http://cancerres.aacrjournals.org/content/46/7/3574.full.pdf

publicité
[CANCER RESEARCH 46, 3574-3579, July 1986]
Selective Killing of Simian Virus 40-transformed Human Fibroblasts by
Parvovirus H-l1
Yong Quan Chen, Françoisede Foresta, Jacqueline Hertoghs, Bernard L. Avalosse, Jan J. Cornelis, and
Jean Rommelaere2
Laboratory of Biophysics and Radiobiology, UniversitéLibre de Bruxelles, B-1640 Rhode St. Genèse,Belgium [Y. Q. C., F. d. F., J. H., B. L. A., J. J. C., J. RJ, and
Laboratory of Molecular Oncology, 1NSERM UI86, Institut Pasteur de Lille, F-59019 Lille, France [B. L. A., J. J. C.. J. RJ
ABSTRACT
A normal strain of human foreskin fibroblasts, two SV40-transformed
derivatives with finite and infinite life spans, and an established line of
SV40-transformed
newborn human kidney cells are compared for their
susceptibility to infection with parvovirus H-l. H-l inocula, which do not
detectably alter the growth of normal cells, cause a progressive degen
eration of all three SV40-transformed cultures. The resistance of normal
cells is not a membrane phenomenon since they adsorb and take up H-l
as efficiently as the transformants. Moreover, the fraction of infected
cells supporting the synthesis and nuclear migration of H-l proteins is
similar in normal and SV40-transformed
cultures. On the other hand,
the enhanced H-l sensitivity of transformed cells correlates with a 5- to
30-fold increase in their accumulation of newly synthesized parvoviral
DNA, as compared with normal cultures. This stimulation of H-l DNA
replication is most pronounced for the amplification of duplex replicative
forms, although the conversion of parental single-stranded DNA to
replicative forms is also enhanced to a smaller extent. In addition, SV40transformed cells support productive H-l infection and release a burst of
infectious virus, whereas no H-l production can be detected in the normal
cell strain. The latter difference was confirmed for another series of 7
normal and 16 SV40-transformed
strains of human skin fibroblasts.
Altogether, these results indicate that intracellular limitations on H-l
DNA replication are associated with the abortive nature of the parvoviral
life cycle in normal human fibroblasts and are overcome after SV40
transformation, resulting in the selective killing of the transformants.
This observation raises the possibility that oncolysis might contribute to
the oncosuppressive activity displayed by parvoviruses in vivo.
INTRODUCTION
Parvoviruses are nuclear replicating, single-stranded DNA
viruses of small size and low genetic complexity, which have
been isolated, in particular, from a number of avian and mam
malian species including humans (1). Parvoviruses have a lytic
replicative life cycle and are devoid of detectable oncogenic
activity, but they have the intriguing ability to suppress cancer
formation in laboratory animals and there is speculation about
their similar effect in humans (for reviews, see Refs. 2 and 3).
Vertebrate parvoviruses can be divided into two subgroups,
nondefective (so-called autonomous) and defective (so-called
adeno-associated) members, respectively. The antineoplastic
spectrum of autonomous parvoviruses is broad and includes
spontaneous (4) as well as \ ¡rally(3) and chemically (5) induced
cancers, whereas adeno-associated viruses specifically inhibit
helper virus oncogenicity (2). The cellular and molecular bases
of oncosuppression by parvoviruses are poorly understood.
Parvoviruses might interfere with complex physiological proc
esses conditioning tumor development (5). On the other hand,
adeno-associated (6, 7) and autonomous (8) parvoviruses have
both been shown to inhibit in vitro transformation of cells in
culture, although they might act by different mechanisms. Thus,
direct interactions between parvoviruses and transformed cells
or their precursors, as occurring in vitro, might also contribute
to oncosuppression.
We reported previously that the autonomous parvovirus
MVM3 prevents the tumor virus SV40 from transforming in
vitro mouse cells which had been selected for their resistance to
MVM infection (8). This inhibition could be ascribed to the
specific lysis of SV40 transformants by MVM. Together with
the known requirements of parvoviruses for the proliferation
(3) and appropriate differentiation (1) of their host cells, this
observation led us to hypothesize that malignant transforma
tion may render normally resistant cells permissive to the
replication of these viruses. Hence, transformed cells would be
a preferential target for the lytic action of autonomous parvo
viruses. The possible involvement of such an oncolysis in cancer
suppression by nondefective parvoviruses remains to be deter
mined.
