Bachmann 2002

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

JOURNAL OF VIROLOGY, Jan. 2002, p. 280–291 Vol. 76, No.

1
0022-538X/02/$04.00⫹0 DOI: 10.1128/JVI.76.1.280–291.2002
Copyright © 2002, American Society for Microbiology. All Rights Reserved.

Disturbance of Tumor Necrosis Factor Alpha-Mediated Beta


Interferon Signaling in Cervical Carcinoma Cells
Anastasia Bachmann, Brigitte Hanke, Rainer Zawatzky, Ubaldo Soto, Jan van Riggelen,
Harald zur Hausen, and Frank Rösl*
Forschungsschwerpunkt Angewandte Tumorvirologie, Abteilung Tumorvirus-Immunologie, Deutsches
Krebsforschungszentrum, Heidelberg, Federal Republic of Germany
Received 3 July 2001/Accepted 28 September 2001

Downloaded from http://jvi.asm.org/ on June 8, 2015 by RUTGERS UNIVERSITY


In the present study we show that malignant human papillomavirus (HPV)-positive cells lost their ability to
synthesize endogenous beta interferon (IFN-␤) upon tumor necrosis factor alpha (TNF-␣) treatment. IFN-␤
transcription, however, was reinducible in nonmalignant HPV-positive cells, which was confirmed in functional
protection assays against encephalomyocarditis virus or vesicular stomatitis virus infections. Addition of
neutralizing antibodies against IFN-␤ blocked the antiviral effect, excluding the possibility that other IFN types
were involved. Conversely, both malignant and immortalized cells could be protected against viral cytolysis
when either IFN-␤, IFN-␣, or IFN-␥ was added exogenously. This indicates that only the cross talk between
TNF-␣ and the IFN-␤ pathways, and not IFN-␣/␤ and IFN-␥ signaling in general, is perturbed in cervical
carcinoma cells. Notably, full virus protection was restricted exclusively to nonmalignant cells, indicating that
the antiviral effect correlates with the growth-inhibitory and virus-suppressive properties of TNF-␣. The
IFN-regulatory factors IRF-1 and p48 (ISGF3␥) emerged as key regulatory molecules in the differential IFN-␤
response, since their transcription was either absent or only inefficiently enhanced in tumorigenic cells upon
treatment with TNF-␣. Inducibility of both genes, however, became reestablished in cervical carcinoma cells,
which were complemented to nontumorigenicity after somatic cell hybridization. Complementation was par-
alleled by the entire reconstitution of cytokine-mediated IFN-␤ expression and the ability of TNF-␣ to exert an
antiviral state. In contrast, under conditions where tumor suppression was not accomplished upon somatic cell
hybridization, neither expression of IRF-1, p48, and IFN-␤ nor antiviral activity could be restored.

Human papillomavirus (HPV)-induced carcinogenesis is a outcome of neoplasias, all cervical carcinoma lines tested up to
multistep process which is initiated by viral infection (68). At now are devoid of significant inducible MCP-1 expression.
what time and to what extent tumor formation takes place, Cytokine inducibility, however, can be completely restored in
however, are dependent mainly on the immunological status of somatic cell hybrids with normal cells (51), which were no
the patient. As deduced from epidemiological data, immuno- longer tumorigenic when heterotransplantated into immuno-
suppressed individuals or persons with impaired immunocom- compromised animals (61). Tumorigenic segregants derived
petence have a significantly higher risk for developing cervical from the same hybrids again lost MCP-1 expression (28, 51),
cancer than corresponding age-matched controls (53). Hence, strongly suggesting that elimination of chemokine expression
tumor appearance can be regarded in part as the result of an may provide a selective advantage for tumor formation (52). It
immunological escape process during which either certain in- should be stressed that this correlation is not restricted to
ter- and intracellular surveillance mechanisms are functionally tissue culture conditions, because in situ hybridization studies
abolished or HPV-positive cells are no longer susceptible to in combination with immunohistochemistry techniques con-
immunological control (69). It is therefore reasonable to as- firmed that MCP-1 expression and infiltrating cells of the
sume that only a physiologically intact communication pathway monocyte/macrophage lineage were detectable only in prema-
between inflammatory and HPV-containing cells guarantees a lignant precursor cells and were absent in high-grade lesions of
proper antiviral response (52). cancer patients (29, 46).
Indeed, when fresh biopsies from patients were evaluated by Recruitment and activation of macrophages can be consid-
immunohistochemistry, cervical cancer sections were found to ered the first line of defense against generalized virus infec-
be significantly depleted of infiltrating macrophages, T lym- tions and viral spread (for reviews, see references 17 and 19).
phocytes, and dendritic cells compared with premalignant tis- For example, spontaneous regression of benign warts is accom-
sue specimens (22, 60, 63, 64). The recruitment of immuno- panied by a strong infiltration of mononuclear cells (41), where
logical effector cells in turn is mediated by chemokines papilloma shrinkage directly correlates with high tumor necro-
(chemotactic cytokines) such as monocyte-chemotactic pro- sis factor alpha (TNF-␣) expression in surrounding macro-
tein-1 (MCP-1) (39, 45, 47). Consistent with the notion that phages (20). Hence, TNF-␣ not only may represent a key
dysregulation in intercellular communication may favor the regulatory cytokine in regression of benign tumors (20) but
also could play a pivotal role in the immunological control of
dysplastic cervical lesions infected with high-risk HPV types
* Corresponding author. Mailing address: Angewandte Tumorvi-
rologie, Deutsches Krebsforschungszentrum, Im Neuenheimer Feld
such as HPV type 16 (HPV16) or HPV18. Although not yet
242, 69120 Heidelberg, Federal Republic of Germany. Phone: 49- directly demonstrated in patients, it is conceivable that mac-
6221-42-4900. Fax: 49-6221-42-4902. E-mail: F.Roesl@DKFZ.de. rophage-specific TNF-␣ synthesis can trigger paracrine MCP-1

280
VOL. 76, 2002 ABSENCE OF IFN-␤ EXPRESSION IN CERVICAL CARCINOMA CELLS 281

gene expression in immortalized cells (and vice versa) which in specificity of endogenous IFN-␤ production were assessed in
turn augments mononuclear cell infiltration as well as secretion antiviral protection assays using encephalomyocarditis virus
of larger amounts of growth-inhibitory cytokines. In support of (EMCV) or vesicular stomatitis virus (VSV) as infectious
this assumption was the finding that cocultivation with acti- agents. In contrast, all malignant cells remained protected
vated macrophages from normal human volunteers can both against viral cytolysis when IFN-␤ was exogenously supple-
suppress viral transcription and induce MCP-1 gene expres- mented. This indicates that the disturbance of the TNF-␣-
sion, but only in nonmalignant HPV-positive cells (51). Con- mediated IFN-␤ expression and the loss of an immediate an-
versely, cervical carcinoma cells were completely refractory, tiviral response are additional central events in the multistep
despite functional TNF-␣ signaling, as monitored by rapid progression to cervical cancer, being more the consequence of
proteolysis of I␬B␣ upon cytokine application (13). The block- the in vivo phenotype of the respective host cell rather than a
age of MCP-1 synthesis and the concomitant resistance to direct effect of E6 and E7 oncoprotein expression.
TNF-␣ may therefore perturb the intercellular cross talk and

