Download as pdf or txt
Download as pdf or txt
You are on page 1of 6

JOURNAL OF VIROLOGY, Jan. 1994, p. 346-351 Vol. 68, No.

1
0022-538X/94/$04.00+0
Copyright © 1994, American Society for Microbiology

A Single Chicken Anemia Virus Protein Induces Apoptosis


M. H. M. NOTEBORN,'* D. TODD,2 C. A. J. VERSCHUEREN,' H. W. F. M. DE GAUW,' W. L. CURRAN,2
S. VELDKAMP,3 A. J. DOUGLAS,2 M. S. McNULTY,3 A. J. VAN DER EB,' AND G. KOCH3
Laboratory for Molecular Carcinogenesis, Sylvius Laboratory, Leiden University, 2300 RA Leiden,' and Virology
Department, DLO-Central Veterinary Institute, 8200 AJ Lelystad, The Netherlands, and Veterinary Sciences Division,
Stormont, Belfast BT4 3SD, Northern Ireland2
Received 19 July 1993/Accepted 19 October 1993

Chicken anemia virus (CAV) causes cytopathogenic effects in chicken thymocytes and cultured transformed
mononuclear cells via apoptosis. Early after infection of chicken mononuclear cells, the CAV-encoded protein

Downloaded from http://jvi.asm.org/ on November 21, 2018 by guest


VP3 exhibits a finely granular distribution within the nucleus. At a later stage after infection, VP3 forms
aggregates. At this point, the cell becomes apoptotic and the cellular DNA is fragmented and condensed. By
immunogold electron microscopy VP3 was shown to be associated with apoptotic structures. In vitro,
expression of VP3 induced apoptosis in chicken lymphoblastoid T cells and myeloid cells, which are susceptible
to CAV infection, but not in chicken embryo fibroblasts, which are not susceptible to CAV. Expression of a
C-terminally truncated VP3 induced much less pronounced apoptosis in the chicken lymphoblastoid T cells.