The work presented in this paper was aimed at analyzing the
mechanism by which SV40 transformation sensitizes cells to
the killing effect of autonomous parvoviruses and at determin
ing the generality of this phenomenon. Since it was known that
the nondefective parvovirus H-l can be propagated in an estab
lished line of SV40-transformed newborn human kidney cells
(9), it was decided to compare a normal diploid strain of human
foreskin fibroblasts and two SV40-transformed derivatives with
finite and infinite life spans for their susceptibility to H-l
infection. Consistent with the oncolysis hypothesis, SV40 trans
formation was found to correlate with an enhanced cell permis
siveness to H-l.
MATERIALS AND METHODS
Cells. Human cell strains were grown as monolayers in Eagle's
minimal essential medium supplemented with 10% fetal calf serum.
NB-E is an established line of S\ 40 transformed newborn human
kidney cells (10). VII 10 is a normal finite-life strain of foreskin human
fibroblasts from which nonestablished (VH-10 1SV) and established
(VH-10 SV), SV40 T-antigen-positive clones were independently de
rived by transformation with the early region of SV40 6-17 mutant
deleted within the replication origin. The VH-10 series was prepared
by B. Klein and kindly provided by A. van der Eb (Leiden University,
Leiden, The Netherlands). Cells were counted with a hemocytometer
after trypsinization and their viability was determined by their ability
to exclude 0.07% (w/v) trypan blue stain.
Virus. H-l parvovirus (wild type) was propagated in NB-E cells and
purified according to the method of Tattersall et al. (11). 3H- and 32Plabeled virus was produced by incubating infected cells with [3H]thymidine (5 Ci/mmol, 25 ^Ci/ml) and 32P¡
(carrier free, 0.1 mCi/ml), as
Received 11/12/85; revised 3/5/86; accepted 3/26/86.
The costs of publication of this article were defrayed in part by the payment
of page charges. This article must therefore be hereby marked advertisement in
accordance with 18 U.S.C. Section 1734 solely to indicate this fact.
1Supported by an Action de Recherche Concertéefrom the Ministère de la
Politique Scientifique and by grants from the Caisse Générale
d'Epargne et de
described by Rhode (12) and Rommelaere and Ward (13), respectively.
Conditions for cell infection, virus harvesting, and titration by plaque
assay have been described previously (14).
H-l Uptake. Cells were incubated for 60 min at 37°Cor 4°Cin the
Retraite, Loterie Nationale, and Fonds de la Recherche Scientifique Médicale.
2 To whom requests for reprints should be addressed.
3The abbreviations used are: MVM, minute virus of mice; SS, single-stranded;
RF, replicative form.
3574
Downloaded from cancerres.aacrjournals.org on July 8, 2017. © 1986 American Association for Cancer Research.
Downloaded from cancerres.aacrjournals.org on July 8, 2017. © 1986 American Association for Cancer Research.
Downloaded from cancerres.aacrjournals.org on July 8, 2017. © 1986 American Association for Cancer Research.
CELL TRANSFORMATION
AND SUSCEPTIBILITY
Table 3 H-l protein synthesis in normal and SV40-transformed human
fibroblasto
H-1-infected cells were tested for viral protein synthesis by an immunoradiometric assay or immunoenzymatic staining. Background radioactivity due to the
immunoradiometric method (-350 cpm) and to input virus proteins (-150 cpm)
was measured 2 h postinfection and was subtracted. Nuclear staining was quantitated in 200 cells and its positiveness and intensity were determined after similar
background (A = 6) subtraction. The relative synthesis of viral proteins is referred
to the corresponding VH-10 value considered as 1.0.
