected IB3-1 cultures, a small number of cells escaped G2 arrest and, because BrdU was absent in the chase medium, subsequently appeared by the 48-h chase as BrdU S-phase cells (Fig. 2B, S). This subpopulation could have arisen from cells resistant to infection by the ΔE1ΔE3 Ad vectors. To explore this possibility, we infected IB3-1 cells with an ΔE1ΔE3 Ad vector coding for bacterial β-galactosidase and quantitated lacZ expression by flow cytometry. At 3 and 5 days after infection, only 0.3 to 0.4% of the cells did not express lacZ (at an MOI of either 5, 25, or 200 PFU/cell; correspondingly, the G2-M fraction at these MOIs increased to 11, 18, and 38%, respectively). This contrasts with the 3 to 10% BrdU S-phase cells in the pulse-chase experiments and excludes the conclusion that this subpopulation arose from uninfected cells. An alternate explanation is that the small number of cells in G2-M at the time of Ad infection were able to overcome growth arrest and undergo subsequent cell division.

Ad vectors affect cyclin protein distribution and expression.

Since progression through the eukaryotic cell cycle is regulated by cyclin-dependent kinase holoenzyme complexes (68), it was possible that the cell cycle perturbations observed in ΔE1 or ΔE1ΔE3 Ad-infected cells involved alterations in cyclin protein expression. From studies using synchronized cells, flow cytometric assessment of cyclin protein expression is comparable to conventional blotting techniques; however, in the case of asynchronous populations, as studied here, this technique permits the detection of cyclin protein expression with high sensitivity between cell cycle compartments (see reference 13 for a review).

Differential expression of cyclin A protein begins at the G1/S transition and reaches maximal levels in the late S and G2 phases (13). Flow cytometric analysis of cyclin A protein expression across the cell cycle confirmed that this observation held true for uninfected IB3-1 cells (Fig. 3). However, in AdCFTR-infected cells, cyclin A protein was overexpressed in late S and G2 (Fig. 3). Similarly, cyclin B1 protein expression, differentially expressed in late S and G2 (34), also increased with Ad dose and was inappropriately expressed to G1 cells (Fig. 3). Ad-infected NHBE cells showed similar aberrant patterns of cyclin A and B protein expression, beginning at an MOI of 5 (Fig. 4).

FIG. 3.

FIG. 3

Overexpression and unscheduled expression of cyclins A, B1, and D in IB3-1 cells exposed to Ad vectors. Control (vehicle) or AdCFTR-infected cells at the viral MOIs indicated were harvested 72 h after viral infection. Similar results were obtained when cells were infected with AdCL. The trapezoidal window represents the immunofluorescence levels of cells stained with an isotype IgG control (cyclin B1 or D) or IgE (cyclin A). In the cyclin B panel, inappropriate expression of cyclin B protein to G1 cells is indicated by the dashed box. At 25 and 200 PFU/cell, cyclin B protein immunofluorescence is present in the portion of this box above background levels (trapezoidal window). Bivariate histograms were obtained from 25,000 cells.

FIG. 4.

FIG. 4

Overexpression and unscheduled expression of cyclins A and B1 in NHBE cells exposed to Ad vectors. Control (vehicle) or AdCFTR-infected cells at the viral MOIs indicated were harvested 72 h after viral infection. Similar results were obtained when cells were infected with AdCL. The positions of the cell cycle compartments (G1, G2-M, G2, and M) are indicated by either broken lines or arrows and are based on DNA content. The trapezoidal window represents the immunofluorescence levels of cells stained with an isotype IgG control (cyclin B1 or D) or IgE (cyclin A). There is inappropriate expression of both cyclin A and B1 proteins to G1-phase cells. Bivariate histograms were obtained from 25,000 cells.

Because D-type cyclins are rate limiting for progression from G1 into S phase (13), the decreased entry of ΔE1 or ΔE1ΔE3 Ad-infected cells into S phase might be due to a reduction in cyclin D protein expression. This was not observed (Fig. 3). Actually, in Ad-infected cells, cyclin D protein expression was noticeably increased, in an Ad dose-dependent manner (Fig. 3) in both G1 and G2-M phases.

The inability of ΔE1 or ΔE1ΔE3 Ad-infected cells to progress from G2 to M may be the result of a decrease in cyclin-dependent kinase levels. To explore this possibility, we analyzed cyclin-dependent kinase p34cdc2 protein levels and kinase activities in control and Ad-infected cells. Normally, p34cdc2 protein is expressed across all cell cycle phases and is absent only in noncycling G1 cells (Fig. 5A, vehicle) (3). In Ad-infected cells however, p34cdc2 protein was increased in an Ad dose-dependent manner (Fig. 5A). Cdc2 and Cdk2 kinase (cyclin A also associates with Cdk2) activities appeared to be unaffected (Fig. 5B).

FIG. 5.

FIG. 5

p34cdc2 kinase expression and kinase activity in AdCFTR-infected IB3-1 cells. (A) Analysis of Cdc2 protein expression across the cell cycle. The trapezoidal window represents the immunofluorescence levels of cells stained with isotype IgG controls. The positions of the G1 and G2-M populations, denoted by dotted boxes, are based on DNA content. Histograms depict measurements from 25,000 cells. (B) Histone H1 kinase activity of Cdk2 (cyclin A) and Cdc2 (cyclin B1) in IB3-1 cells exposed to vehicle buffer (VHC) or infected with AdCFTR at the indicated MOIs. Cells were harvested 72 h after infection.

Ad gene transfer is not limited to specific cell cycle compartments.

G2 arrest by Ad vectors raised the possibility that gene transfer and subsequent reconstitution of protein and/or function are restricted to a specific cell cycle phase. To address this question, CFTR protein expression following infection with the AdCFTR vector was measured by immunofluorescence and correlated with DNA content. CFTR protein was expressed throughout the cell cycle in asynchronously growing T84 cells, which endogenously express abundant levels of CFTR protein (5) (Fig. 6). By contrast, CFTR protein was absent in the control (Fig. 3, vehicle) or AdCL-infected IB3-1 cells (data not shown). In AdCFTR-infected IB3-1 cells, CFTR protein expression increased in an Ad dose-dependent manner across all cell cycle compartments (Fig. 6), thus confirming that Ad transgene expression was not limited to a specific cell cycle compartment.