Downloaded from http://jvi.asm.org/ on June 8, 2015 by RUTGERS UNIVERSITY


favor the accumulation of malignant cells (52). MATERIALS AND METHODS
Another interesting property of TNF-␣ is its ability to confer Cell lines, hybrid formation, and cytokine treatment. The cervical carcinoma
an antiviral state through induction of the beta interferon cell lines HeLa, SW756, CaSki, and SiHa; the nontumorigenic somatic cell
(IFN-␤) gene (23, 38). IFN-␣/␤ production is the most rapid hybrids made between HeLa cells and normal human fibroblasts (444 cells) and
their tumorigenic segregants (CGL3 cells) (61); HeLa ⫻ CaSki and HeLa ⫻
host immune response against a variety of viral infections (9).
SW756 hybrids (15, 59); SiHa ⫻ HeLa hybrids (see below); malignant HPK Ia
Moreover, it also became evident that the elimination of virus- cells (10); and the human lung carcinoma cell line A549 (66) were maintained in
positive cells is not entirely exerted through major histocom- Dulbecco’s modified Eagle’s medium supplemented with 10% fetal calf serum
patibility complex class I-restricted CD8⫹ cytotoxic T lympho- and 1% penicillin-streptomycin. Modified SiHa cells (so-called universal fuser)
cytes but also relies on appropriate temporal and local (50) harboring dominant (G418 resistance) and recessive (hypoxanthine-phos-
phoribosyltransferase-negative) phenotypes were grown in the same medium
expression of cytokines such as TNF-␣ and downstream IFN containing 1 mg of G418 (Gibco BRL) per ml and 10⫺4 M 6-thioguanine
signaling (19, 35). (Sigma). Somatic cell hybridization between SiHa and HeLa cells was done
Considering this scenario in the context of the transforming exactly as described previously (50). For cytokine treatment, the cells were
potential of high-risk HPVs, there are a quite substantial num- incubated with TNF-␣ (Strathman Biotech GmbH, Hannover, Germany), IFN-␣
(Alphaferon3 [natural human IFN-␣; kindly provided by Rentschler, Laupheim,
ber of studies which show that the IFN signal transduction
Germany]), IFN-␤ (IFN-␤1; [natural human IFN-␤; Rentschler), and IFN-␥
pathway is a target for viral oncoproteins E6 and E7. For (IF-RC [Thomae, Biberach, Germany]) for different periods of time as described
example, HPV18 E6 can interfere with the IFN-␣ response in in the figure legends.
human fibrosarcoma HT1080 cells by impairing Jak-STAT1/2 RNA analysis. RNA was isolated with the RNeasy kit according to the instruc-
tyrosine phosphorylation after binding of the ligand to the tions of the manufacturer (Qiagen, Hilden, Germany). Approximately 5 ␮g of
RNA was separated on 1% agarose gels in the presence of ethidium bromide
corresponding receptor (32). Furthermore, E7 expression in under nondenaturing conditions (26) and transferred to GeneScreen Plus mem-
spontaneously immortalized human keratinocytes (HaCaT branes (DuPont, NEN). Specific probes for the hybridization were labeled with
cells) can inhibit p48 (ISGF3␥) protein translocation into the [32P]dCTP by random priming (12). The filters were washed in 2⫻ SSC (1⫻ SSC
nucleus (3). p48 (ISGF3␥) is part of IFN-stimulated gene fac- is 0.15 M NaCl plus 0.015 M sodium citrate)–0.1% sodium dodecyl sulfate (SDS)
at 68°C and exposed to Kodak films as indicated in the figure legends.
tor 3 (ISGF3), a trimeric trans-activating complex formed be-
RT-PCR. DNA digestion of total RNA was carried out with 2 U of RQ1-
tween p48, STAT1, and STAT2 which induces transcription of DNase (specific activity, 1 U/␮l) (Promega, Mannheim, Germany) in 40 mM
IFN-regulated genes via binding to cognate IFN-stimulated Tris-HCl (pH 8.0)–10 mM MgSO4–1 mM CaCl2 in the presence of 20 U of
response elements (5). However, all of these properties of the RNase inhibitor (specific activity, 40,000 U/ml) (Hybaid, Heidelberg, Germany)
viral oncogenes do not provide a reliable explanation of why in a total volume of 100 ␮l at 37°C for 10 min. DNase was inactivated by addition
of 2 ␮l of 0.5 M EDTA (pH 8.0), followed by acidic phenol-chloroform extraction
IFN still has a curative and growth-inhibitory effect on prema- (8). RNA was diluted in RNase-free water. One microgram of DNase-treated
lignant HPV-positive cells (6, 14, 25), despite the fact that E6 RNA was mixed with 0.5 ␮g of oligo(dT) primer (Promega) added in a total
and E7 are found to be expressed in those lesions. volume of 12 ␮l, heated at 70°C for 10 min, and chilled on ice. The mixture was
To gain insight into IFN signaling and the antiviral activity of supplemented with 5⫻ reverse transcription (RT) buffer (250 mM Tris-HCl [pH
8.3], 375 mM KCl, 15 mM MgCl2), 10 mM dithiothreitol, 500 ␮M deoxynucleo-
TNF-␣, we used cervical carcinoma cells and malignant and
side triphosphate mix (Roche Diagnostic, Mannheim, Germany), and 20 U of
nonmalignant somatic cell hybrids derived therefrom as a RNase inhibitor and incubated at 42°C for 2 min in a total volume of 19 ␮l. After
model system (61). For example, HPV18-positive HeLa cells the annealing, 200 U of reverse transcriptase SuperScript II (Gibco BRL) (spe-
can be complemented to nontumorigenic phenotype growth cific activity, 200 U/␮l) was added and the reaction mixture was incubated for 1 h
after somatic cell hybridization with normal human fibroblasts at 42°C. The RT products were heated to 72°C for 10 min and chilled on ice.
PCRs were performed in a solution containing 10 mM Tris-HCl (pH 8.3), 200
or keratinocytes. Occasionally, however, rare tumorigenic seg- ␮M deoxynucleoside triphosphate mix (Roche Diagnostic), 40 pmol of upstream
regants of the same hybrids arise, which is accompanied by the and downstream primers, 5 U of Taq polymerase (Sigma) (specific activity, 5
nonrandom loss of chromosomes, in particular of chromosome U/␮l), and 2 ␮l of reverse-transcribed product. The amplification was performed
11 (for a review, see reference 52). In the case of HPV18- in an MJ Research PTC-200 thermal cycler in a total volume of 50 ␮l. For IFN-␤
detection, the primers 5⬘-GATTCATCTAGCACTGGCTTG-3⬘ and 5⬘-CTTCA
positive cervical carcinoma cells, chromosome 11 indeed seems
GGTAAATGCAGAATCC-3⬘ (23) were used. The PCR was performed for 40
to play a key role in malignant transformation, since the rein- cycles consisting of 1 min at 94°C, 45 s at 55°C, and 30 s at 72°C. During the last
troduction of the corresponding normal allele via microcell cycle, the extension time was increased to 10 min, and the reaction mixture was
transfer is sufficient to completely suppress tumor formation of rapidly cooled to 4°C. IFN-regulatory factor 3 (IRF-3) expression was monitored
either the parental cells or the tumorigenic segregants (52a). using the upstream primer 5⬘-GGTTGCGTTTAGCAGAGGAC-3⬘ and the
downstream primer 5⬘-AGGAGATGGTCTGCTGGAAG-3⬘. Amplification (35
Here we demonstrate that, similar to the case for the MCP-1 cycles) was done at 94°C for 30 s, 57°C for 45 s, and 72°C for 30 s. In the last cycle,
gene (28, 51), IFN-␤ expression could be induced by TNF-␣ the extension time was increased (to 10 min) and then the mixture was rapidly
only in nonmalignant HPV-positive cells. The functionality and cooled to 4°C. GAPDH (glyceraldehyde-3-phosphate dehydrogenase) was used
282 BACHMANN ET AL. J. VIROL.

as an internal control with primers 5⬘-TGGATATTGTTGCCATCAATGAC


C-3⬘ and 5⬘-GATGGCATGGACTGTGGTCATG-3⬘ (18). The amplification
was performed for 35 cycles of 1 min at 94°C, 45 s at 65°C, and 30 s 72°C, with
a prolonged extension time of 10 min in the last cycle before cooling to 4°C. The
PCR products were analyzed in 1 to 2% agarose gels.
Antiviral titration assays. Cells were infected with either EMCV or VSV using
multiplicities of infections (MOIs) of between 0.1 and 0.2 (37, 66). The cells,
seeded in 96-well plates (104 cells/well) were pretreated with cytokines as de-
scribed in the figure legends. After 24 h, the medium was discarded, fresh
medium (37°C) with virus was added, and the plates were incubated at 37°C until
the viral lysis in the controls was complete (16 to 20 h). All cells were susceptible
to EMCV, except CaSki ⫻ HeLa hybrids. For those cells, antiviral activity was
monitored after infection with VSV. To quantify the cytopathic effect (CPE), the
cells were fixed with 4% glutardialdehyde and stained with 1% crystal violet. The