Chicken anemia virus (CAV) transiently causes severe ane- isolated in Germany (34). For DNA transfections, we used the
mia due to destruction of erythroblastoid cells and immuno- cell line MDCC-MSB1; the myeloid cell line LSCC-HD11
deficiency due to depletion of cortical thymocytes in young transformed by avian myeloblastosis virus (4), which was kindly
chickens (10, 38). Jeurissen et al. (11) have provided evidence provided by T. Graf; and primary chicken embryo fibroblasts
that the observed depletion of the thymocytes occurs via (CEFs).
CAV-induced apoptosis. Apoptosis is considered to be a Immunoblotting. The nuclear protein preparations that
physiological process of cell depletion that is part of the were used for immunoblotting were produced as follows.
homeostatic regulation of normal tissues (5). CAV in addition MDCC-MSB1 cells that had been infected at a multiplicity of
to several other viruses, such as human immunodeficiency virus infection of about 0.2 median tissue culture infective dose per
type 1 (2) and parvovirus B19 (19), seems to use apoptosis to cell were collected by centrifugation at 48 h after infection, and
exert its cytopathogenic effect. a nucleus-rich pellet was obtained (32). Nuclear material that
CAV is a small virus with a diameter of about 23 nm and had been resuspended in 10 mM Tris HCl-1 mM EDTA (pH
contains a circular single-stranded DNA of 2.3 kb (27). CAV 8.0), dispersed by thorough sonication, and solubilized with 2%
multiplies via a circular double-stranded DNA replicative sodium dodecyl sulfate (SDS) was fractionated by gel filtration
intermediate, which was recently cloned (18, 21). The cloned through a column (45.1 by 5 cm) containing Sephacryl S200.
CAV genome was proven to be representative for CAV Noninfected MDCC-MSB1 cells were similarly processed for
isolates collected worldwide (23, 29). A polycistronic polyade- control purposes.
nylated mRNA (22) which comprises three overlapping open Proteins were separated by SDS-polyacrylamide gel electro-
reading frames encoding proteins VP1 (51.6 kDa), VP2 (24.0 phoresis, electroblotted, and subjected to immunodetection as
kDa), and VP3 (13.6 kDa) is transcribed from the CAV described by Todd et al. (28). Blots were incubated with a
genome. 1:10,000 dilution of an immunoglobulin fraction (approximate-
In the present paper, we report studies on the expression of ly 28.7 mg/ml), which had been produced by precipitation with
the VP3 protein in CAV-infected cells. VP3 is a putative CAV (NH4)2SO4, of the CAV-specific monoclonal antibody (MAb)
protein of 121 amino acids and contains two proline-rich 3B1 (17) and a 1:1,000 dilution of a peroxidase-labeled goat
stretches and two positively charged regions. So far, there is no anti-mouse conjugate.
evidence that VP3 is present in purified CAV particles (27). Immunoperoxidase assay and immunofluorescence assay.
However, VP3 seems to be important for the virus life cycle, as MDCC-MSB1 cells, which were synchronized by growing in
found by VP3-deletion studies (19a). By way of immunolabel- medium without leucine and fetal bovine serum, were infected
ing experiments with CAV-infected chicken lymphoblastoid T with CAV at a multiplicity of infection of about 1 median tissue
cells in vitro, we present evidence that VP3 accumulates in culture infective dose of virus per cell. The cells were harvested
condensed DNA. We also show that expression of VP3 alone at 0, 16, 24, 32, and 64 h after infection; spun on coverslips; and
is sufficient to induce apoptosis in chicken mononuclear cells. fixed with 100% acetone. Immunoperoxidase staining was
carried out with a 100-fold dilution of the MAb CVI-CAV-85.1
MATERIALS AND METHODS (85.1) (21) and a 500-fold dilution of a peroxidase-labeled
rabbit anti-mouse immunoglobulin G conjugate in phosphate-
Viruses and cells. The chicken lymphoblastoid T-cell line buffered saline, as described by Jeurissen et al. (9). Immuno-
MDCC-MSBI, transformed by Marek's disease virus (1), was fluorescence assays were carried out on DNA-transfected cells
infected with the CAV-Cux-1 isolate, which was originally by using MAb CVI-CAV-85.1 as described by Noteborn et al.
(20).
*
Corresponding author. Mailing address: Laboratory for Molecular Immunoelectron microscopy. MDCC-MSB1 cells were in-
Carcinogenesis, Sylvius Laboratory, Leiden University, P.O. Box 9003, fected with CAV at a multiplicity of infection of about 0.2
2300 RA Leiden, The Netherlands. Phone: (31) 71 276113. Fax: (31) median tissue culture infective dose of virus per cell. The
71 276125. infected and noninfected control cells were harvested 48 h
346
VOL. 68? 1994

KDa

925-
66-2-

45-

31-
A B
o

24

:-y.oW!.;_0-,X0'Viste<;r
APOPTOSIS INDUCED BY A SINGLE CAV PROTEIN

i.-

s
i:ffEt-.

4S-

.:
s
to .-0t; C0.:
fi:f

0::::
16

32
347

Downloaded from http://jvi.asm.org/ on November 21, 2018 by guest


.$_
.7 .*.2_ t_
S.z_:

..g ?S - .
_r

::0 ::v _
21-5- A.su ;00: it. _t
F

14-4-
40 56

FIG. 1. Immunoblot of nuclear protein fractions from noninfected


(lane A) and CAV-infected (lane B) MDCC-MSBl cells. The blot was
initially allowed to react with MAb 3B1, which is directed against VP3.