FIG. 6.

FIG. 6

Immunofluorescence measurement of CFTR protein expression across the cell cycle. Control (vehicle) or cells infected with AdCFTR at the MOIs indicated were labeled by indirect immunofluorescence with an anti-CFTR antibody (vertical axis) and counterstained with PI for DNA content (horizontal axis). The trapezoidal window represents the immunofluorescence levels of cells stained with isotype IgG controls. The G1 and G2-M populations are indicated by broken lines. The solid line in the T84 bivariate distribution (arrow) shows the level of background immunofluorescence. In this experiment, >98% of T84 reacted with the CFTR antibody. Bivariate histograms were obtained from 25,000 cells.

Aneuploid DNA histogram patterns following ΔE1ΔE3 Ad infection.

In the case of 3T3 mouse fibroblasts, commonly used as a host cell to express foreign genes, infection with the ΔE1 or ΔE1ΔE3 Ad vector induced an additional G0/1 peak (Fig. 7), consistent with the presence of aneuploidy in DNA histograms (76). This DNA aneuploid peak began to appear after infection at an MOI of 20 and became more clearly discernible with increasing Ad dose. Peak position remained constant, denoting a stable population with abnormal DNA content. Interestingly, when Ad-infected IB3-1 cells were passaged by trypsinization and recultured for 72 h, a proliferating DNA aneuploid population was also observed (Fig. 7).

FIG. 7.

FIG. 7

Aneuploidy in Ad-infected cells. (A) 3T3 cells infected with vehicle buffer (VHC) or AdCL at the MOIs indicated for 48 h. A solid arrow shows the position of the aneuploid population in the Ad-infected cells. (B) Appearance of aneuploid subpopulations in recultured Ad-infected IB3-1 cells. Cells were exposed to vehicle buffer or Ad vector for 72 h, trypsinized, and recultured for an additional 72 h in fresh growth medium before DNA cell cycle analysis. The positions of the G1 diploid and G1 aneuploid peaks are labeled. The solid arrow depicts the position of the diploid G2-M peak, while the open arrow indicates the position of aneuploid G2-M cells.

Infection of HT-29 cells, a model for colonic cell differentiation (58) and the in vivo assessment of the response of human colon cancer cells to therapeutic modalities in immunodeficient animals (51), with an ΔE1ΔE3 Ad vector induced polyploidy (cells having >4N DNA content), where G2-arrested cells undergo subsequent rounds of DNA synthesis without proceeding through mitosis. Polyploidization occurred in an Ad dose-dependent manner independent of vector (AdCL, AdCFTR, or Addl312) at an MOI of 5 or greater and was accompanied by the elevated levels of cyclin A and B1 proteins (Fig. 8).

FIG. 8.

FIG. 8

Ad vectors induce polyploidization in HT-29 cells. Cells were treated with either vehicle buffer or AdCFTR at the indicated MOIs and harvested after 72 h for BrdU analysis or for analysis of cyclin A or B1 protein expression. Prior to harvesting, cells were pulsed with 10 μM BrdU for 30 min. The trapezoidal window represents the immunofluorescence levels of cells stained with isotype IgG controls. Data represent measurements from 10,000 cells.

G2 growth arrest is absent in an ΔE1E4ORF6 Ad vector.

To address the possibility that the E4 gene region was responsible for G2 growth arrest caused by ΔE1 and ΔE1ΔE3 Ad vectors, IB3-1 and HT-29 cells were infected with a second-generation Ad2-based vector, Ad2/CMVβgal-5, that had been modified in the E4 region to contain only ORF6 (ΔE1E4ORF6). In contrast to the results observed with the ΔE1-only or ΔE1ΔE3 Ad vector, the percentage of G2-M cells remained essentially unchanged compared to control cells over a wide range of MOIs (5 to 1,000 PFU/cell) in IB3-1 cells infected with ΔE1E4ORF6 (Fig. 9). Only at ≥1,500 PFU/cell did the G2-M fraction significantly increase. A similar situation was observed for NHBE cells (data not shown). While polyploid HT-29 cells were detected after infection with the ΔE1 or ΔE1ΔE3 Ad vector at 5 PFU/cell (Fig. 7), infection with the ΔE1E4ORF6 required an MOI of >1,000. At 400 PFU/cell, DNA histograms showed increased G2-M and a small number of cells having >4N DNA content (Fig. 9). DNA histograms from HT29 or IB3-1 cells infected with an ΔE4 vector (Addl366) at an MOI estimated at >1,000 PFU/cell likewise did not show increased levels of G2-M cells.

FIG. 9.

FIG. 9

Cell cycle arrest and polyploidization is absent in cells infected with Ad2/CMVβgal-5, an Ad vector that contains only the E4 region ORF6. In IB3-1 cells, the G2-M fraction remained unchanged up to 1000 PFU/cell (12% versus 16%, P > 0.8). Between 50 and 200 PFU/cell, the percentage of G2-M cells remained essentially unchanged in HT-29 cells, and polyploid cells having DNA content of >4N were absent.

DISCUSSION

G2 cell cycle arrest was a direct consequence of infection with a replication-defective ΔE1 or ΔE1ΔE3 Ad vector. Growth arrest was absent in cells exposed to UV-irradiated Ad vectors (Table 1), indicating the requirement for Ad gene expression in this process. Moreover, cells infected with a clinical-grade AdCFTR vector, required for use in human gene therapy trials, became G2 arrested (Table 1), thus eliminating the possibility that contaminants in the laboratory-grade constructs were responsible for growth arrest. (Clinical-grade vector is prepared under Good Laboratory Practices and Good Manufacturing Practices for administration in human trials. Vector prepared to these specifications has a frequency of replication-competent viral particles of <1 in 109 [6a]). Interestingly, sporadic examples found in the literature show decreased [3H]thymidine uptake in primary vascular smooth muscle cells (8), approximately a 50% decrease in cell proliferation in U373MG cells (9), a fourfold increase in the G2-M fraction in human non-small-cell lung cancer cells lines (34), and elevated numbers of G2-M cells in numerous tumor cell lines (60) infected with control ΔE1ΔE3 Ad vectors (expressing lacZ) compared to uninfected cells. These changes, although not addressed by the authors, further support our conclusion that cell cycle arrest in primary and immortalized cells is an inherent characteristic of infection with ΔE1 and ΔE1ΔE3 Ad vectors.