Downloaded from http://jvi.asm.org/ on June 8, 2015 by RUTGERS UNIVERSITY


dye was solubilized in 33% acetic acid, and the optical density of the eluate was
measured at 570 nm in a Labsystem Multiscan MS enzyme-linked immunosor-
bent assay reader. To quantify the amount of endogenously synthesized IFN-␤ in
444 cells after treatment with TNF-␣, the supernatants were transferred on A549
indicator cells (66) (3 ⫻ 104/well). On the next day, the cells were infected with
EMCV. After 26 to 30 h, medium was removed and the cytopathogenic effect was
quantified as described above. The amounts of IFN-␤ are expressed as interna-
tional reference units per milliliter, using exogenously added National Institutes
of Health human IFN-␤ as a reference.
Neutralization assay. Cells (104) were seeded in 96-well plates. Prior to the
addition of TNF-␣, as indicated in the figure legends, the cells were incubated for
1 h at 37°C with antibodies neutralizing 8 U of IFN-␤ (clone 39C; Biotrend, Köln,
Germany) or 10 U of IFN-␣ (Hoffmann La Roche Inc., Basel, Switzerland).
After 24 h, medium was discarded and the cells were infected with EMCV and
incubated at 37°C until viral controls showed complete lysis. Cells were fixed with
4% glutardialdehyde and stained with 1% crystal violet. The eluate was quanti-
fied as described above.
EMSAs. For gel retardation, the following oligonucleotides were used: the
positive regulatory domain II (PRDII) sequence 5⬘-GGGAAATTCCGGGAA
ATTCC-3⬘, which contains two copies of the PRDII core element (1); the
PRDIV sequence 5⬘-AATGTAAATGACATAGGAAAACTGA-3⬘ (33); the
PRDIII-I sequence 5⬘-GAAAACTGAAAGGGAGAAGTGAAA-3⬘ (33); and FIG. 1. Selective IFN-␤ induction by TNF-␣ in nonmalignant
an NF-␬B sequence, 5⬘-AGTTGAGGGGACTTTCCCAGGC-3⬘, derived from HPV18-positive cells, showing intactness of the MAPK pathway. (A)
the immunoglobulin ␬ light-chain gene (44). The DNAs were synthesized in an RT-PCR products generated by IFN-␤- and GAPDH-specific primer
Applied Biosystems synthesizer using phosphoramitide chemistry and further sets (185 and 460 bp, respectively) were separated on 2% agarose gels.
purified by high-pressure liquid chromatography. For electrophoretic mobility The superimposed ethidium bromide-stained gel shows the quality of
shift assays (EMSAs), the annealed oligonucleotides were labeled with the RNAs used for the RT-PCR. The positions of the 18S and 28S
[␥-32P]ATP (Amersham; 3,000 Ci/mmol) with T4 polynucleotide kinase and gel rRNAs are indicated. Nonmalignant 444 hybrids, their tumorigenic
purified from a 15% polyacrylamide gel. Cellular extracts were prepared as segregants (CGL3 cells), and parental HeLa cells were incubated with
described previously (54), with the only modification being that N,N-(L-3-trans- 10 ng of TNF-␣ per ml for different periods of time. Lanes (⫺),
carboxyoxirane-2-carbonyl)-L-leucyl-agmatine (E64), 4-(2-aminoethyl)-benzol- untreated controls. (B) Western blot analysis of p38 MAPK. Twenty
sulfonylfluoride (Pefabloc SC), 1 mM NaF, and 0.2 mM Na3VO4 were included micrograms of total cellular extract per lane was separated in two
as protease inhibitors in concentrations suggested by the manufacturer (Roche). identical SDS–10% polyacrylamide gels. After transfer, the filters were
The protein concentration was determined by the Bradford method (Bio-Rad) incubated with either a phosphorylation-specific (Phospho-p38MAPK)
using defined amounts of bovine serum albumin as standards. The binding of or a phosphorylation-unspecific (p38MAPK) p38 antibody. Equal load-
NF-␬B, activation factor-2 (ATF-2), and c-Jun was performed in a 20-␮l reaction ing and protein transfer were confirmed by incubating the upper filter
volume containing 10% glycerol, 12 mM HEPES (pH 7.9), 4 mM Tris-HCl (pH with an actin-specific antibody. The molecular masses are indicated.
7.9), 60 mM KCl, 1 mM EDTA, 1 mM dithiothreitol, 0.6 mg of bovine serum
albumin per ml, 2.0 ␮g of poly(dI-dC), and 2 to 4 ␮g of nuclear extract. After 5
min, 10,000 cpm of the [␥-32P]ATP 5⬘-end-labeled double-stranded oligonucle-
otide probe was added, and the incubation was continued for an additional 30 an IRF-1 antibody specific for the C-terminal region of human IRF-1 (sc-497x),
min at room temperature (58). EMSAs for the IRF family were performed in an IRF-2 antibody specific for the carboxy terminus of human IRF-2 (sc-498x),
20-␮l reaction volumes containing 10% glycerol, 70 mM Tris (pH 7.5), 30 mM Na an IRF-3 antibody with an epitope corresponding to full-length IRF-3 of human
acetate, 10 mM EDTA, 1 ␮g of poly(dI-dC), and 5 ␮g of nuclear extract. After origin (sc-9082x), and an IRF-7 antibody which recognizes amino acids 1 to 246
5 min, 10,000 cpm of the [␥-32P]ATP 5⬘-end-labeled double-stranded oligonu- of human IRF-7 (sc-9083x). Antibody to p48 (ISGF3␥) was from Transduction
cleotide probe was added, and the incubation was continued for an additional 15 Laboratories. The DNA-protein complexes were resolved on 5.5% nondenatur-
min at 4°C (7a). The sequence specificity of the binding was routinely controlled ing polyacrylamide gels (29:1 cross-linking ratio), dried, and exposed overnight to
in competition experiments by the addition of a 100-fold molar excess of either Fuji medical X-ray films.
unlabeled homologous or heterologous oligonucleotides. For monitoring the SDS-polyacrylamide gel electrophoresis and Western blotting. The same ex-
c-Jun and ATF-2 composition in supershift assays, 2 ␮g of polyclonal antibodies tracts (25 to 50 ␮g) used for the band-shift analyses were separated in 8 to 10%
directed against c-Jun or ATF-2 was added and the reaction mixture was further SDS-polyacrylamide gels, electrotransferred to polyvinylidene difluoride mem-
incubated for 1 h at 4°C. Selective binding of different IRF members was ana- branes (Immobilon-P; Millipore Corporation, Bedford, Mass.), and probed with
lyzed by adding 2 ␮g of poly- or monoclonal antibodies to the reaction mixture antibodies to IRF-1 (sc-497x; Santa Cruz), IRF-2 (sc-498x; Santa Cruz), and p48
prior to oligonucleotide supplementation. Specifically, the following antibodies (Transduction Laboratories). The incubation was carried out overnight in Tris-
(all obtained from Santa Cruz Biotechnology as TransCruz supershift reagents) buffered saline supplemented with 5% skim milk powder (Merck), 0.05% Tween
were used: a c-Jun antibody which recognizes both the nonphosphorylated and 20 (Sigma), and a 1:5,000 (IRF-1 and IRF-2) or 1:500 (p48) dilution of antibod-
phosphorylated forms of c-Jun (epitope corresponding to amino acids 56 to 69 ies. p38 and phospho-p38 mitogen-activated protein kinase (MAPK) were di-
mapping within the amino-terminal domain of the mouse c-Jun protein) (sc- rectly monitored in total cellular extracts obtained after lysis in SDS-polyacryl-
822x), an ATF-2 antibody specific for Thr-71-phosphorylated ATF-2 (sc-8398x), amide gel electrophoresis buffer (60 mM Tris, 2% SDS, 10% glycerol, 50 mM
VOL. 76, 2002 ABSENCE OF IFN-␤ EXPRESSION IN CERVICAL CARCINOMA CELLS 283

dithiothreitol, 0.1% bromophenol blue, pH 6.8). Antibody dilutions were used as


recommended by the supplier (New England Biolabs, Frankfurt, Germany). The
bands were visualized with an anti-rabbit immunoglobulin G antibody conju-
gated with horseradish peroxidase using the ECL detection system (Amersham).
Equal protein transfer and loading were routinely controlled by reincubating the
filters with a monoclonal actin-specific antibody (ICN Biomedicals). For reincu-
bation with additional antibodies, the filters were stripped with 200 mM NaOH
for 5 min at room temperature.

RESULTS

Restoration of the TNF-␣-induced antiviral response in


nonmalignant HPV18-positive somatic cell hybrids: reexpres-

Downloaded from http://jvi.asm.org/ on June 8, 2015 by RUTGERS UNIVERSITY


sion of the IFN-␤ gene. To monitor the antiviral effect of
TNF-␣ on the IFN-␣/␤-regulatory pathway, we used HPV18-
positive HeLa cells and derived somatic cell hybrids as an
experimental model system (61). Tumorigenic HeLa cells can
be reverted to nonmalignant growth when fused to primary
human fibroblasts (designated 444 cells). Long-term in vitro
cultivation, however, leads to rare tumorigenic segregants of
the same hybrids (referred to as CGL3 cells), which permits
the investigation of cytokine signaling in a cellular environ-
ment harboring identical transcription cassettes of HPV18 (49)
but where the viral oncoproteins E6 and E7 are expressed in a
different genetic background. When such cells were treated
with TNF-␣, IFN-␤ mRNA could be detected as a 185-bp
RT-PCR fragment in nonmalignant hybrids (444 cells) which
became discernible as saturated quantities 4 h after TNF-␣
application and persisted for at least 24 h. When the same
experiment was carried out with RNAs obtained from malig-
nant segregants (CGL3 cells) or with parental HeLa cells, no
IFN-␤ expression occurred (Fig. 1A). The selective inducibility
of the IFN-␤ gene could not be attributed to a disturbance of
the TNF-␣ signal response or receptor engagement in malig-
nant cells, because p38MAPK MAPK phosphorylation, a known
hallmark for functional proinflammatory cytokine signaling
(65), became visible and disappeared with approximately the
same kinetics in all three cell lines without quantitative
changes of the net amount of nonphosphorylated p38 (Fig.
1B). Identical loading and protein transfer after Western blot-
ting were assessed by reincubating the same filter with an
actin-specific antibody.
To verify that the IFN-␤-specific mRNA was translated into
functional protein, a biological assay based on the susceptibil-
ity of nonresponsive cells to lysis after infection with EMCV
was used (37, 66) (Fig. 2). After being seeded in 96-well plates,
the cells were first preincubated with different amounts of
TNF-␣ ranging from 10 to 2.5 ng/ml. Only 444 cells were
protected (Fig. 2A), while both HeLa cells (Fig. 2B) and the
CGL3 tumorigenic segregants (Fig. 2C) were completely sen-
sitive to EMCV-mediated cell lysis, even at higher TNF-␣
FIG. 2. TNF-␣ confers selective protection against EMCV infec-
tion in nontumorigenic HPV18-positive cells. Malignant and nonma-
lignant cells grown in microtiter plates were first pretreated for 24 h
with serial dilutions (1:2, indicated by the arrow) of TNF-␣ (10, 5, and tected cells relative to the untreated and the TNF-␣-incubated cell
2.5 ng/ml) and infected with EMCV at an MOI of 0.1 as described in controls (cc and Tc, respectively). (E) Determination of IFN-␤ syn-
Materials and Methods. To monitor the CPE, the cells were fixed and thesis in 444 cells. Supernatants (in twofold serial dilutions) were
stained with crystal violet. cc, uninfected control cells; vc, EMCV- added to A549 indicator cells, and antiviral activity was determined as
infected cells without prior TNF-␣ addition; Tc, TNF-␣ treated cells described in Materials and Methods. The amounts of IFN-␤ produced
not infected with EMCV. (A to C) 444, HeLa, and CGL3 cells, re- at different periods of time (4, 8, and 16 h) after treatment with 10 ng
spectively. (D) Quantification of the CPE after staining with crystal of TNF-␣ per ml are expressed as international units per milliliter. All
violet. The dye was eluted, and its absorbance at 570 nm was deter- standard deviations are given for three independent experiments per-
mined spectroscopically. The bars indicate the percentages of pro- formed in triplicate.
284 BACHMANN ET AL. J. VIROL.