Immunoradiometric
Cells
cpm/10* cells
assay
Relative
synthesis
TO PARVOVIRUSES
Table 4 H-l virus replication in human fibroblasts
Cultures of human fibroblasts (5 x 10* cells) were infected with H-l (multi
plicity of infection, 10~2), incubated for 4 days, and tested for the production of
infectious virus. Increases in tiler of at least 3-fold over the input virus were
scored as positive. The virus titer in nonproducing cells actually decreased with
time postinfection. Input virus was titered 2 h postinfection.
of
SV40
H-l
strains
transferproducRef.
tion38
tested
mation
CellsVH-10VH-10SVGM2149
Immunoenzymatic staining
% of Absorbance/
positive
positive
nuclei
nucleus
139
+139
2
+
GM2149SVGM1061 +140
3
+
GM1061
SVGM1085
+140
2
+
GM1085SVAT5BI
+41
8
+
AT5BI
VA2BI
a41
46BRXP30RO
4241a10+-:
Relative
synthesis
VH-10VH-10
SVVH-10
1SVNB-E12422420325348471.01.952.63.926.5292628111621211.01.61.92.0
GM0637 B
NB-ENo.
:
" National Institute of General Medical Sciences Human Genetic Mutant Cell
Repository.
DISCUSSION
Abortive Interaction between Parvovirus H-l and Normal Hu
man Fibroblasts
012345
Time postinfection
(days)
Fig. 4. H-l virus production in normal and SV40-transformed human fibro
blasts. Cultures (5 x 10s cells) were infected with H-l virus (multiplicity of
infection, IO"2) and analyzed at intervals for the production of infectious virus.
Average values for 2 experiments (SD less than 15%). PFU, plaque-forming units.
Same symbols as for Fig. 1.
2) synthesis correlated, although the former was much lower
than the latter.
Production of Infectious H-l Particles. Cultures of normal
VH-10 cells were unable to amplify the H-l inoculum; the titer
of infectious virus harvested from these cultures rather declined
with time (Fig. 4). Thus, most normal fibroblasts did not appear
to sustain a productive H-l infection. In contrast, SV40-transformed strains were all productively infected and released a
burst of infectious H-l (Fig. 4). NB-E and transformed VH-10
cells displayed high and intermediate degrees of permissiveness
compared to normal VH-10 cultures, respectively, which cor
related with their relative abilities to support H-l DNA repli
cation (Table 2) and susceptibilities to the lytic action of H-l
(Fig. 1). The induction of permissiveness to H-l as a result of
SV40 transformation is not unique to VH-10 cells. As shown
by Table 4, a similar effect was observed for a number of other
human skin fibroblasts. None of the normal cultures tested were
able to replicate H-l whereas SV40-transformed strains were
the hosts of a productive H-l infection.
A series of nonpermanent cultures of normal human skin
fibroblasts were found to resist productive infection with parvovirus H-l. Resistance could be ascribed to the failure of an
intracellular stage of the replicative life cycle of H-l, resulting
in a lack of amplification of the virus inoculum. Similar results
have been reported for human amnion, embryo kidney (25),
and lung (9) cells. A quantitative comparison of H-l replication
was undertaken between normal skin fibroblasts and corre
sponding permissive cultures, namely SV40 transformants de
rived therefrom (see below). A striking difference was observed
at the level of the replication of RF DNA, which was much
enhanced in transformed cultures and correlated with their
ability to produce infectious virus. The defect of the H-l life
cycle in diploid human fibroblasts might concern RF DNA
replication itself. Alternatively, another step of the viral cycle
conditioning the extent of RF DNA replication might be im
paired, although our data suggest that virus uptake, nuclear
translocation, and parental DNA conversion are unlikely to be
primarily involved.
The inability of normal human fibroblasts to support RF
DNA replication efficiently might result from their failure to
express nonessential host functions usurped by H-l for its own
growth. In addition, H-l DNA replication appears to involve
virus-encoded functions (26). Nonpermissive cells might also
fail to synthesize or modify these viral products, although no
major impairment of H-l capsid gene expression was detected
in diploid human fibroblasts. Our data indicate that the limi
tation placed by normal cells on H-l replication can be reversed
as a result of their in vitro transformation by SV40. A parallel
can be drawn between this observation and other reports show
ing that cellular changes can render normally resistant cultures
susceptible to autonomous parvoviruses. Permissiveness to parvoviruses has been shown to be dependent on host cell prolif
eration (27,28) and differentiation (29-31). Various physiolog-
3577
Downloaded from cancerres.aacrjournals.org on July 8, 2017. © 1986 American Association for Cancer Research.
CELL TRANSFORMATION
AND SUSCEPTIBILITY
ical states can lead to the arrest of the parvoviral life cycle at
different steps, both on the surface and on the inside of the cell
(32). Therefore, there appear to be multiple conditional host
factors which determine permissiveness to parvoviruses. It fol
lows that the specific abortive interaction reported here for
diploid human fibroblasts cannot be necessarily generalized to
other cell types.