The ability of the ΔE1 or ΔE1ΔE3 and not the ΔE1E4ORF6 Ad vector to induce G2 cell cycle arrest suggests that gene products from the Ad E4 region other than ORF6, present in the vectors tested here and demonstrated to have oncogenic properties (48), may affect eukaryotic cell cycle regulation. While the E4 ORF1 encodes a transforming protein (75), the E4 ORF4 protein induces apoptosis in a p53-independent manner in transformed (69) and rodent (41) cells. Biochemically, the product of E4 ORF4 interacts with protein phosphatase 2A (43). Progression through the G2-M phase of the cell cycle requires dephosphorylation of the inactive mitosis-promoting factor complex, consisting of cyclin B and its cyclin-dependent kinase p34cdc2. Passage through mitosis and subsequent division requires MPF inactivation by degradation of cyclin B via the ubiquitin pathway (21). In this context, the elevation of cyclin B protein and p34cdc2 kinase protein levels in Ad-infected cells suggests the possibility that Ad E4 gene products remaining in the ΔE1 and ΔE1ΔE3 Ad vectors can interfere with the degradation of cyclin B by ubiquination or affect the activity of upstream regulators of the cyclin B-cyclin kinase complex, notably Cdc25C, a phosphatase which activates p34cdc2 (10).

The E1a region of the wild-type Ad genome is potentially oncogenic (79) and in this respect was considered to be the most likely candidate to affect the host cell cycle. Screening of our Ad vectors by PCR confirmed the absence of E1a transcripts. While Addl312, defective in E1a, has been reported to replicate in HeLa cells at MOIs of ≥80 PFU/cell (67), viral replication could not be detected in AdCFTR- or AdCL-infected IB3-1 or HT-29 cells (24 or 72 h at MOIs of 5, 25, and 200 [data not shown]), confirming the absence of wild-type E1a virus or cell-derived E1a-like function. Since G2 arrest was observed in cells infected with clinical-grade AdCFTR vector, Ad replication could be further excluded. Ad5 E1a induces cellular DNA synthesis in quiescent cells (35), while infection of asynchronous populations of A549, HeLa, or KB cells with Ad12 E1A induces S-phase arrest (22). However, the S-phase fraction was unchanged in serum-starved quiescent IB3-1 cells infected with the ΔE1 or ΔE1ΔE3 Ad vector (data not shown), and Ad-infected cells did not arrest in S phase (Fig. 1 and 2), further confirming the absence of Ad E1a proteins.

Arrest at the G2 stage of the cell cycle is not a phenomenon unique to adenoviruses. G2-M arrest is observed in human foreskin fibroblasts infected with human CMV (31, 59) and in HeLa (55), 293 cells (28), and monocytes (56) infected with human immunodeficiency virus type 1. While the specific portion of the CMV genome responsible for cell cycle arrest is unknown, the human immunodeficiency virus vpr gene product appears to prevent activation of the p34cdc2-cyclin B complex, possibly by interfering with upstream regulation of this complex (28). Consequently, the highly conserved mechanism regulating the G2/M transition of eukaryotic cells appears to be a common target for a diverse group of viral gene products.

The overexpression of cyclin proteins and their inappropriate appearance in cell cycle compartments are important hallmarks in the oncogenic progression of cells (13, 29). Cyclin A overexpression has been implicated in the neoplastic transformation of alveolar epithelial cells (6), and cyclin D1, overexpressed in a number of diverse human cancers, may have a role in the development of non-small-cell lung cancer (61). Interestingly, hepatitis B virus integrates into the human genome at the cyclin A locus, resulting in the constitutive overexpression of cyclin A in hepatocarcinomas (72).

Aneuploid peaks have been observed in DNA histograms from human tissues infected by papillomavirus (7), herpes simplex virus types 1 and 2 (71), and Epstein-Barr virus (52). Several lines of experimental evidence support the conclusion that the increase in G2-M cells (Fig. 1 and Table 1), aneuploidy in recultured IB3-1 cells (Fig. 7), and polyploidy in HT-29 cells (Fig. 8) results from the interaction of viral regions remaining in ΔE1 and ΔE1ΔE3 Ad vectors with cellular elements and does not reflect spurious histogram peaks arising from the contribution of Ad vector DNA itself upon PI staining. AdCFTR infections at 5 PFU/cell (G2-M arrest in NHBE cells [Table 1]) correspond to 40 particles/cell (approximately 1/2,500 of genomic DNA content) and, at 200 PFU/cell, range from 1,600 (AdCFTR) to 4,400 particles/cell (AdCL), or 1/60 and 1/25 of genomic DNA, respectively, levels far lower than the DNA content of 96 human chromosomes present in G2-M cells. More importantly, infection with the Ad2/CMVβgal-5 vector at 200 (HT-29) or 1,000 (IB3-1) PFU/cell corresponding to 1/14 or ∼1/3 of genomic DNA, respectively, had no effect on cell cycle distributions (Fig. 9). In the case of the recultured IB3-1 cells, the most probable explanation is that the aneuploid population represents proliferating Ad-infected cells while the diploid fraction corresponds to cells that escaped cell cycle arrest. By contrast, the single aneuploid DNA stemline observed in Ad-infected 3T3 cells that increases in an Ad dose-dependent manner could be the result of Ad infection limited to a specific cell cycle phase.