Downloaded from http://jvi.asm.org/ on June 8, 2015 by RUTGERS UNIVERSITY


FIG. 3. EMCV protection is mediated by IFN-␤. 444 cells were
pretreated with different concentrations of TNF-␣ prior to EMCV
infection in the presence of antibodies sufficient to neutralize IFN-␣
(␣), IFN-␤ (␤), or both (␣ ⫹ ␤) as described in Materials and Methods.
cc, uninfected control cells; vc, EMCV-infected cells without prior
TNF-␣ addition; Tc, TNF-␣-treated cells not infected with EMCV.

concentrations. These data illustrate that the antiviral activity


of TNF-␣ was reconstituted in nonmalignant hybrids even in
the presence of the viral oncoproteins but was lost in tumori-
genic segregants or parental HeLa cells. To estimate how
much IFN-␤ was definitively synthesized, the supernatants of
nonmalignant hybrids were removed and tested for IFN-␤ ac-
tivity against EMCV on A549 indicator cells, where TNF-␣
itself had no antiviral activity (66). Concordant with the RT-
PCR data (Fig. 1A), biologically active IFN-␤ was first detect-
able after 4 h, leading to an average accumulation of IFN-␤
ranging between 18 and 33 IU at 16 h after TNF-␣ adminis-
tration (Fig. 2E). To ensure that endogenous IFN-␤ synthesis
was responsible for the protective effect against EMCV infec-
tion, the same assay was performed in the presence of neutral-
izing IFN-␤ antibodies added 1 h prior to TNF-␣ supplemen-
tation. Figure 3 shows that only the addition of antibodies FIG. 4. Overall protection of HPV18-positive cells after exogenous
against IFN-␤, and not addition of an IFN-␣-specific anti- IFN-␤ supplementation. Malignant and nonmalignant cells grown in
serum, significantly inhibited the TNF-␣-induced antiviral ac- microtiter plates were first pretreated for 24 h with serial dilutions (1:2,
tivity. This supports the notion that IFN-␤ was in fact the key indicated by the arrow) of IFN-␤ (100, 50, and 25 U) and infected with
effector protein which selectively protected nontumorigenic EMCV at an MOI of 0.1 as described for Fig. 2. (A to C) 444, HeLa,
and CGL3 cells, respectively. (D) Quantification of the degree of
cells against EMCV infection. protection. The bars indicate the percentages of IFN-␤-protected cells
Exogenous IFN-␤ supplementation protects HPV-positive relative to untreated and IFN-␤-incubated control cells used as a
cells from EMCV infection independently from the in vivo reference (indicated as cc and Tc, respectively). Standard deviations
phenotype. Since it has been reported that the expression of are given for three independent experiments performed in triplicate.
viral oncogenes can interfere with IFN signaling (for a review,
see reference 30), it was mandatory to examine whether ma-
lignant and nonmalignant cells still reacted selectively when IRF-1 and p48 (ISGF3␥) are selectively induced in nonma-
IFN-␤ was added exogenously. In this case, all cell lines could lignant cells upon TNF-␣ treatment. To gain further insight
be protected against EMVC-mediated cytolysis independently into the differential regulation of the IFN-␤ gene in nonma-
of whether IFN-␤ (Fig. 4) or IFN-␣ or IFN-␥ (A. Bachmann et lignant hybrids, we next examined the transcription factors
al., unpublished observation) was added to the tissue culture involved in IFN-␤ induction. One major key regulatory factor
medium. Note that protection was successful even after appli- which is activated after addition of proinflammatory cytokines
cation of less than 25 U of IFN-␤ per ml, which was in agree- is NF-␬B (31). NF-␬B binds to the IFN-␤ promoter as a part of
ment with our preceding data measuring the bioavailability of a coordinately assembled multiprotein complex called the en-
444 cell-secreted IFN-␤ at 16 h after TNF-␣ addition (compare hanceosome (for a review, see reference 36). At the DNA
Fig. 2E and 4A). Contrary to previous observations (3, 32, 42, level, the IFN-␤ upstream region is composed of four PRDs
48), the present data unambiguously demonstrate that, as far (PRDI to -IV) which act synergistically in stimulating tran-
as the antiviral activity is concerned, both IFN-␣/␤ and IFN-␥ scription (17, 24, 36) (Fig. 5A shows a schematic overview).
can still protect HPV18-positive cells against EMCV infection Using parts of PRDII in comparison with an NF-␬B site ob-
(Fig. 4D), even after ongoing oncogene expression (see Dis- tained from the immunoglobulin ␭ light-chain gene (44) for
cussion). EMSAs, both oligonucleotides showed approximately the
VOL. 76, 2002 ABSENCE OF IFN-␤ EXPRESSION IN CERVICAL CARCINOMA CELLS 285

same affinity in all three cell lines investigated. NF-␬B binding


already became discernible 30 min after TNF-␣ addition (Fig.
5B), which makes it unlikely that the absence of IFN-␤ induc-
ibility was due to a failure in cytokine signaling towards the
NF-␬B branch. When the same set of experiments were carried
out with PRDIV containing the cis-regulatory sequences for
c-Jun and ATF-2, a more complex binding pattern appeared.
When the origins and specificities of the various bands were
examined by addition of c-Jun (Fig. 5C) and phosphorylation-
specific ATF-2 (Fig. 5D) antibodies in supershift EMSAs, both
transcription factors again revealed roughly the same binding
kinetics.

Downloaded from http://jvi.asm.org/ on June 8, 2015 by RUTGERS UNIVERSITY


A completely different picture emerged when we monitored
the expression of IRF-1, which was originally identified as a
critical mediator of the IFN response (for a review, see refer-
ence 62). As depicted in Fig. 6A, TNF-␣ treatment led to a
strong induction of IRF-1 transcription exclusively in nontu-
morigenic hybrids (444 cells), whereas the gene was only mar-
ginally elevated in the malignant cells (CGL3 and HeLa cells).
The effect became even more pronounced when IRF-1 expres-
sion was examined by Western blot analysis (Fig. 6B). When
EMSAs were performed with oligonucleotides encompassing
the PRDIII-I region (Fig. 5A), significant IRF-1 binding was
obtained only with nuclear extracts derived from TNF-␣-
treated 444 cells (Fig. 6C, left). The authenticity of IRF-1 was
verified after addition of a specific antibody, which leads to a
disappearance of the induced band. IRF-3, IRF-7, and p48 do
not bind at this region after short-term TNF-␣ application (4
h), since the binding pattern was not affected after addition of
the respective antibodies (Fig. 6C, right). Conversely, consis-
tent with the EMCV infection assays described above (Fig. 4),
IRF-1 could be activated independent of the cell phenotype
when either IFN-␤ or IFN-␥ was exogenously supplemented
(Fig. 6D and E, respectively). No selective induction or over-
expression in tumorigenic cells was seen in analysis of the
expression of IRF-2 (Fig. 7A), a transcription factor which
antagonizes the function of IRF-1 by competing for the same
binding site (PRDI and -II) (for a schematic overview, see Fig.
5A) (62). The same was true for IRF-3 (2), encoded by a
transcriptional activator gene, which was constitutively tran-
scribed and not further stimulated after TNF-␣ application
(Fig. 7B).
p48 (ISGF3␥) is another IRF family transcription factor and
is the major binding component of ISFG3 (a multimeric com-
plex between STAT1 and -2) (5). Although p48 (ISGF3␥)
seems to be more involved in the autocrine amplification of the
so-called delayed IFN-␣/␤ response (24), knockout experi-

FIG. 5. EMSAs of NF-␬B, c-Jun, and ATF-2 at the IFN-␤ up-


stream regulatory region. (A) Schematic overview of the IFN-␤-regu-
latory region (enhanceosome). I to IV, PRDs and the relative binding
positions of c-Jun; ATF-2; IRF-1, -2, -3, and -7; and NF-␬B (p50/p65).
(B to D) HeLa, 444, and CGL3 cells were treated with TNF-␣ for 30
min and 4 h. Lanes (⫺), untreated controls. Nuclear extracts were
prepared for EMSAs with a PRDII and NF-␬B probe (B) or an
oligonucleotide probe harboring PRDIV (C and D). PRDIV EMSAs
were performed either in the absence (⫺) or in the presence (⫹) of
supershift antibodies directed against c-Jun (C) or phosphorylated
ATF-2 (D). Specific bands are marked by an arrow. Asterisks indicate
nonspecific binding.
286 BACHMANN ET AL. J. VIROL.

Downloaded from http://jvi.asm.org/ on June 8, 2015 by RUTGERS UNIVERSITY


FIG. 7. IRF-2 and IRF-3 expression in HPV18-positive cells. (A)
IRF-2 Western blot analysis of nuclear extracts (50 ␮g of protein/lane)
of each cell line treated with TNF-␣ as indicated. After electrotransfer,
the same filters were consecutively incubated with antibodies raised
against IRF-2 and actin. (B) RT-PCR products of IRF-3 and GAPDH
(399 and 460 bp, respectively) after separation on 2% agarose gels. The
quality of the RNAs used for the RT-PCR is shown at the top. The
positions of the 18S and 28S rRNAs are indicated. Cells were incu-
bated for 16 h in the presence of 10 ng of TNF-␣ per ml. Lanes (⫺),
untreated controls.