TO PARVOVIRUSES
ACKNOWLEDGMENTS
The authors are greatly indebted to Drs. A. van der Eb and B. Klein
for their generous gift of VH-10 and derived cells, to C. Kumps for
technical assistance, and to Dr. P. Tattersall for critical reading of the
manuscript. Some human mutant cells were kindly provided by Drs. C.
Arieti, A. Lehmann, and D. Bootsma.
Induction of 11-1 Sensitivity by SV40 Transformation
REFERENCES
The acquisition of permissiveness to H-l replication by SV40
transformants correlated with their sensitization to parvoviral
lytic action. H-l inocula which had little effect on the growth
of normal cultures caused a progressive degeneration of trans
formed derivatives. Thus, the preferential killing of transformed
cells is likely to result from their propensity to replicate H-l
rather than their lower resistance to its cytopathic effect. A
similar sensitization of SV40 transformants to the autonomous
parvovirus M VM was reported previously for mouse cells which
had been selected in vitro for their resistance to this virus (8).
Therefore, SV40 transformation overcomes both natural and
selected cellular protections against parvoviral attack.
The enhanced susceptibility of SV40 transformants to H-l
infection does not require their establishment into a permanent
culture. Thus, SV40 transformation appears to act directly and
not merely by facilitating cell immortalization. Since the expres
sion of the SV40 genome in transformants is restricted to the
early region-encoding T-antigens (33), the latter protein(s) is
likely to mediate sensitization to H-l. Large T-antigen is known
to activate cellular gene expression (33, 34). As stated above,
cell permissiveness to parvoviruses involves host functions,
some of which are not permanently expressed and might con
ceivably be induced by SV40. On the other hand, a direct
interaction between the H-l genome and SV40 T-antigen(s)
might also potentiate H-l replication. Another tumor virus,
adenovirus, was found to provide helper functions to H-l in
coinfected human cells (35) and encodes a tumor antigen which
could possibly interact directly with the DNA of the defective
parvovirus adeno-associated virus (7). Whatever mechanism it
involves, sensitization to H-l is apparently not specific for
tumor virus-transformed cells. Indeed we have recently ex
tended the present observation to human diploid fibroblasts
transformed by 7-irradiation (36). Nonvirally transformed cells
therefore appear to supply helper functions similar to those
encoded or induced by SV40.
Altogether, our data point to a positive interrelation between
two complex cellular phenotypes each composed of multiple
components, i.e., permissiveness to autonomous parvoviruses
and /// vitro malignant transformation. This result raises two
intriguing possibilities, (a) The lytic life cycle of parvoviruses
may provide specific markers for cell transformation. We report
here that SV40 transformation bypasses a limitation to H-l
DNA replication. In contrast, a postreplicative step of the
growth of parvovirus MVM was found to be stimulated as a
result of the transformation of rat cells with the retrovirus avian
erythroblastosis virus (37). An interesting issue is whether the
overcoming of distinct barriers to parvovirus replication re
quires the expression of different transformation traits, (b) The
suppresion of cancer development by autonomous parvoviruses
may result, at least in part, from oncolysis. The preferential
cytolytic replication of autonomous parvoviruses in trans
formed cells accounts for the ability of these viruses to specifi
cally inhibit in vitro malignant transformation (8). Whether
cancer cells provide a similar target for destruction by parvo
viruses in vivo remains to be determined.
1. Tattersall, P., and Ward, D. The parvoviruses: an introduction. In: D. C.
Ward and P. Tattersall (eds.), Replication of Mammalian Parvoviruses, pp.
3-12. Cold Spring Harbor, NY: Cold Spring Harbor Laboratory, Publisher,
1978.
2. Cukor, G., Blacklow, N. R., Hoggan, D., and Berns, K. I. Biology of adenoassociated virus. In: K. I. Berns (ed.). The Parvoviruses, pp. 33-36. New
York: Plenum Publishing Corp., 1984.
3. Siegl, G. Biology of pathogenicity of autonomous parvoviruses. In: K. I.
Berns (ed.). The Parvoviruses, pp. 297-362. New York: Plenum Publishing
Corp., 1984.
4. Toolan, H. W. Lack of oncogenic effect of the H-viruses. Nature (Lond.),
214: 1036, 1967.