In a typical Ad clinical protocol, a single dose of 109 virus particles is administered (19), and infection with 10,000 PFU/cell may be required to impart CFTR function to CF epithelia in vivo (24). The degree of cell cycle inhibition induced by ΔE1 and ΔE1ΔE3 Ad vectors varied between cell types (i.e., abnormalities in cyclin protein expression at an MOI of 5 in primary NHBE cells; polyploidization in HT29 cells at an MOI of 5; nearly a 100% increase in G2-M cells in IB3-1 cells at an MOI of 20). In this context, it is conceivable that the cellular target which interacts with the gene product(s) from the Ad E4 region to induce G2 growth arrest may be present at different levels in a variety of cells. The broad range of MOIs used to infect cells and deliver transgenes (0.5 to 1,000 PFU/cell and >10,000 PFU/cell) in various gene therapy studies reflects both the lack of consensus among investigators for standardization of Ad vector concentrations (47) and the fact that MOIs, especially in ex vivo and in vivo experiments, are estimated values based on arbitrary assumptions (for example, the density of cells in the nasal epithelium has been estimated at 2 × 106 [45] or 3 × 106 [83] cells/cm2). Consequently, the levels used here (i.e., an MOI of 5 to roughly double the percentage of G2-M cells in NHBE cells) are within the range (MOI of 66 to 200) used clinically in an attempt to correct the defect in Cl transport in the nasal epithelium of CF individuals with an Ad vector containing the CFTR cDNA (83).

In the context of gene therapy for inherited diseases, G2 arrest could conceivably prolong gene expression, normally reduced as cells undergo successive cycles of cell division. Cell differentiation usually occurs in the G0/1 phase; however, in the case of CF, where nasal polyps have been shown to have increased proliferative activity (27) and it is estimated that nearly 20% of the airway cells are proliferating, G2 growth arrest by ΔE1 and ΔE1ΔE3 Ad vectors may potentially slow remodeling of the respiratory epithelium (42). Although speculative, this hypothesis is consistent with the observation that the majority of the regenerating cells in explant cultures of human nasal polyp tissue, which were targeted by an AdLacZ vector, lacked reactivity with the Ki-67 antibody, which recognizes proliferating cells and is absent in only quiescent cell cycle compartments (16). In human bronchial xenograft models, Ad-mediated transgene expression was present in all cell types of the surface epithelium except basal cells (18). While it is presumed that these transfected cells arose from Ad-infected basal cells that subsequently underwent division and differentiation, these results are at odds with the concept of differentiation as a stochastic event, implying that all infected basal cells would have differentiated within 72 h of Ad infection.

Based on their ability to induce G2 growth arrest, first-generation ΔE1 and ΔE1ΔE3 Ad vectors may be more appropriate for gene therapy of human cancers (15). Coadministration of genotoxic agents with Ad vectors conveying the cDNAs for G1-phase growth regulatory genes (8, 11, 60), along with the cellular and humoral immune responses evoked by ΔE1 and ΔE1ΔE3 Ad vectors (81), might provide a multifaceted approach to target selectively proliferating tumor cells. In this respect, G2 growth arrest was observed in a wide variety of colonic and mammary adenocarcinoma cell lines commonly used to assess the efficacy of chemotherapeutic agents in vitro and in vivo (77a). By contrast, second-generation vectors, having deletions in the E1 and E4 regions, may alleviate the problems reported here and minimize potential safety concerns regarding use of Ad vectors for gene therapy of inherited diseases of humans.

ACKNOWLEDGMENTS

We thank Harold Ginsberg, National Institute for Allergy and Infectious Diseases, NIH, for critical review and Thomas Shenk, Howard Hughes Medical Institute, Department of Molecular Biology, Princeton University, for providing Ad deletion mutants and helpful comments. We deeply appreciate Alan Smith, Sam Wadsworth, and the members of the Virus Production Group of Genzyme Corp. for providing the ΔE1E4ORF6 Ad vector.

ADDENDUM

While this report was under review, Brand and Strauss (4a) reported that first-generation ΔE1 and ΔE1ΔE3 Ad vectors induce growth retardation and prolongation of the G2-M phase in p53 knockout murine hepatocytes.