ments have revealed that p48⫺/⫺ mice were impaired in their


antiviral activity against EMCV and VSV infections (27). To
assess the role of p48 (ISGF3␥) in the perturbation of TNF-
␣-mediated IFN signaling in our experimental system, p48
expression was examined. As depicted in Fig. 8A, only nonma-
lignant cells revealed significant p48 (ISGF3␥) mRNA levels at
4 and 8 h after TNF-␣ treatment, while the gene remained
transcriptionally silent in the malignant counterparts. Since it
has been reported that E7 of HPV16 can potentially interfere
with p48 (ISGF3␥) translocation into the nucleus (3), cells
were fractionated and the nuclear extracts were monitored by
Western blot analysis. To exclude cytoplasmic contamination,
the quality of nucleus-cytoplasm separation was controlled by
incubation of the filter with a cytoplasmic (pyruvate kinase
M2) marker protein (Bachmann et al., unpublished data) (34).
Similar to the case for IRF-1 (Fig. 6B), nuclear p48 (ISGF3␥)
accumulation occurred only in nonmalignant cells (Fig. 8B),
while gene expression was inducible with similar kinetics in all
three cell line when IFN-␤ or IFN-␥ was administered (Fig.
8C).
FIG. 6. Selective IRF-1 induction by TNF-␣ in nonmalignant Complementation of a nontumorigenic phenotype between
HPV18-positive cells. (A) Transcriptional analysis of IRF-1 after
TNF-␣ treatment for 0.5, 1, 2, 4, and 8 h. Total RNA (5 ␮g/lane) was different cervical carcinoma cell line restores antiviral activity.
separated on 1% agarose gels. Filters were consecutively hybridized
with probes specific for IRF-1 and ␤-actin. IRF-1 was exposed to
Kodak Biomax film and ␤-actin was exposed to Kodak X-Omat film for
1 day. The positions of the 18S and 28S rRNAs are indicated. (B) TNF-␣ for 1, 4, and 16 h. Lanes (⫺), untreated controls. Right, PRD
Western blot analysis of nuclear extracts (50 ␮g/lane) after TNF-␣ III-I EMSAs with TNF-␣-treated (4 h) 444 extracts in the presence of
application. After electrotransfer, the filters were incubated with an- supershift antibodies directed against IRF-1, -2, -3, and -7 and p48.
tibodies directed against IRF-1. Equal loading was assessed with an The specific IRF-1 and supershift bands are indicated by arrows. (D
actin-specific antibody. (C) Left, EMSAs using an oligonucleotide and E) Same as panel A but after treatment with IFN-␤ (10 U/ml) or
probe harboring PRDIII-I. HeLa and 444 cells were treated with with IFN-␥ (10 U/ml). The IFN application was extended to 16 h.
VOL. 76, 2002 ABSENCE OF IFN-␤ EXPRESSION IN CERVICAL CARCINOMA CELLS 287

Downloaded from http://jvi.asm.org/ on June 8, 2015 by RUTGERS UNIVERSITY


FIG. 8. Selective p48 (ISGF3␥) induction by TNF-␣ in nonmalig-
nant HPV18-positive cells. (A) Transcriptional analysis of p48 after
TNF-␣ treatment for 0.5, 1, 2, 4, and 8 h. Total RNA (5 ␮g/lane) was
separated on 1% agarose gels. Filters were consecutively hybridized
with probes specific for p48 (ISGF3␥) and ␤-actin. Exposure was done
on Kodak Biomax films (p48) or Kodak X-Omat films (␤-actin) for 3
days and 1 day, respectively. The positions of the 18S and 28S rRNAs
are indicated. (B) Western blot analysis of nuclear extracts (25 ␮g/
lane) at different times after TNF-␣ application. After electrotransfer,
the filters were incubated with a polyclonal p48 antibody. Equal load-
ing was confirmed with an actin-specific antibody. (C) Same as panel A FIG. 9. Virus protection in nontumorigenic hybrids between cervi-
but after treatment with IFN-␣ (10 U/ml). IFN-␣ application was cal carcinoma cells. (A) CaSki, SW756, SW756 ⫻ HeLa, and CaSki ⫻
extended to 16 h. HeLa hybrids were first pretreated for 24 h with serial dilutions (1:2,
indicated by the arrow) of TNF-␣ (10, 5, and 2.5 ng/ml) and infected
with EMCV at an MOI of 0.1 as described for Fig. 2. CaSki ⫻ HeLa
hybrids were infected with VSV at an MOI of 0.2. cc, uninfected
Based on the preceding experiments, we reasoned that cervical control cells; vc, infected cells without prior TNF-␣ addition; Tc,
carcinoma cells in general may lack TNF-␣-induced antiviral TNF-␣ treated cells not infected with virus. (B) RT-PCRs of RNAs
obtained from SW756, CaSki, and somatic cell hybrids using HeLa
activity, which should correlate with IRF dysregulation and cells as a fusion partner after treatment with TNF-␣ for 16 h. For
tumorigenicity. To test this prediction, HPV16-positive CaSki details, see the legend to Fig. 1A. Lanes (⫺), untreated controls.
cells and HPV18-positive SW756 cells were seeded in 96-well
plates and then tested in EMCV and VSV infection assays.
Both the CaSki and SW756 malignant cell lines were unable to
resist viral infection after pretreatment with TNF-␣ (Fig. 9A, experiments, were partially protected against EMCV infection
upper panels). As shown by RT-PCR analysis, deficiency of after TNF-␣ treatment (ranging between 30 to 40%), but these
viral resistance was again paralleled by an absence of IFN-␤ cells were again completely sensitive when infected with VSV
expression (Fig. 9B). Conversely, as already shown for HeLa (Table 1).
cells (Fig. 3), protection could be achieved when either IFN- In a recent study we have demonstrated that tumorigenicity
␣/␤ or IFN-␥ was exogenously supplemented, which was again of HeLa and CaSki cells can be entirely suppressed after so-
paralleled by IRF-1 and p48 expression (Bachmann et al., matic cell hybridization. In contrast, hybrid formation between
unpublished observations). When other malignant cells, such HeLa and SW756 resulted in cell clones which were still ma-
as tumorigenic variants of in vitro-immortalized HPV16-posi- lignant after heterotransplanation into immunocompromised
tive human keratinocytes (HPK Ia cells) (10) were tested, animals (59). Utilizing this complementation system in the
these cells were also found to be highly sensitive to EMCV and context of TNF-␣-mediated IFN-␤ signaling, antiviral activity
VSV infection (Table 1). Only HPV16-positive SiHa cervical and protection against both EMCV and VSV infection could
carcinoma cells, which had low tumorigenic potential in animal be completely restored in nonmalignant CaSki ⫻ HeLa hy-
288 BACHMANN ET AL. J. VIROL.

TABLE 1. Summary of results of EMCV and VSV protection


studies in relation to the proliferative phenotype
of the cells in nude mice
Cell linea Tumorigenicityb Protection by TNF-␣

HPV-18
444 ⫺ ⫹
␤-444 ⫺ ⫹
CGL3 ⫹ ⫺
HeLa ⫹ ⫺
SW756 ⫹ ⫺

HPV-16
SiHa ⫹ ⫹/⫺

Downloaded from http://jvi.asm.org/ on June 8, 2015 by RUTGERS UNIVERSITY


CaSki ⫹ ⫺
HPK Ia late passage ⫹ ⫺

Hybrids
CaSki ⫻ HeLa ⫺ ⫹c
SW756 ⫻ HeLa ⫹ ⫺
SiHa ⫻ HeLa ⫹ ⫺
a
HPV 16- or 18-positive cervical carcinoma cells and derived cell hybrids.
b
Formation of tumors after subcutanous inoculation of 107 cells in both flanks
of 6-week-old female nude mice (59).
c
CaSki ⫻ HeLa hybrids were susceptible only to VSV infections.

brids, while SW756 ⫻ HeLa hybrids remained sensitive to


EMCV infection after TNF-␣ treatment (Fig. 9A, lower pan-
els). When IRF-1 and p48 (ISGF3␥) transcription in these cell
lines was monitored by Northern blotting, both genes were
found to become significantly reinduced by TNF-␣ in nonma-
lignant CaSki ⫻ HeLa cells but not in SW756 ⫻ HeLa hybrids,
where only marginal transcription was detectable (Fig. 10).
IRF-1 and p48 (ISGF3␥) reexpression resulted in complete FIG. 10. Restoration of IRF-1 and p48 (ISGF3␥) expression in
restoration of IFN-␤ transcription, as confirmed by RT-PCR nonmalignant CaSki ⫻ HeLa hybrids. (A and B) Transcriptional anal-
analyses (Fig. 9B). ysis of IRF-1 in CaSki (A) and SW756 (B) cells and the corresponding
HeLa hybrids after TNF-␣ treatment for 1, 4, and 8 h. Total RNA (5
␮g/lane) was separated on 1% agarose gels. Filters were consecutively
DISCUSSION hybridized with probes specific for IRF-1 and ␤-actin. (C) Same as
panels A and B but after hybridization with a p48 (ISGF3␥)-specific
Extending our previous studies to unravel immunological probe. Exposure was done on Kodak Biomax films (p48) or Kodak
escape mechanisms during HPV-induced carcinogenesis (for a X-Omat films (␤-actin) for 3 days and 1 day, respectively. Lanes (⫺),
untreated controls.
review, see reference 52), we found that the inducibility of the
IFN-␤ gene by TNF-␣ was eliminated in tumorigenic HPV18-
positive HeLa cells but was reconstituted in nonmalignant
HeLa ⫻ fibroblast hybrids (Fig. 1). Restoration of IFN-␤ ex- carcinoma cells. This may have considerable implications for
pression resulted in successful protection of cells against infec- immune evasion processes during progression to cervical can-
tion with EMCV or VSV (37) (Fig. 2). Furthermore, the fail- cer (52, 67). TNF-␣ represents an important regulatory cyto-
ure of TNF-␣ to induce an effective antiviral response in 444 kine with immunomodulatory and growth-inhibitory functions
cells in the presence of neutralizing antibodies against IFN-␤ in nonmalignant HPV-positive keratinocytes (35, 51). TNF-␣
(but not against IFN-␣) confirmed a direct involvement of both suppresses transcription of the viral E6 and E7 oncogenes
autocrine secreted IFN-␤ as an antiviral mediator elicited by and induces the expression of MCP-1, exclusively in nontu-
TNF-␣ (Fig. 3). Of particular interest also was the fact that morigenic cells (51). MCP-1 belongs to a superfamily of small
malignant cells remained protected against virus when either secretory proteins called chemokines (39, 47), which recruit
IFN-␣/␤ or IFN-␥ was directly supplemented into the tissue and activate mononuclear cells, the first line of defense against
culture medium (Fig. 4). These data provide compelling evi- viral infection (45). Activated macrophages in turn not only
dence that IFN signaling and the induction of an antiviral secrete additional TNF-␣, thereby amplifying the cytokine re-
function operate equally well in all cell lines and independently sponse, but also are capable of inducing IFN-␣/␤, which have
from the proliferative phenotype in immunocompromised an- strong antiviral functions, in their target cells (23, 38). It should
imals. be emphasized that IFN-␤ induction represents the earliest
The observation that TNF-␣-mediated induction of IFN-␤ antiviral response which occurs by an protein synthesis-inde-
was restricted to nontumorigenic hybrids strongly suggests that pendent pathway (for reviews, see references 17 and 19). Since
the cross talk between the TNF-␣ pathway towards transcrip- IFN-␣ is not able to be induced in cells lacking both copies of
tional activation of the IFN-␤ gene is disturbed in cervical the IFN-␤ gene (IFN-␤⫺/⫺ cells), it is thought that IFN-␤
VOL. 76, 2002 ABSENCE OF IFN-␤ EXPRESSION IN CERVICAL CARCINOMA CELLS 289