5. Toolan, H. W., Rhode, S. L., and Gierthy, J. F. Inhibition of 7,12-dimethylbenz(a)anthracene-induced tumors in Syrian hamsters by prior infection
with H-l parvovirus. Cancer Res., 42: 2552-2555, 1982.
6. Casto, B. C., and Goodheart, C. R. Inhibition of adenovirus transformation
in vitro by AAV-1. Proc. Soc. Exp. Biol. Med., 140: 72-78, 1972.
7. Ostrove, J. M., Duckworth, D. H., and Berns, K. I. Inhibition of adenovirustransformed cell oncogenicity by adeno-associated virus. Virology, 113: 521533, 1981.
8. Mousset, S., and Rommelaere, J. Minute virus of mice inhibits cell transfor
mation by simian virus 40. Nature (Lond.), 300: 537-539, 1982.
9. Singer, I. I., and Rhode, S. L. Electron microscopy and cytochemistry of H1 parvovirus intracellular morphogenesis. In: D. C. Ward and P. Tattersall
(eds.). Replication of Mammalian Parvoviruses, pp. 479-504. Cold Spring
Harbor, NY: Cold Spring Harbor Laboratory, Publisher, 1978.
10. Shein, H. M., and Enders, J. F. Multiplication and cytopathogenicity of
simian vacuolating virus 40 in cultures of human tissues. Proc. Soc. Exp.
Biol. Med., 709:495-500, 1962.
11. Tattersall, P., Cawte, P. J., Shatkin, A. J., and Ward, D. C. Three structural
polypeptides coded for by minute virus of mice, a parvovirus. J. Virol., 20:
273-289, 1976.
12. Rhode, S. L. H-l DNA synthesis. In: D. C. Ward and P. Tattersall (eds.),
Replication of Mammalian Parvoviruses, pp. 279-296. Cold Spring Harbor,
NY: Cold Spring Harbor Laboratory, Publisher, 1978.
13. Rommelaere, J., and Ward, D. C. Effect of UV-irradiation on DNA repli
cation of the parvovirus minute virus of mice in mouse fibroblasts. Nucleic
Acids Res., 10: 2577-2596, 1982.
14. Cornells, J. J., Su, Z. Z., Ward, D. C., and Rommelaere, J. Indirect induction
of mutagenesis of intact parvovirus H-l in mammalian cells treated with
UV-light or with UV-irradiated H-I or SV40 viruses. Proc. Nati. Acad. Sci.
USA, 78:4480-4484, 1981.
15. Linser, P., Bruning, H., and Armentrout, R. W. Uptake of minute virus of
mice into cultured rodent cells. J. Virol., 31: 537-545, 1979.
16. Lavi, S. Carcinogen-mediated amplification of viral DNA sequences in simian
virus 40-transformed Chinese hamster embryo cells. Proc. Nati. Acad. Sci.
USA, 78: 6144-6148, 1981.
17. Southern, E. M. Detection of specific sequences among DNA fragments
separated by gel electrophoresis. J. Mol. Biol., 98: 503-517, 1975.
18. Winocour, E., and Keshet, I. Indiscriminate recombination in simian virus
40-infected monkey cells. Proc. Nati. Acad. Sci. USA, 77:4861-4865, 1980.
19. Merchlinsky, M. J., Tattersall, P. J., Leary, J. J., Cotmore, S. F., Gardiner,
E. M., and Ward, D. C. Construction of an infectious molecular clone of the
autonomous parvovirus minute virus of mice. J. Virol., 47: 227-232, 1983.
20. Paradiso, P. R., Williams, K. R., and Costantino, R. L. Mapping of the
amino terminus of the H-l parvovirus major capsid proteins. J. Virol., 52:
77-81, 1984.
21. Rigby, P. W. J., Dieckmann, M., Rhodes, C., and Berg, P. Labeling deoxyribonucleic acid to high spa-ilk activity in vitro by nick translation with DNA
polymerase I. J. Mol. Biol., 113: 237-251, 1977.
22. Luo, Z. Y., Cornells, J. J., Vos, J. M., and Rommelaere, J. UV-enhanced
reactivation of capsid protein synthesis and infectious centre formation in
mouse cells infected with UV-irradiated minute virus of mice. Int. J. Radiât.