after infection. Thin-section electron microscopy and immu- 64 64


nogold labeling were carried out as described by McNulty et al. D : .
(16). The grids were stained with a 10,000-fold dilution of the
CAV-specific MAb 3B1 and a 1:50 dilution of 15-nm gold-
labeled goat anti-mouse immunoglobulin G conjugate.
Construction of VP3 expression vectors. Plasmid DNA
manipulations were performed essentially according to the
methods described by Maniatis et al. (14). The CAV sequences
originated from plasmid pCAV/E, described by Noteborn et al.
(21). FIG. 2. Indirect immunoperoxidase staining of CAV-infected
The sequences encoding VP3 were cloned in the expression MDCC-MSB1 cells with antibody 85.1 directed against VP3. The cells
were fixed and stained 0, 16, 24, 32, 40, 56, and 64 h after infection.
vector pRSV-H20, which contains the Rous sarcoma virus
promoter. From plasmid pEP-VP3 (19a) we isolated a 0.46-kb
BamHI-EcoRI DNA fragment with CAV DNA sequences
from positions 427 to 868 (21). It contains the coding region of
VP3, a 58-bp 5'-flanking sequence, and a 25-bp 3-flanking HindIII-SalI DNA linker. Plasmid pRSV-H20 was digested
sequence. Two synthetic DNA oligomers, 5'-GATCCAACCC with BglII and Sall and treated with calf intestine alkaline
GGGTFG-3' and 5'-AATTCAACCCGGGTTG-3', were hy- phosphatase, and a 4.3-kb fragment was isolated. The HindIll-
bridized to form a double-stranded BamHI-EcoRl DNA Sall DNA linker and the 0.38-kb BamHI-HindIII DNA frag-
linker. The vector pRSV-H20 was linearized with BglII and ment were ligated with the 4.3-kb BglII-SalI DNA fragment.
treated with calf intestine alkaline phosphatase, after which a The final construct, pRSV-tr, was analyzed by restriction
4.3-kb fragment was isolated. The BamHI-EcoRI DNA linker enzyme digestions and by sequencing of the newly introduced
and the 0.46-kb BamHI-EcoRI DNA fragment were ligated bases (see Fig. 4).
with the 4.3-kb BglII DNA fragment. The final construct, DNA transfection and analysis of cellular DNA. Plasmid
pRSV-VP3, contains the VP3 coding region under the control DNA was purified by centrifugation in a CsCl gradient and by
of the Rous sarcoma virus promoter as shown in Fig. 4 and was column chromatography in Sephacryl S500 (Pharmacia). For
analyzed by restriction enzyme digestions. DNA transfections of MDCC-MSB1 and LSCC-HD11 cells,
A truncated VP3 product was made by deleting 11 codons at the DEAE-dextran method of Luthman and Magnusson (13),
the 3' terminus of the VP3 coding sequences and introducing in which the chloroquine treatment was replaced with a
a new stop codon. Plasmid pEP-VP3 was digested with BamHI dimethyl sulfoxide boost, was used. CEFs were transfected
and HindIlI. The 0.38-kb BamHI-HindIIl DNA fragment was with DNA according to the calcium phosphate precipitation
isolated. Two synthetic DNA oligomers, 5'-AGCTTGATTAC method of Graham and Van der Eb (8). DNA extracted from
CACTACTCCCTGAG-3' and 5'-TCGACTCAGGGAGTAG DNA-transfected MDCC-MSB1 cells was analyzed as de-
TGGTAATCA-3', were hybridized to form a double-stranded scribed by Jeurissen et al. (11).
348 NOTEBORN ET AL. J. VIROL.

A.

Downloaded from http://jvi.asm.org/ on November 21, 2018 by guest


B.