REFERENCES

  • 1.Akhter S A, Skaer C A, Kypson A P, McDonald P H, Peppel K C, Glower D D, Lefkowitz R J, Koch W J. Restoration of beta-adrenergic signaling in failing cardiac ventricular myocytes via adenoviral-mediated gene transfer. Proc Natl Acad Sci USA. 1997;94:12100–12015. doi: 10.1073/pnas.94.22.12100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Armentano D, Sookdeo C C, Hehir K M, Gregory R J, St. George J A, Prince G A, Wadsworth S C, Smith A E. Characterization of an adenovirus gene transfer vector containing an E4 deletion. Hum Gene Ther. 1995;6:1343–1353. doi: 10.1089/hum.1995.6.10-1343. [DOI] [PubMed] [Google Scholar]
  • 3.Baroja A, de la Hoz C, Alvarez A, Ispizua A, Bilbao J, de Gandarias J M. Genesis and evolution of high-ploidy tumor cells evaluated by means of the proliferation markers p34cdc2, cyclin B1, PCNA, and 3[H]-thymidine. Cell Prolif. 1996;29:89–100. [PubMed] [Google Scholar]
  • 4.Boucher R C. Current status of CF gene therapy. Trends Genet. 1996;12:81–84. doi: 10.1016/0168-9525(96)81410-7. [DOI] [PubMed] [Google Scholar]
  • 4a.Brand K, Strauss M. Presented at the 1st Annual Meeting of the American Society for Gene Therapy, May 28–31, Seattle, Wash. 1998. [Google Scholar]
  • 5.Buchwald M, Sood R, Auerbach W. Expression of CFTR in cultured human intestinal epithelial cells. In: Tsui L-C, Romeo G, Gieger R, Gorini S, editors. The identification of the CF (cystic fibrosis) gene: recent progress and new research strategies. New York: Plenum Press; 1991. pp. 241–252. [Google Scholar]
  • 6.Bui K C, Wu F, Buckley S, Wu L, Williams R, Carbonaro-Hall D, Hall F L, Warburton D. Cyclin A expression in normal and transformed alveolar epithelial cells. Am J Respir Cell Mol Biol. 1993;9:115–125. doi: 10.1165/ajrcmb/9.2.115. [DOI] [PubMed] [Google Scholar]
  • 6a.Center for Biologics Evaluation and Research. Points to consider in human somatic cell therapy and gene therapy. Rockville, Md: Center for Biologics Evaluation and Research, Food and Drug Administration; 1996. [Google Scholar]
  • 7.Chacho M S, Eppich E, Wersto R P, Koss L G. Influence of human papillomavirus on DNA ploidy determination in genital condylomas. Cancer. 1990;65:2291–2294. doi: 10.1002/1097-0142(19900515)65:10<2291::aid-cncr2820651021>3.0.co;2-m. [DOI] [PubMed] [Google Scholar]
  • 8.Chang M W, Barr E, Seltzer J, Jiang Y-Q, Nabel G J, Nabel E G, Paracek M S, Leiden J M. Cytostatic gene therapy for vascular proliferative disorders with a constitutively active form of the retinoblastoma gene product. Science. 1995;267:518–522. doi: 10.1126/science.7824950. [DOI] [PubMed] [Google Scholar]
  • 9.Chen J, Willingham T, Shuford M, Nisen P D. Tumor suppression and inhibition of aneuploid cell accumulation in human brain tumor cells by ectopic overexpression of the cyclin-dependent kinase inhibitor p27KIP1. J Clin Investig. 1996;97:1983–1988. doi: 10.1172/JCI118631. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Coleman T R, Dunphy W G. Cdc2 regulatory factors. Curr Opin Cell Biol. 1994;6:877–882. doi: 10.1016/0955-0674(94)90060-4. [DOI] [PubMed] [Google Scholar]
  • 11.Craig C, Wersto R, Kim M, Ohri E, Li Z, Katayose D, Lee S J, Trepel J, Cowan K, Seth P. A recombinant adenovirus expressing p27kip1 induces cell cycle arrest and loss of cyclin-cdk activity in human breast cancer cells. Oncogene. 1997;14:2283–2289. doi: 10.1038/sj.onc.1201064. [DOI] [PubMed] [Google Scholar]
  • 12.Darzynkiewcz Z, Bruno S, Del Bino G, Gorczyca W, Hotz M A, Lassota P, Tragnos F. Features of apoptotic cells measured by flow cytometry. Cytometry. 1992;13:795–808. doi: 10.1002/cyto.990130802. [DOI] [PubMed] [Google Scholar]
  • 13.Darzynkiewcz Z, Gong J, Juan G, Ardelt B, Tragnos F. Cytometry of cyclin proteins. Cytometry. 1996;25:1–13. doi: 10.1002/(SICI)1097-0320(19960901)25:1<1::AID-CYTO1>3.0.CO;2-N. [DOI] [PubMed] [Google Scholar]
  • 14.Ding M, Feng Y, Vandre D D. Partial characterization of the MPM-2 phosphoepitope. Exp Cell Res. 1997;231:3–13. doi: 10.1006/excr.1996.3439. [DOI] [PubMed] [Google Scholar]
  • 15.Douglas J T, Curiel D T. Targeted adenoviral vectors for cancer gene therapy. Int J Oncol. 1997;11:341–348. doi: 10.3892/ijo.11.2.341. . (Review.) [DOI] [PubMed] [Google Scholar]
  • 16.Dupuit F, Zahm J-M, Pierrot D, Brezillon S, Bonnet N, Imler J-L, Pavirini A, Puchelle E. Regenerating cells in human airway epithelium represent preferential targets for recombinant adenovirus. Hum Gene Ther. 1995;6:1185–1193. doi: 10.1089/hum.1995.6.9-1185. [DOI] [PubMed] [Google Scholar]
  • 17.Eissa N T, Chu C-S, Danel C, Crystal R G. Evaluation of the respiratory epithelium of normals and individuals with cystic fibrosis for the presence of adenovirus E1a sequences relevant to the use of E1a− adenovirus vectors for gene therapy for the respiratory manifestations of cystic fibrosis. Hum Gene Ther. 1994;5:1105–1114. doi: 10.1089/hum.1994.5.9-1105. [DOI] [PubMed] [Google Scholar]
  • 18.Engelhardt J F, Yang Y, Stratford-Perricaudet L D, Allen E D, Kozarsky K, Perricaudet M, Yankaskas J R, Wilson J M. Direct gene transfer of human CFTR into bronchial human epithelia of xenografts with E1-deleted adenoviruses. Nat Genet. 1993;4:27–33. doi: 10.1038/ng0593-27. [DOI] [PubMed] [Google Scholar]
  • 19.Felgner P L. Improvements in catonic liposomes for in vivo gene transfer. Hum Gene Ther. 1996;7:1791–1793. doi: 10.1089/hum.1996.7.15-1791. [DOI] [PubMed] [Google Scholar]