Biol., 40:119-126, 1982.
23. Ward, D. C., and Tattersall, P. J. Minute virus of mice. In: H. L. Foster, J.
D. Small, and J. G. Fox (eds.), The Mouse in BiomédicalResearch, Vol. 2,
pp. 313-334. New York: Academic Press, 1982.
24. Astell, C. R., Chow, M. B., and Ward, D. C. Sequence analysis of the termini
of virion and replicative forms of minute virus of mice DNA suggests a
modified rolling hairpin model for autonomous parvovirus DNA replication.
J. Virol., 54: 171-177, 1985.
25. Toolan, H., and Ledinko, N. Growth and cytopathogenicity of H-viruses in
human and simian cell cultures. Nature (Lond.), 208: 812-813, 1965.
26. Rhode, S. L., and Paradiso, P. R. Parvovirus genome: nucleotide sequence
3578
Downloaded from cancerres.aacrjournals.org on July 8, 2017. © 1986 American Association for Cancer Research.
CELL TRANSFORMATION
27.
28.
29.
30.
31.
32.
33.
34.
35.
AND SUSCEPTIBILITY
of H-l and mapping of its genes by hybrid-arrested translation. J. Virol., 45:
173-184, 1983.
Rhode, S. L. Replication process of the parvovirus H-l. III. Factors affecting
H-l RF DNA synthesis. J. Virol., 14: 791-801, 1974.
Wolter, S., Richards, R., and Armentrout, R. W. Cell cycle-dependent
replication of the DNA of minute virus of mice, a parvovirus. Biochim.
Biophys. Acta, 007:420-431, 1980.
Tattersall, P. Susceptibility to minute virus of mice as a function of host-cell
differentiation. In: D. C. Ward and P. Tattersall (eds.). Replication of
Mammalian Parvoviruses, pp. 131-149. Cold Spring Harbor, NY: Cold
Spring Harbor Laboratory, Publisher, 1978.
Tattersall, P., and Bratton, J. Reciprocal productive and restrictive virus-cell
interactions of immunosuppressive and prototype strains of minute virus of
mice. J. Virol., 46:944-955, 1983.
Ron, D., and Tal, J. Coevolution of cells and virus as a mechanism for the
persistence of lymphotropic minute virus of mice in L-cells. J. Virol., 55:
424-430, 1985.
Spulhol/. B. A., and Tattersall, P. Interaction of minute virus of mice with
differentiated cells: strain-dependent target cell specificity is mediated by
intracellular factors. J. Virol.. 46:937-943, 1983.
Martin, R. G. The transformation of cell growth and transmogrification of
DNA synthesis by simian virus 40. Adv. Cancer Res., 34: 1-68, 1981.
Schutzbank, T., Robinson, R., Oren, M., and Levine, A. J. SV40 large tumor
antigen can regulate some cellular transcripts in a positive fashion. Cell, Ml:
481-490, 1982.
Ledinko, V. Hopkins, S., and Tonkin, H. Relationship between potentiation
36.
37.
38.
39.
40.
41.
42.
TO PARVOVIRUSES
of H-l growth by human adenovirus 12 and inhibition of the helper adenovirus by H-l. J. Gen. Virol., 5:19-31, 1969.
Cornells, J. J., Avalosse, H., Mousset, S., Namba, M.. and Rommelaere, J.
Selective destruction by parvoviruses of human and mouse fibroblasts trans
formed with gamma irradiation. Int. J. Radiât.Biol., 49:529, 1986.
Cornelis. J. J., Avalosse, B., Guetta, E., Tal, J., and Rommelaere, J. Trans
formation in vitro of rat cells correlates with an enhancement of their
susceptibility to the production of, and killing by parvovirus minute-virus-ofmice. Arch. Int. Physiol. Biochim., 92:14-15, 1984.
Abrahams, P. J., Huitema, B. A., and van der Eb, A. J. Enhanced reactivation
and enhanced mutagenesis of herpes simplex virus in normal human and
xeroderma pigmentosum cells. Mol. Cell. Biol., 4: 2341-2346, 1984.
de Foresta, F., Deleys-Hertoghs, H., Cornelis, J. J., Klein, B., and Romme
laere, J. Transformation by simian virus 40 sensitizes human skin fibroblasts
to the lytic effect of parvovirus H-l. Arch. Int. Physiol. Biochim., 92: 123,
1984.