1-M N A L Q E D T P P G P S T V
F R P P T S S R P L E T P H C
R E I R I G I A G I T I T L S
L C G C A N A R A P T L R S A
T A D N S E S T G F K N V P D
L R T D Q P K P P S K K R S C
D P S E Y R V S E LK E S L I
T T T P S R P R T A K R R I R
L-1 21
FIG. 3. Immunogold labeling of electron-dense nuclear inclusions FIG. 4. (A) Schematic representation of the expression vectors
in CAV-infected MDCC-MSB1 cells. Infected cells harvested 48 h pRSV-VP3 and pRSV-tr. Both vectors are constructed as described in
after infection were embedded in Lowicryl and allowed to react with Materials and Methods. The CAV sequence encoding VP3 is drawn as
MAb 3B1, which is directed against VP3. Magnification, x 26,000. a filled box, that for VP3-tr is drawn as a striped box, the Rous sarcoma
virus promoter region is drawn as a dotted box, and the simian virus 40
sequences, containing a poly(A) addition site, is drawn as an open box.
RESULTS (B) Amino acid sequence of VP3. The 11 amino acids which were
deleted from the C terminus of the truncated form of VP3 are
Expression of VP3 during CAV infection of cultured cells. underlined. The proline residues are in italics, and the lysine and
We have analyzed the expression of VP3 during a CAV arginine residues are in boldface type.
infection of chicken lymphoblastoid T cells. The CAV-infected
cells were analyzed by immunoblotting and an immunoperoxi-
dase assay using CAV-specific MAbs 3B1 (17) and 85.1 (21). structures in the nucleus of CAV-infected cells (Fig. 3).
Both MAbs are specific for CAV VP3, since these specifically Apparently, VP3 accumulates in these apoptotic structures.
bind to recombinant VP3 synthesized with a baculovirus VP3 induces apoptosis in cultured avian mononuclear cells.
system (19a). Immunoblotting experiments showed that frac- To establish whether VP3 alone is able to induce apoptosis, the
tions of the nuclear extracts of CAV-infected cells that eluted expression vector pRSV-VP3 was constructed (Fig. 4). This
close to the void volume of the Sephacryl S200 column vector contains CAV DNA sequences encoding only VP3.
contained a 16-kDa protein that specifically reacted with MAb After transfection with DNA of pRSV-VP3, VP3 was tran-
3B1 (Fig. 1). Under the immunodetection conditions used, siently expressed in cultured chicken lymphoblastoid T cells.
faint nonspecific staining of other proteins in the profiles The cells were harvested after 48 h, acetone fixed, and stained
derived from infected and noninfected cells was also observed. with MAb 85.1. In addition, the cells were treated with
Expression of sequences encoding VP3 in a baculovirus system propidium iodide (PI), which is known to stain intact nuclei
yields a protein with an identical molecular mass (19a). strongly but apoptotic nuclei relatively weakly (26). CAV-
Immunoperoxidase staining of CAV-infected MDCC-MSB1 infected cells containing apoptotic inclusion structures, which
cells with MAb 85.1 was carried out at several time points after were comparable to the structures observed at 64 h after
infection (Fig. 2). As early as 24 h after infection, VP3 was infection (Fig. 2), were weakly and irregularly stained by PI
present in the nucleus. The distribution of VP3 was very faint (data not shown). We observed 60% of the transfected cells
and finely granular. Later after infection, the granules in- with nuclei exhibiting a very finely granular distribution of
creased in size, and gradually aggregates appeared. The aggre- VP3. The DNA of cells with this type of VP3 staining was
gates were especially prominent around 60 h after infection. At always clearly stained by PI. In contrast, 40% of VP3-positive
this stage of infection, the CAV-specific cytopathogenic effect cells contained aggregates of VP3, and their DNA invariably
was clearly visible. stained weakly and irregularly with PI. Three days after
The aggregates containing VP3 resemble the electron-dense transfection, almost all VP3-positive cells contain VP3 aggre-
structures described by Jeurissen et al. (11) that occur during gates and negatively PI-stained DNA (Fig. 5). In contrast,
the apoptotic process of CAV-infected cells. We have shown expression of a C-terminally truncated VP3 (Fig. 4) yielded
by immunogold electron microscopy that MAb 3B1 binds almost no apoptosis in MDCC-MSB1 cells. Three days after
strongly to many, but not all, of the electron-dense apoptotic transfection, 80 to 90% of the cells expressing truncated VP3
VOL. 68, 1994 APOPTOSIS INDUCED BY A SINGLE CAV PROTEIN 349

A B m 1 2 3 4

slot-

Downloaded from http://jvi.asm.org/ on November 21, 2018 by guest


C

FIG. 6. DNA fragmentation in MDCC-MSBI cells undergoing


VP3-mediated cell death. DNAs from CAV-infected MDCC-MSBI
E F cells which were harvested 72 h after infection (lane 1) and from
MDCC-MSB1 cells transfected with DNA of plasmids pRSV-VP3
(lane 2), pRSV-tr (lane 3), and pRSV-CAT (lane 4) were analyzed on
a 1% agarose-ethidium bromide gel. Lane m, DNA markers (lambda-
HindIll fragments; Promega). About 10% of the MDCC-MSBI cells
expressed VP3 or VP3-tr protein, as determined by immunofluores-
cence (data not shown).