  • 20.Gahery-Segard H, Molinier-Frenkel V, Boulare C L, Saulnier P, Opolon P, Lengagne R, Gautier E, Cesne A L, Zitvogel L, Venet A, Schatz C, Courtney M, Chevalier T L, Tursz T, Guillet J G, Farace F. Phase I trial of recombinant adenovirus gene transfer in lung cancer: longitudinal study of immune responses to transgene and viral proteins. J Clin Investig. 1997;100:2218–2226. doi: 10.1172/JCI119759. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Glotzer M, Murray A W, Kirschner M W. Cyclin is degraded by the ubiquitin pathway. Nature. 1991;349:132–138. doi: 10.1038/349132a0. [DOI] [PubMed] [Google Scholar]
  • 22.Grand R J A, Ibrahim A P, Taylor A M R, Milner A E, Gregory C D, Gallimore P H, Turnell A S. Human cells arrest in S phase in response to adenovirus 12 E1A. Virology. 1998;244:330–342. doi: 10.1006/viro.1998.9102. [DOI] [PubMed] [Google Scholar]
  • 23.Griscelli F, Li H, Bennaceur-Griscelli A, Soria J, Opolon P, Soria C, Perricaudet M, Yeh P, Lu H. Angiostatin gene transfer: inhibition of tumor growth in vivo by blockage of endothelial cell proliferation associated with a mitosis arrest. Proc Natl Acad Sci USA. 1998;95:6367–6372. doi: 10.1073/pnas.95.11.6367. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Grubb B R, Pickles R J, Ye H, Yankaskas J R, Vick R N, Engelhardt J F, Wilson J M, Johnson L G, Boucher R C. Inefficient gene transfer by adenovirus vector to cystic fibrosis airway epithelia of mice and humans. Nature. 1994;371:802–806. doi: 10.1038/371802a0. [DOI] [PubMed] [Google Scholar]
  • 25.Gruenert D C, Basbaum C B, Welsh M J, Li M, Finkbeiner W E, Nadel J A. Characterization of human tracheal epithelial cells transformed by an origin-defective simian virus 40. Proc Natl Acad Sci USA. 1988;85:5951–5955. doi: 10.1073/pnas.85.16.5951. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Halbert D N, Cutt J R, Shenk T. Adenovirus early region 4 encodes functions required for efficient DNA replication, late gene expression, and host cell shutoff. J Virol. 1985;56:250–257. doi: 10.1128/jvi.56.1.250-257.1985. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Hassid S, Degaute M P, Dawance S, Rombaut K, Nagy N, Choufani G, Decaestecker C, Danguy A, Salmon I, Kiss R. Determination of proliferative activity in nasal polyps. J Clin Pathol. 1997;50:923–928. doi: 10.1136/jcp.50.11.923. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.He J, Choe S, Walker R, Di Marzio P, Margon D O, Landau N R. Human immunodeficiency virus type 1 viral protein R (Vpr) arrests cells in the G2 phase of the cell cycle by inhibiting p34cdc2 activity. J Virol. 1995;69:6705–6711. doi: 10.1128/jvi.69.11.6705-6711.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Hinds P W, Dowdy S F, Eaton E N, Arnold A, Weinberg R A. Function of a human cyclin gene as an oncogene. Proc Natl Acad Sci USA. 1994;91:709–713. doi: 10.1073/pnas.91.2.709. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Horwitz M S. Adenoviridae and their replication. In: Fields B N, Knipe D M, editors. Virology. New York, N.Y: Raven Press; 1990. pp. 1679–1721. [Google Scholar]
  • 31.Jault F M, Jault J-M, Ruchti F, Fortunato E A, Clark C, Corbeil J, Richman D D, Spector D H. Cytomegalovirus infection induces high levels of cyclins, phosphorylated Rb, and p53, leading to cell cycle arrest. J Virol. 1995;69:6697–6704. doi: 10.1128/jvi.69.11.6697-6704.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Jones N, Shenk T. Isolation of adenovirus type 5 host range deletion mutants defective for transformation of rat embryo cells. Cell. 1979;17:683–689. doi: 10.1016/0092-8674(79)90275-7. [DOI] [PubMed] [Google Scholar]
  • 33.Jooss K, Yang Y, Wilson J M. Cyclophosphamide diminishes inflammation and prolongs transgene expression following delivery of adenoviral vectors to mouse lung and liver. Hum Gene Ther. 1996;7:1555–1566. doi: 10.1089/hum.1996.7.13-1555. [DOI] [PubMed] [Google Scholar]
  • 34.Joshi U S, Chen Y Q, Kalemkerian G P, Adil M R, Kraut M, Sarkar F H. Inhibition of tumor cell growth by p21WAF1 adenoviral gene transfer in lung cancer. Cancer Gene Ther. 1998;5:183–191. [PubMed] [Google Scholar]
  • 35.Kaczmarek L, Ferguson B, Rosenberg M, Baserga R. Induction of cellular DNA synthesis by purified adenovirus E1a proteins. Virology. 1986;152:1–10. doi: 10.1016/0042-6822(86)90366-1. [DOI] [PubMed] [Google Scholar]
  • 36.Kaplan J M, St. George J A, Pennington S E, Keyes L D, Johnson R P, Wadsworth S C, Smith A E. Humoral and cellular immune responses of non-human primates to long-term repeated lung exposure to Ad2/CFTR-2. Gene Ther. 1996;3:117–127. [PubMed] [Google Scholar]
  • 37.Kaplan J M, Smith A E. Transient immunosuppression with deoxyspergualin improves longevity of transgene expression and ability of readminister adenoviral vector to the mouse lung. Hum Gene Ther. 1997;8:1095–1104. doi: 10.1089/hum.1997.8.9-1095. [DOI] [PubMed] [Google Scholar]
  • 38.Kay M A, Liu D, Hoogerbrugge P M. Gene therapy. Proc Natl Acad Sci USA. 1977;94:12744–12746. doi: 10.1073/pnas.94.24.12744. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Knowles M R, Hohneker K W, Zhou Z, Olsen J C, Noah T L, Hu P C, Leigh M W, Englehardt J F, Edwards L J, Jones K R, Grossman M, Wilson J M, Johnson L G, Boucher R C. A controlled study of adenoviral-vector-mediated gene transfer in the nasal epithelium of patients with cystic fibrosis. N Engl J Med. 1995;333:823–831. doi: 10.1056/NEJM199509283331302. [DOI] [PubMed] [Google Scholar]
  • 40.Kozarsky K, Wilson J M. Gene therapy: adenovirus vectors. Curr Opin Genet Dev. 1993;3:499–503. doi: 10.1016/0959-437x(93)90126-a. [DOI] [PubMed] [Google Scholar]