Hilgers, G., Deleys-Hertoghs, J., Cornelis, J. J., Klein, B., and Rommelaere,
J. Probing human hypersensitivity syndromes with parvoviruses: aberrant
conditioned recovery responses in irradiated ataxia telangiectasia cells. Arch.
Int. Physiol. Biochim., 92: 140-141, 1984.
Arlett, C. F., and Harcourt, S. A. Survey of radiosensitivity in a variety of
human cell strains. Cancer Res., 40:926-932, 1980.
Teo, I. A., Broughton, B. C., Day, R. S., James, M. R., Karran, P., Mayne,
L. V., and Lehmann, A. R. A biochemical defect in the repair of alkylated
DNA in cells from an immunodeficient patient (46BR). Carcinogenesis
(Lond.), 4: 559-564, 1983.
3579
Downloaded from cancerres.aacrjournals.org on July 8, 2017. © 1986 American Association for Cancer Research.
0
Announcements
MEETING
OF THE RADIATION
The annual meeting of the Radiation Research Socie
ty will be held at the State University of Iowa, Iowa
City, on June 22—24,
1953. The Society will be the guest
of the University, and all meetings will be held on the
campus. The program will consist of: (1) Two symposia,
one on “TheEffects of Rwliation on Aqueous Solu
tions,― which includes
the following
speakers:
E. S. G.
Barren,
Edwin J. Hart, Warren
Garrison,
J. L. Magee,
and A. 0. Allen. The second is “PhysicalMeasurements
for Radiobiology―and companion talks by Ugo Fano,
Burton J. Moyer, G. Failla, L. D. Marinelli, and Payne
RESEARCH
S. Harris.
SOCIETY
(2) On Monday
night,
June
22, a lecture
by
Dr. L. W. Alvarez on meson physics has been tentative
ly scheduled. On Tuesday night, June 23, Dr. L. H.
Gray of the Hammersmith Hospital, London, will speak
on a topic to be announced. Dr. Gray's lecture is spon
sored by the Iowa Branch of the American Cancer Soci
ety. Those desiring to report original research in radia
tion effects, or interested in attending or desiring addi
tional information, please contact the Secretary of the
Society, Dr. A. Edelmann, Biology Department, Brook
haven National Laboratory, Upton, L.I., New York.
ERRATUM
The following correction should be made in the arti
cle by Beck and Valentine, “The
Aerobic Carbohydrate
Metabolism
Glycolysis
substitute
of Leukocytes
in Health and Leukemia.
I.
and Respiration,―
November,
1952, page 821;
for the last paragraph:
The data in Table 3 permit several interesting calcu
lations.
If one compares
the amount
of glucose
actually
disappearing with the sum of the amount equivalent to
lactic
acid
produced
plus
that
equivalent
to 02 con
by the glucose utilized by 16 per cent in CLL. If the as
sumption is made that, in this respect, the myeloid and
lymphoid celLsof leukemia are similar to those of nor
ma! blood, it may be that the computed
normal figure
represents a summation of the myeloid (M) and
lymphoid (L) cells that make up the normal leukocyte
population.
Thus,
if M = +0.27
and
L = —0.16 and
the normal differential is 65 per cent M and So per cent
L, then
0.65 (+0.27)
+ 0.35 (—0.16) = +0.12
sumption, it is seen that the amount of glucose “cleav
age products―exceeds the amount of glucose utilized b
a figure identical to the observed +0.12 for normal
12 per cent in N and 27 per cent in CML and is exceeded leukocytes.
308
Selective Killing of Simian Virus 40-transformed Human
Fibroblasts by Parvovirus H-1
Young Quan Chen, Françoise de Foresta, Jacqueline Hertoghs, et al.
Cancer Res 1986;46:3574-3579.
Updated version
E-mail alerts
Reprints and
Subscriptions
Permissions
Access the most recent version of this article at:
http://cancerres.aacrjournals.org/content/46/7/3574
Sign up to receive free email-alerts related to this article or journal.
To order reprints of this article or to subscribe to the journal, contact the AACR Publications
Department at [email protected].
To request permission to re-use all or part of this article, contact the AACR Publications
Department at [email protected].
Downloaded from cancerres.aacrjournals.org on July 8, 2017. © 1986 American Association for Cancer Research.
Téléchargement