binding to its cognate receptor is a prerequisite for activation gene regulation is presently not understood. Suppression or
of further IFN-␣ production (11). Consequently, loss of both inefficient expression of IRF-1, however, can be epigenetically
TNF-␣-mediated MCP-1 and IFN-␤ inducibility in tumori- modified by different degrees of chromatin condensation (55).
genic cells concomitant with the absence of a negative regula- This notion was reinforced by a recent study demonstrating
tory effect on viral E6 and E7 expression could provide an that HPV16 E7 can recruit to the IFN-␤ promoter region a
explanation for the observed depletion of immunological ef- histone deacetylase which blocks IRF-1 trans activation on
fector cells in dysplastic lesions (22, 29, 46, 60, 63, 64), which corresponding reporter constructs (42). Whether an altered
not only diminishes the immediate-early antiviral response but nucleosomal organization may account for inefficient IRF-1
also may increase the incidence of cervical cancer. transcription upon TNF-␣ treatment remains to be elucidated.
Although both E6 and E7 can counteract the function of In any case, IRF-1 dysregulation in tumorigenic HPV-positive
regulatory proteins involved in the ultimate IFN response (3, cells might be of potential biological interest, especially in light
40, 42, 48), it was amazing that tumorigenic cells still respond of the fact that IRF-1 can act as a tumor suppressor under

Downloaded from http://jvi.asm.org/ on June 8, 2015 by RUTGERS UNIVERSITY


to exogenous IFN treatment (Fig. 4). As reported recently, specific conditions (for a review, see reference 62). Since over-
HPV18 E6 affects IFN-␣ signaling by reducing Jak-STAT1/2 expression of IRF-1 induces apoptosis (62), it will be worth-
tyrosine phosphorylation in human fibrosarcoma cells (32). while in further studies to test whether the ectopic expression
Additionally, HPV16 E6 can bind to IRF-3, thereby impairing of a dominant-negative mutant of IRF-1 alters the growth
Sendai virus-induced activation of IFN-␤ and 2⬘,5⬘-oligoadeny- properties of 444 cells towards malignancy in nude mice.
late synthetase transcription (48). However, it must be stressed IRF-1 is functionally counteracted by IRF-2 by competition
that both processes were not completely perturbed by E6, for the same binding site within the IFN-␤ promoter (21).
strongly indicating that, at least in the latter case, other factors Moreover, IRF-2 has a considerably longer half-life than
(such as IRF-1 [see below]) can functionally substitute for IRF-1 (approximately 8 h versus 30 min) (62) and is present in
IRF-3. Nonetheless, because TNF-␣ can selectively suppress uninduced cells [Fig. 7A, lanes (⫺)], probably to prevent un-
HPV transcription in nontumorigenic cells (51), it was still controlled IFN-␤ synthesis. As further outlined in Fig. 7A,
conceivable that the reduction of oncogene expression to IRF-2 remained constitutively expressed throughout the cell
threshold levels may partially allow an IFN-␤ response. To cycle, and no obvious quantitative difference in the protein
clarify this point, we used modified nontumorigenic HeLa ⫻ amount could be seen when extracts from malignant and non-
fibroblast hybrids which were additionally transfected with an malignant cells were compared. An analogous situation was
HPV18 E6-E7 transcription cassette under the control of the found for the transcription of the IRF-3 gene, whose expres-
␤-actin promoter. In those transfectants, only endogenous sion was also not further augmented after TNF-␣ addition
transcription, and not the ␤-actin-driven E6-E7 transcription, (Fig. 7B). Conversely, like the case for IRF-1, only nonmalig-
became suppressed upon cytokine treatment. Nevertheless, nant cells retained their ability to selectively synthesize p48
TNF-␣-treated ␤-actin 444 cells were still protected against (ISGF3␥) (Fig. 8A). p48 (ISGF3␥) represents the DNA bind-
EMCV infection, arguing against a direct involvement of viral ing component of a trimeric complex (ISGF3) which induces,
oncogene expression in the outcome of the antiviral response jointly with STAT1 and STAT2, the transcription of antiviral
(Bachmann et al., unpublished results). genes such as those for the 2⬘,5⬘-oligoadenylate synthetase or
By compiling the results of experiments which monitor tran- double-stranded RNA-dependent protein kinase R (5). Al-
scription factors engaged in the differential regulation of though p48 (ISGF3␥) is structurally related to IRF-1 and binds
IFN-␤ gene expression (36), the following picture emerged. to the IFN-␤ promoter (24), the protein does not execute a
Using duplicated parts of PRDII in comparison to an NF-␬B redundant function in the cell but rather complements IRF-1
binding site derived from the immunoglobulin ␬ light chain in inducing both IFN-␣/␤ and IFN-␥ responses (21). The pref-
(44), TNF-␣ addition resulted in similar binding patterns when erential accumulation of p48 (ISGF3␥) in the nuclear fraction
nuclear extracts were analyzed in EMSAs (Fig. 5B). Therefore, of 444 cells (Fig. 8B) is in contrast to recent results showing
the absence of IFN-␤ inducibility in tumorigenic cells cannot that HPV16 E7 can block p48 (ISGF3␥) translocation into the
be attributed to inefficient cytokine signaling towards NF-␬B nucleus in spontaneously immortalized human keratinocytes
activation, since no obvious differences in affinity and binding (HaCaT cells) after IFN-␣ treatment (3). Since this study in-
kinetics could be discerned. An equivalent situation was found vestigated the role of E7 in IFN-␣ signaling in cells not asso-
with PRDIV-derived oligonucleotides harboring the recogni- ciated with a natural HPV infection, it is likely that the out-
tion sequences for c-Jun and ATF-2. Binding of the latter to come of the response may reflect the biological properties of
PRDIV was paralleled by a threonine-specific phosphorylation the respective model system, the dosage of the transduced
at position 71 (Fig. 5B), which occurred in the same temporal exogenous viral oncogene, the nature of the exogenous stim-
relationship as detected for the phosphorylation of the up- ulus, and/or the phenotype of the host cell.
stream MAPK p38MAPK (Fig. 1B). p38MAPK represents a ma- Another important aspect of our analysis is the correlation
jor effector MAPK of ATF-2, which becomes transiently acti- between nontumorigenicity and the antiviral activity induced
vated after addition of proinflammatory cytokines such as by TNF-␣. Considering our preceding experiments, we realized
TNF-␣ (65). that HeLa ⫻ fibroblast hybrids, which were converted to ma-
Regarding IRF-1, however, strong inducibility and DNA lignancy via ectopic c-fos expression (58), have almost com-
binding to PRDIII-I after TNF-␣ treatment was achieved only pletely lost their ability to block EMCV cytolysis after TNF-␣
in nonmalignant cells, while the corresponding protein was not addition (Bachmann et al., unpublished observations). We
detectable or was barely detectable in the tumorigenic coun- therefore went on to use an additional cell system which is
terparts (Fig. 6A, B, and C). The reason for this discrepancy in based on the fact that fusion of two malignant cells results in
290 BACHMANN ET AL. J. VIROL.

nonmalignant hybrids when different tumor suppressor genes J. M. Arbeit, L. M. Ellis, K. R. Cleary, and I. J. Fidler. 1999. Expression of
interferon-␤ is associated with growth arrest of murine and human epider-
are affected. Complementation to nontumorigenicity cannot mal cells. J. Investig. Dermatol. 112:802–809.
be accomplished when the same gene or pathway is defective 5. Bluyssen, H. A. R., J. E. Durbin, and D. E. Levy. 1996. ISGF3␥ p48, a
(7, 43). Accordingly, when HPV18-positive HeLa cells or specificity switch for interferon activated transcription factors. Cytokine
Growth Factor Rev. 7:11–17.
HPV16-positive CaSki cells, both of which are highly suscep- 6. Bornstein, J., Y. Ben-David, J. Atad, B. Pascal, M. Revel, and H. Abramovici.
tible to viral infection after pretreatment with TNF-␣ (Fig. 9A, 1993. Treatment of cervical intraepithelial neoplasia and invasive squamous
upper panels), were fused, the resulting hybrids were nontu- cell carcinoma by interferon. Obstet. Gynecol. Surv. 4504:252–260.
7. Chen, T. M., G. Pecoraro, and V. Defendi. 1993. Genetic analysis of in vitro
morigenic after inoculation into immunocompromised ani- progression of human papillomavirus-transfected human cervical cells. Can-
mals. In contrast, hybrid formation between HeLa cells and cer Res. 53:1167–1171.
7a.Chen, C.-J., T.-T. Lin, and J. E. Shively. 1996. Role of interferon regulatory
TNF-␣-unresponsive HPV18-positive SW756 cervical carci- factor-1 in the induction of biliary glycoprotein (cell CAM-1) by interferon-
noma cells (Fig. 9A, lower panels) failed to suppress tumor gamma. J. Biol. Chem. 271:2818–28188.
formation (59). When those hybrid clones were challenged 8. Chomczynski, P., and N. Sacchi. 1987. Single-step method of RNA isolation