  • 41.Lavoie J N, Nguyen M, Marcellus R C, Branton P E, Shore G C. E4orf4, a novel adenovirus death factor that induces p53-independent apoptosis by a pathway that is not inhibited by zVAD-fmk. J Cell Biol. 1998;140:637–645. doi: 10.1083/jcb.140.3.637. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Leigh M W, Kylander J, Yankaskas J R, Boucher R C. Cell proliferation in bronchial epithelium and submucosal glands of cystic fibrosis patients. Am J Respir Cell Mol Biol. 1995;12:695–612. doi: 10.1165/ajrcmb.12.6.7766425. [DOI] [PubMed] [Google Scholar]
  • 43.Leppard K N. E4 gene function in adenovirus, adenovirus vector and adeno-associated virus infections. J Gen Virol. 1997;78:2131–2138. doi: 10.1099/0022-1317-78-9-2131. [DOI] [PubMed] [Google Scholar]
  • 44.Li X, Darzynkiewicz Z. Labeling DNA strand breaks with BrdUTP. Detection of apoptosis and cell proliferation. Cell Prolif. 1995;28:572–579. doi: 10.1111/j.1365-2184.1995.tb00045.x. [DOI] [PubMed] [Google Scholar]
  • 45.Mariassy A T. Epithelial cells of trachea and bronchi. In: Parent R A, editor. Comparative biology of the normal lung. Boca Raton, Fla: CRC Press; 1992. pp. 63–73. [Google Scholar]
  • 46.Marino C R, Matovcik L M, Gorrelick F S, Cohn J A. Localization of the cystic fibrosis transmembrane conductance regulator in pancreas. J Clin Investig. 1991;88:712–716. doi: 10.1172/JCI115358. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Mittereder N, March K L, Trapnell B C. Evaluation of the concentration and bioactivity of adenovirus vectors for gene therapy. J Virol. 1996;70:7498–7509. doi: 10.1128/jvi.70.11.7498-7509.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Moore M, Horikoshi N, Shenk T. Oncogenic potential of the adenovirus E4orf6 protein. Proc Natl Acad Sci USA. 1996;93:11295–11301. doi: 10.1073/pnas.93.21.11295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Morgan R A, Anderson W F. Human gene therapy. Annu Rev Biochem. 1993;62:191–217. doi: 10.1146/annurev.bi.62.070193.001203. [DOI] [PubMed] [Google Scholar]
  • 50.Nordstrom T, Nevanlinna H A, Andersson L C. Mitosis-arresting effect of the calcium channel inhibitor SK&F 96365 on human leukemia cells. Exp Cell Res. 1992;202:487–494. doi: 10.1016/0014-4827(92)90103-f. [DOI] [PubMed] [Google Scholar]
  • 51.Ohwada A, Hirschowitz E A, Crystal R G. Regional delivery of an adenovirus vector containing the Escherichia coli cytosine deaminase gene to provide local activation of 5-fluorocytosine to suppress the growth of colon carcinoma metastatic to liver. Hum Gene Ther. 1996;7:1567–1576. doi: 10.1089/hum.1996.7.13-1567. [DOI] [PubMed] [Google Scholar]
  • 52.Overton W R. False DNA aneuploidy possibly caused by viral DNA. Cytometry. 1995;22:158. doi: 10.1002/cyto.990220214. [DOI] [PubMed] [Google Scholar]
  • 53.Pickles R J, Barker P M, Ye H, Boucher R C. Efficient adenovirus-mediated gene transfer to basal but not columnar cells of cartilaginous airway epithelia. Hum Gene Ther. 1996;7:921–931. doi: 10.1089/hum.1996.7.8-921. [DOI] [PubMed] [Google Scholar]
  • 54.Pollack A. Flow cytometric cell-kinetic analysis by simultaneously staining nuclei with propidium iodide and fluorescein isothiocyanate. Methods Cell Biol. 1990;33:315–323. doi: 10.1016/s0091-679x(08)60535-x. [DOI] [PubMed] [Google Scholar]
  • 55.Re F, Braaten D, Franke E K, Luban J. Human immunodeficiency virus type 1 vpr arrests the cell cycle in G2 by inhibiting the activation of p34cdc2-cyclin B. J Virol. 1995;69:6859–6984. doi: 10.1128/jvi.69.11.6859-6864.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Rogel M E, Wu L I, Emerman M. The human immunodeficiency virus type 1 vpr gene prevents cell proliferation during chronic infection. J Virol. 1995;69:882–888. doi: 10.1128/jvi.69.2.882-888.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Ross G, Erickson R, Knorr D, Motulsky A G, Parkman R, Samulski J, Strauss S E, Smith B R. Gene therapy in the United States: a five-year status report. Hum Gene Ther. 1996;7:1781–1790. doi: 10.1089/hum.1996.7.14-1781. [DOI] [PubMed] [Google Scholar]
  • 58.Rousset M. The human colon carcinoma cell lines HT-29 and Caco-2: two in vitro models for the study of intestinal differentiation. Biochemie. 1986;68:1035–1040. doi: 10.1016/s0300-9084(86)80177-8. [DOI] [PubMed] [Google Scholar]
  • 59.Salvant B S, Fortunato E A, Spector D H. Cell cycle dysregulation by human cytomegalovirus: influence of the cell cycle phase at the time of infection and effects on cyclin transcription. J Virol. 1998;72:3729–3741. doi: 10.1128/jvi.72.5.3729-3741.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Sandig V, Brand K, Herwig S, Lukas J, Bertek J, Strauss M. Adenovirally transferred p16INK4/CDKN2 and p53 genes cooperate to induce apoptotic tumor cell death. Nat Med. 1997;3:313–319. doi: 10.1038/nm0397-313. [DOI] [PubMed] [Google Scholar]
  • 61.Schauer I E, Siriwardana S, Langan T A, Sclafani R A. Cyclin D1 overexpression vs. retinoblastoma inactivation: implications for growth control evasion in non-small cell and small cell lung cancer. Proc Natl Acad Sci USA. 1994;91:7827–7831. doi: 10.1073/pnas.91.16.7827. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Schiavi S C, Abdelkader N, Reber S, Pennington S, Narayana R, McPherson J M, Smith A E, Hoppe IV H, Cheng S H. Biosynthetic and growth abnormalities are associated with high-level expression of CFTR in heterologous cells. Am J Physiol. 1996;270:C341–C351. doi: 10.1152/ajpcell.1996.270.1.C341. [DOI] [PubMed] [Google Scholar]
  • 63.Schoumacher R A, Ram J, Iannuzzi M C, Bradbury N A, Wallace R W, Tom Hon C, Kelly D R, Schmid S M, Gelder F B, Rado T A, Frizzell R A. A cystic fibrosis pancreatic adenocarcinoma cell line. Proc Natl Acad Sci USA. 1990;87:4012–4016. doi: 10.1073/pnas.87.10.4012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Schutte B, Reynders M M J, van Assche C L M V J, Hupperets P S G J, Bosman F T, Blijham C H. An improved method for the immunocytochemical detection of bromodeoxyuridine labeled nuclei using flow cytometry. Cytometry. 1987;8:372–376. doi: 10.1002/cyto.990080405. [DOI] [PubMed] [Google Scholar]