Downloaded from http://jvi.asm.org/ on June 8, 2015 by RUTGERS UNIVERSITY


by acid guanidinium thiocyanate-phenol-chloroform extraction. Anal. Bio-
with viral infection after TNF-␣ treatment, antiviral activity chem. 162:156–159.
could be completely restored in nonmalignant CaSki ⫻ HeLa 9. Doly, J., A. Civas, S. Navarro, and G. Uze. 1998. Type I interferons: expres-
hybrids, whereas SW756 ⫻ HeLa hybrids (and SiHa ⫻ HeLa sion and signalization. Cell. Mol. Life Sci. 54:1109–1121.
10. Dürst, M., S. Seagon, S. Wanschura, H. zur Hausen, and J. Bullerdiek. 1995.
hybrids) remained sensitive (Fig. 9A; Table 1). Reconstitution Malignant progression of an HPV16-immortalized human keratinocyte cell
of a functional antiviral response was therefore not a peculiar- line (HPK Ia) in vitro. Cancer Genet. Cytogenet. 85:105–112.
ity of the initially utilized HeLa ⫻ fibroblast hybrid system but 11. Erlandsson, L., R. Blumenthal, M. L. Eloranta, H. Engel, G. Alm, S. Weiss,
and T. Leanderson. 1998. Interferon-beta is required for interferon-alpha
rather correlated with nonmalignancy and the capability to production in mouse fibroblasts. Curr. Biol. 8:223–226.
reexpress IRF-1 and p48 (ISGF3␥) (Fig. 10) and, in turn, 12. Feinberg, A. P., and B. Vogelstein. 1983. A technique for radiolabeling DNA
restriction endonuclease fragments to high specific activity. Anal. Biochem.
IFN-␤ (Fig. 9B). This supports the notion that the antiviral 132:6–13.
response of TNF-␣ is determined by the in vivo phenotype of 13. Finzer, P., U. Soto, H. Delius, A. Patzelt, J. F. Coy, A. Poustka, H. zur
the respective HPV-positive host cell line rather than by on- Hausen, and F. Rösl. 2000. Differential transcriptional regulation of the
monocyte-chemoattractant-protein-1 (MCP-1) gene in tumorigenic and non-
cogene expression per se. tumorigenic HPV 18 positive cells: the role of the chromatin structure and
Besides immunostimulatory and antiviral activities, the re- AP-1 composition. Oncogene 19:3235–3244.
striction of endogenous IFN-␤ production to nontumorigenic 14. Gangemi, J. D., L. Pirisi, M. Angell, and J. W. Kreider. 1994. HPV replica-
tion in experimental models: effects of interferon. Antiviral Res. 24:175–190.
cells also involves an additional interesting feature which may 15. Geisen, C., C. Denk, J. H. Küpper, and E. Schwarz. 2000. Growth inhibition
explain the long latency period between viral infection and the of cervical cancer cells by the human retinoic acid receptor beta gene. Int. J.
Cancer 85:289–295.
final progression to cervical cancer (67, 68). IFN-␤ synthesis 16. Gohji, K., I. J. Fidler, R. Tsan, R. Radinsky, A. C. Eschenbach, T. Tsuruo,
inversely correlates with angiogenesis as well as with cell pro- and M. Nakjima. 1994. Human recombinant interferon-beta and -gamma
liferation and, under some circumstances, with terminal differ- decrease gelatinase production and invasion by human KG-2 renal carci-
noma cells. Int. J. Cancer 58:380–384.
entiation (4). Notably, IFN-␤ can down-regulate angiogenic 17. Goodbourn, S., L. Didcock, and R. E. Randell. 2000. Interferons: cell sig-
factors such as basic fibroblast growth factor (56), interleukin-8 nalling, immune modulation, antiviral responses and virus countermeasures.
(57), and matrix metalloproteases (16), all of which are neces- J. Gen. Virol. 81:2341–2364.
18. Griffiths, D. J., P. J. Venables, R. A. Weiss, and M. T. Boyd. 1997. A novel
sary to promote tumor growth and metastasis. Furthermore, exogenous retrovirus sequence identified in humans. J. Virol. 71:2866–2872.
upon analysis of human keratinocytes in medium either favor- 19. Guidotti, L. G., and F. V. Chisari. 2000. Cytokine-mediated control of viral
infections. Virology 273:221–227.
ing or preventing terminal differentiation, IFN-␤ expression 20. Hagari, Y., L. R. Budgeon, M. D. Pickel, and J. W. Kreider. 1995. Association
was detectable only in cells without proliferating cell nuclear of tumor necrosis factor-␣ gene expression and apoptotic cell death with
antigen (PCNA). Similar results were obtained from the im- regression of Shope papillomas. J. Investig. Dermatol. 104:526–529.
21. Harada, H., T. Fujita, M. Miyamoto, Y. Kimura, M. Maruyama, A. Furia, T.
munohistological evaluation of normal tissue sections, where Miyata, and T. Taniguchi. 1989. Structurally similar but functionally distinct
IFN-␤ production was restricted to nondividing suprabasal cell factors, IRF-1 and IRF-2, bind to the same regulatory elements of IFN and
layers (4). It will therefore be the aim of further studies to IFN-inducible genes. Cell 58:729–739.
22. Hughes, R. G., M. Norval, and S. E. M. Howie. 1988. Expression of major
analyze these additional properties of IFN-␤ in the context of histocompatibility class II antigens by Langerhans’ cells in cervical intraepi-
HPV-induced carcinogenesis in greater detail. thelial neoplasia. J. Clin. Pathol. 41:253–259.
23. Jacobsen, H., J. Mestan, S. Mittnacht, and C. W. Dieffenbach. 1989. Beta
interferon subtype 1 induction by tumor necrosis factor. Mol. Cell. Biol.
ACKNOWLEDGMENTS 9:3037–3042.
24. Kawakami, T., M. Matsumoto, M. Sato, H. Harada, T. Taniguchi, and M.
We thank Eric Stanbridge (University of California, Irvine), Claudia Kitagawa. 1995. Possible involvement of the transcription factor ISGF3␥ in
Denk (DKFZ, Heidelberg, Germany), and Matthias Dürst (University virus-induced expression of the IFN-␤ gene. FEBS Lett. 358:225–229.
of Jena, Jena, Germany) for providing cell lines. The technical help of 25. Khan, M. A., W. H. Tolleson, J. D. Gangemi, and L. Pirisi. 1993. Inhibition
Anita Weyland is appreciated. of growth, transformation, and expression of human papillomavirus type 16
E7 in human keratinocytes by alpha interferons. J. Virol. 67:3396–3403.
26. Khandjian, E. W., and C. Meric. 1986. A procedure for Northern blot
REFERENCES
analysis of native RNA. Anal. Biochem. 159:227–232.
1. Algarte, M., H. Kwon, P. Genin, and J. Hiscott. 1999. Identification by in vivo 27. Kimura, T., Y. Kadokawa, H. Harada, M. Matsumoto, M. Sato, Y. Kashi-
genomic footprinting of a transcriptional switch containing NF-␬B and Sp1 wazaki, M. Tarutani, R. S. P. Tan, T. Takasugi, T. Matsuyama, T. W. Mak,
that regulates the I␬B␣ promoter. Mol. Cell. Biol. 19:6140–6153. S. Noguchi, and T. Taniguchi. 1996. Essential and non-redundant role of p48
2. Au, W.-C., P. A. Moore, W. Lowther, Y.-T. Juang, and P. M. Pitha. 1995. (ISGF3␥) and IRF-1 in both type I and type II interferon responses, as
Identification of a member of the interferon regulatory factor family that revealed by gene targeting studies. Genes Cells 1:115–124.
binds to the interferon-stimulated response element and activates expression 28. Kleine, K., G. König, J. Kreuzer, D. Komitowski, H. zur Hausen, and F.
of interferon-induced genes. Proc. Natl. Acad. Sci. USA 92:11657–11661. Rösl. 1995. The effect of the JE (MCP-1) gene, which encodes the monocyte
3. Barnard, P., and N. A. J. McMillan. 1999. The human papillomavirus E7 chemoattractant protein-1, on the growth of HeLa cells and derived somatic
oncoprotein abrogates signaling mediated by interferon-␣. Virology 259: cell hybrids in nude mice. Mol. Carcinogenesis 14:179–189.
305–313. 29. Kleine-Lowinski, K., R. Gillitzer, H. Kühne-Heid, and F. Rösl. 1999. Mono-
4. Bielenberg, D. R., M. F. McCarty, C. D. Bucana, S. H. Yuspa, D. Morgan, cyte-chemoattractant protein-1 (MCP-1) gene expression in cervical intra-
VOL. 76, 2002 ABSENCE OF IFN-␤ EXPRESSION IN CERVICAL CARCINOMA CELLS 291