  • 65.Seth P K, Fitzgerald D, Ginsberg H, Willingham M, Pastan I. Evidence that the penton base of adenovirus is involved in potentiation of toxicity of Pseudomonas exotoxin conjugated to epidermal growth factor. Mol Cell Biol. 1984;4:1528–1533. doi: 10.1128/mcb.4.8.1528. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Seth P K, Rosenfeld M, Higginbothom J, Crystal R G. Mechanism of enhancement of DNA expression consequent to cointernalization of a replication-deficient adenovirus and unmodified plasmid DNA. J Virol. 1994;68:933–940. doi: 10.1128/jvi.68.2.933-940.1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Shenk T, Jones N, Colby W, Fowlkes D. Functional analysis of adenovirus-5 host-range deletion mutants defective for transformation of rat embryo cells. Cold Spring Harbor Symp Quant Biol. 1980;44:367–375. doi: 10.1101/sqb.1980.044.01.041. [DOI] [PubMed] [Google Scholar]
  • 68.Sherr C J. Mammalian G1 cyclins. Cell. 1993;73:1059–1065. doi: 10.1016/0092-8674(93)90636-5. [DOI] [PubMed] [Google Scholar]
  • 69.Shtrichman R, Kleinberger T. Adenovirus type 5 E4 open reading frame protein induces apoptosis in transformed cells. J Virol. 1988;72:2975–2982. doi: 10.1128/jvi.72.4.2975-2982.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Teramotto S, Johnson L G, Huang W, Leigh M W, Boucher R C. Effect of adenoviral vector infection on cell proliferation in cultured primary human airway epithelial cells. Hum Gene Ther. 1995;6:1045–1053. doi: 10.1089/hum.1995.6.8-1045. [DOI] [PubMed] [Google Scholar]
  • 71.Trusal L R, Anthony A, Docherty J J. Differential Feulgen-deoxyribonucleic acid hydrolysis patterns of herpes simplex virus type 1 and type 2 infected cells. J Histochem Cytochem. 1975;23:283–288. doi: 10.1177/23.4.47871. [DOI] [PubMed] [Google Scholar]
  • 72.Wang J, Chenivesse X, Henglein B, Brechot C. Hepatitis B virus integration in a cyclin A gene in a hepatocellular carcinoma. Nature. 1990;343:555–557. doi: 10.1038/343555a0. [DOI] [PubMed] [Google Scholar]
  • 73.Wang L, Zoppe M, Hackeng T M, Griffin J H, Lee K F, Verma I M. A factor IX-deficient mouse model for hemophilia B gene therapy. Proc Natl Acad Sci USA. 1997;94:11563–11566. doi: 10.1073/pnas.94.21.11563. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Wang Q, Finer M H. Second-generation adenovirus vectors. Nat Med. 1996;2:714–716. doi: 10.1038/nm0696-714. [DOI] [PubMed] [Google Scholar]
  • 75.Weiss R S, Lee S S, Prasad V B, Javier R T. Human adenovirus early region 4 open reading frame 1 genes encode growth-transforming proteins that may be distantly related to dUTP pyrophosphatase enzymes. J Virol. 1997;71:1857–1870. doi: 10.1128/jvi.71.3.1857-1870.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Wersto R P, Liblit R L, Koss L G. Flow cytometric DNA analysis of human solid tumors: a review of the interpretation of DNA histograms. Hum Pathol. 1991;22:1085–1098. doi: 10.1016/0046-8177(91)90260-v. [DOI] [PubMed] [Google Scholar]
  • 77.Wersto R P, Rosenthal E R, Crystal R G, Spring K R. Uptake of fluorescent dyes associated with functional expression of the cystic fibrosis transmembrane conductance regulator in epithelial cells. Proc Natl Acad Sci USA. 1996;93:1167–1172. doi: 10.1073/pnas.93.3.1167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77a.Wersto, R. P., and P. K. Seth. Unpublished data.
  • 78.Wersto R P, Stetler-Stevenson M A. Debris compensation of DNA histograms and its effect on S-phase analysis. Cytometry. 1995;20:43–52. doi: 10.1002/cyto.990200108. [DOI] [PubMed] [Google Scholar]
  • 79.White E. Regulation of p53-dependent apoptosis by E1A and E1B. Curr Top Microbiol Immunol. 1995;199:34–58. [PubMed] [Google Scholar]
  • 80.Yang Y, Nunes F A, Bercencsi K, Gonczol E, Engelhardt J F, Wilson J M. Inactivation of E2a in recombinant adenoviruses improves the prospect for gene therapy in cystic fibrosis. Nat Genet. 1994;7:362–369. doi: 10.1038/ng0794-362. [DOI] [PubMed] [Google Scholar]
  • 81.Yang Y, Li Q, Ertl H C, Wilson J M. Cellular and humoral immune responses to viral antigens create barriers to lung-directed gene therapy with recombinant adenoviruses. J Virol. 1995;69:2004–2015. doi: 10.1128/jvi.69.4.2004-2015.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Zabner J, Peterson D M, Puga A P, Graham S M, Couture L A, Keyes L D, Lukason M J, St. George J A, Gregory R J, Smith A E, Welsh M J. Safety and efficacy of repetitive adenovirus-mediated transfer of CFTR cDNA to airway epithelia of primates and cotton rats. Nat Genet. 1994;6:75–83. doi: 10.1038/ng0194-75. [DOI] [PubMed] [Google Scholar]
  • 83.Zabner J, Ramsey B W, Meeker D P, Aitken M L, Balfour R P, Gibson R L, Launspach J, Moscicki R A, Richards S M, Standaert T A, Williams-Warren J, Wadsworth S C, Smith A E, Welsh M J. Repeat administration of an adenovirus vector encoding cystic fibrosis transmembrane conductance regulator to the nasal epithelium of patients with cystic fibrosis. J Clin Investig. 1996;97:1504–1511. doi: 10.1172/JCI118573. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Zeitlin P L, Lu L, Rhim J, Cutting G, Stetten G, Kiefer K A, Craig R, Guggino W B. A cystic fibrosis bronchial epithelial cell line: immortalization by adeno12-SV40 infection. Am J Respir Cell Mol Biol. 1991;4:313–319. doi: 10.1165/ajrcmb/4.4.313. [DOI] [PubMed] [Google Scholar]

Articles from Journal of Virology are provided here courtesy of American Society for Microbiology (ASM)

RESOURCES