epithelial hyperplasia and cervical carcinomas. Int. J. Cancer 82:6–11. 51. Rösl, F., M. Lengert, J. Albrecht, K. Kleine, R. Zawatzky, B. Schraven, and
30. Koromilas, A. E., S. Li, and G. Matlashewski. 2001. Control of interferon H. zur Hausen. 1994. Differential regulation of the JE gene encoding the
signalling in human papillomavirus infection. Cytokine Growth Factor Rev. monocyte chemoattractant protein (MCP-1) in cervical carcinoma cells and
12:157–170. derived hybrids. J. Virol. 68:2142–2150.
31. Lenardo, M. J., C.-M. Fan, T. Maniatis, and D. Baltimore. 1989. The in- 52. Rösl, F., K. Kleine-Lowinski, and H. zur Hausen. 1999. The possible role of
volvement of NF-␬B in ␤-interferon gene regulation reveals its role as a chemokines in HPV-linked carcinogenesis, p. 207–225. In B. J. Rollins (ed.),
widely inducible mediator of signal transduction. Cell 57:287–294. Chemokines and cancer. Humana Press Inc., Totowa, N.J.
32. Li, S., S. Labrecque, M. C. Gauzzi, A. R. Cuddihy, A. H. T. Wong, S. 52a.Saxon, P. J., E. S. Srivatsan, and E. J. Stanbridge. 1986. Introduction of
Pellegrini, G. J. Matlashewski, and A. E. Koromilas. 1999. The human human chromosome 11 via microcell transfer controls tumorigenic expres-
papillomavirus (HPV)-18 E6 oncoprotein physically associates with Tyk2 sion of HeLa cells. EMBO J. 5:3461–3466.
and impairs Jak-STAT activations by interferon-␣. Oncogene 18:5727–5737. 53. Schneider, A., and L. A. Koutsky. 1992. Natural history and epidemiological
33. Lopez, S., and S. Navarro. 1998. Transcriptional repression of type I IFN features of genital HPV infection, p. 25–52. In N. Munoz et al. (ed.), The
genes. Biochimie 80:689–701. epidemiology of human papillomavirus and cervical cancer. International
34. Maehama, T., A. Patzelt, M. Lengert, K.-J. Hutter, K. Kanazawa, H. zur Agency for Research on Cancer, Lyon, France.
Hausen, and F. Rösl. 1998. Selective down-regulation of human papilloma- 54. Schreiber, E., P. Matthias, M. M. Müller, and W. Schaffner. 1989. Rapid
virus transcription by 2-deoxyglucose. Int. J. Cancer 76:639–646. detection of octamer binding proteins with ‘mini-extracts’ prepared from a

Downloaded from http://jvi.asm.org/ on June 8, 2015 by RUTGERS UNIVERSITY


35. Majewski, S., J. Malejczyk, and S. Jablonska. 1996. The role of cytokines small number of cells. Nucleic Acids Res. 15:6419–6436.
and other factors in HPV infection and HPV-associated tumors. Papilloma- 55. Shestakova, E., M.-T. Bandu, J. Doly, and E. Bonnefoy. 2001. Inhibition of
virus Rep. 7:143–155. histone deacetylase induces constitutive derepression of the beta interferon
36. Maniatis, T., J. V. Falvo, T. H. Kim, T. K. Kim, C. H. Lin, B. S. Parekh, and promoter and confers antiviral activity. J. Virol. 75:3444–3452.
M. G. Wathelet. 1998. Structure and function of the interferon-beta enhan- 56. Singh, R. K., M. Gutman, C. D. Bucana, R. Sanches, N. Llansa, and I. J.
ceosome. Cold Spring Harbor Symp. Quant. Biol. 63:609–620. Fidler. 1995. Interferons alpha and beta downregulate the expression of
37. McNeill, T. A. 1981. Interferon assay. J. Immunol. Methods 46:121–127. basic fibroblast growth factor in human carcinomas. Proc. Natl. Acad. Sci.
38. Mestan, J., W. Digel., S. Mittnacht, H. Hillen, D. Blohm, A. Moeller, H. USA 92:4562–4566.
Jacobsen, and H. Kirchner. 1986. Antiviral effects of recombinant tumour 57. Singh, R. K., M. Gutman, N. Llansa, and I. J. Fidler. 1996. Interferon-␤
necrosis factor in vitro. Nature (London) 323:816–819. prevents the up-regulation of interleukin-8 expression in human melanoma
39. Miller, M. D., and M. S. Krangel. 1992. Biology and biochemistry of the cells. J. Interferon Cytokine Res. 16:577–584.
chemokines: a family of chemotactic and inflammatory cytokines. Crit. Rev. 58. Soto, U., B. C. Das, M. Lengert, P. Finzer, H. zur Hausen, and F. Rösl. 1999.
Immunol. 12:17–46. Conversion of HPV 18 positive non-tumorigenic HeLa-fibroblast to invasive
40. Nees, M., J. M. Geoghegan, T. Hyman, S. Frank, L. Miller, and C. D. growth involves loss of TNF-␣ mediated repression of viral transcription and
Woodworth. 2001. Papillomavirus type 16 oncogenes downregulate expres- modification of the AP-1 transcription complex. Oncogene 18:3187–3198.
sion of interferon-responsive genes and upregulate proliferation-associated 59. Soto, U., C. Denk, P. Finzer, K.-J. Hutter, H. zur Hausen, and F. Rösl. 2000.
and NF-␬B-responsive genes in cervical keratinocytes. J. Virol. 75:4283– Genetic complementation to non-tumorigenicity in cervical carcinoma cells
4296. correlates with alterations in AP-1 composition. Int. J. Cancer 86:811–817.
41. Oguchi, M., J. Komura, H. Tagami, and S. Ofuji. 1981. Ultrastructural 60. Spinillo, A., P. Tenti, R. Zappatore, F. De Seta, E. Silini, and S. Guaschino.
studies of spontaneously regressing plane warts. Macrophages attack verru- 1993. Langerhans’ cell counts and cervical intraepithelial neoplasia in
ca-epidermal cells. Arch. Dermatol. Res. 270:403–411. women with human immunodeficiency virus infection. Gynecol. Oncol. 48:
42. Park, J.-S., E.-J. Kim, H.-J. Kwon, E.-S. Hwang, S.-E. Namkoong, and S.-J. 210–213.
Um. 2000. Inactivation of interferon-regulatory factor-1 tumor suppressor 61. Stanbridge, E. 1984. Genetic analysis of tumorigenicity in human cell hy-
protein by HPV E7 oncogene. J. Biol. Chem. 275:6764–6769. brids. Cancer Surv. 3:335–350.
43. Pereira-Smith, O. M., and J. R. Smith. 1988. Genetic analysis of indefinite 62. Taniguchi, T., N. Tanaka, K. Ogasawara, S. Taki, M. Sato, and A. Takaoka.
division in human cells: identification of four complementation groups. Proc. 1999. Transcription factor IRF-1 and its family members in the regulation of
Natl. Acad. Sci. USA 85:6042–6045. the host response. Cold Spring Harbor Symp. Quant. Biol. 64:465–472.
44. Pierce, J. W., M. Lenardo, and D. Baltimore. 1988. Oligonucleotide that 63. Tay, S. K., D. Jenkins, P. Maddox, N. Hogg, and A. Singer. 1987. Tissue
binds nuclear factor NF-kappa B acts as a lymphoid-specific and inducible macrophage response in human papillomavirus infection and cervical intra-
enhancer element. Proc. Natl. Acad. Sci. USA 85:1482–1486. epithelial neoplasia. Br. J. Obstet. Gynaecol. 94:1094–1097.
45. Rappolee, D. A., and Z. Werb. 1992. Macrophage-derived growth factors. 64. Viac, J., I. Guerin-Reverchon, Y. Chardonnet, and A. Bremond. 1990. Lang-
Curr. Top. Microbiol. Immunol. 181:87–140. erhans cells and epithelial cell modifications in cervical intraepithelial neo-
46. Riethdorf, L., S. Riethdorf, K. Gützlaff, F. Prall, and T. Löning. 1996. plasia: correlation with human papillomavirus infection. Immunobiology
Differential expression of the monocyte chemoattractant protein-1 in human 180:328–338.
papillomavirus-16 infected squamous intraepithelial lesions and squamous 65. Wysk, M., D. D. Yang, H.-T. Lu, R. A. Flavell, and R. J. Davis. 1999.
cell carcinomas of the cervix uteri. Am. J. Pathol. 149:1469–1476. Requirement of mitogen-activated protein kinase kinase 3 (MKK3) for tu-
47. Rollins, B. J. 1991. JE/MCP-1: an early-response gene encodes a monocyte- mor necrosis factor-induced cytokine expression. Proc. Natl. Acad. Sci. USA
specific cytokine. Cancer Cells 3:517–524. 96:3763–3768.
48. Ronco, L. V., A. Y. Karpova, M. Vidal, and P. M. Howley. 1998. Human 66. Yousefi, S., M. R. Escobar, and C. W. Gouldin. 1985. A practical cytopathic
papillomavirus 16 E6 oncoprotein binds to interferon regulatory factor-3 and effect/dye-uptake interferon assay for routine use in the clinical laboratory.
inhibits its transcriptional activity. Genes Dev. 12:2061–2072. Am. J. Clin. Pathol. 83:735–740.
49. Rösl, F., E.-M. Westphal, and H. zur Hausen. 1989. Chromatin structure and 67. zur Hausen, H. 1994. Disrupted dichotomous intracellular control of human
transcriptional regulation of human papillomavirus type 18 DNA in HeLa papillomavirus infection in cancer of the cervix. Lancet 343:955–957.
cells. Mol. Carcinogenesis 2:72–80. 68. zur Hausen, H., and F. Rösl. 1994. Pathogenesis of cancer of the cervix. Cold
50. Rösl, F., T. Achtstätter, T. Bauknecht, G. Futterman, K. J. Hutter, and H. Spring Harbor Symp. Quant. Biol. 59:623–628.
zur Hausen. 1991. Extinction of the HPV18 upstream regulatory region in 69. zur Hausen, H. 2000. Papillomaviruses causing cancer: evasion from host-
cervical carcinoma cells after fusion with non-tumorigenic human keratino- cell control in early events in carcinogenesis. J. Natl. Cancer Inst. 92:690–
cytes under non-selective conditions. EMBO J. 10:1337–1345. 698.

You might also like