DISCUSSION
CAV induces cell death of chicken lymphoblastoid T cells
FIG. 5. Indirect immunofluorescence of MDCC-MSB1 cells trans- via apoptosis (11). By use of immunoassays and DNA analysis,
fected with plasmid pRSV-VP3 (A to D) or pRSV-tr (E and F). The we have established that the putative open reading frame for
cells were harvested 48 h (A and B) or 72 h (C to F) after transfection,
fixed, and stained either with MAb 85.1 directed against VP3 (A, C, VP3 of the CAV genome (18, 21) is indeed expressed in
and E), or with PI (B, D, and F). CAV-infected cells. Early after infection, VP3 is colocalized
with cellular DNA. Later after infection, VP3 is bound to
nuclear aggregates that are known to be apoptotic bodies (5,
1 1). Expression of only VP3 in MDCC-MSB1 cells mimics the
still have normally PI-stained nuclei (Fig. 5). At this time point, process of CAV-induced apoptosis. In general, apoptosis is a
the cellular DNA of MDCC-MSB1 cells expressing VP3 process leading to cell death that occurs rapidly (36). The
showed the apoptosis-specific nucleosomal laddering. This majority of CAV-infected MDCC-MSB1 cells become apop-
laddering was proven to be much less in DNA from cells totic within 64 h after infection. In vitro expression of VP3 in
transfected with plasmid pRSV-tr or absent in DNA from transfected cells causes CAV-like apoptosis within 3 days.
MDCC-MSB1 cells transfected with plasmid pRSV-CAT, These data suggest that VP3 alone can trigger the apoptotic
which expressed the chloramphenicol transferase gene (Fig. 6). pathway in CAV-infected cells.
We conclude that expression of VP3 alone can induce the How does the expression of VP3 cause apoptosis? In
apoptotic process that is observed during CAV infection and general, apoptosis can be induced by a variety of viral and
results in cell death. nonviral external stimuli, which can converge on a common
To test whether VP3 can also induce apoptosis in other intracellular pathway, resulting in the activation of an endonu-
chicken cell types, it was expressed in the myeloid cell line clease (15). The amino acid sequence of VP3 does not show
LSCC-HD11 and in primary CEFs. LSCC-HD11 is susceptible any distinct homology with the sequences of the receptors that
to CAV, whereas CEFs are not. Transfection of DNA encod- mediate apoptosis (6, 24, 25, 35) or of c-myc and p53 oncogene
ing VP3 in LSCC-HD1 1 cells yielded the formation of VP3 products (7, 37), known to induce apoptosis. So, it is unlikely
aggregates and weak and irregular staining of the cellular DNA that VP3 mimics the activity of one of these cellular proteins.
by PI. CEFs expressing VP3 revealed only granular staining of Further experiments have to be carried out to prove whether
VP3 and strong staining with PI (Fig. 7). This suggests that VP3 causes apoptosis by inhibition of the antiapoptosis activity
VP3 can induce apoptosis in LSCC-HD 11 cells but not in of the oncogene product Bcl-2 (33). We also cannot exclude
CEFs, or at least to a much lesser extent. the possibility that VP3 induces apoptosis by binding to one of
350 J. VIROL.
NOTEBORN ET AL.J.Vto.

densation of the cellular DNA. A disturbance of the supercoil-


A B ing organization by VP3 might be caused by its high proline
content. Induction of apoptosis by disruption of the superchro-
matin structure was described for apoptotic rat thymocytes
(31) and rat ventral prostate cells after castration (12). An
alternative explanation might be that VP3 interacts directly
with the histone protein HI, which binds to the intranucleo-
somal DNA. For instance, binding of VP3 to this region
instead of histone HI might enable endonucleases to digest the
intranucleosomal DNA. Arends et al. (3) have observed that
the oligonucleosomes become depleted of histone HI during
the apoptotic process.
Under our experimental conditions, the expression of VP3
in CEFs and cells of the human epithelia] cell line HeLa (data
not shown) did not result in the observed disruption of the

Downloaded from http://jvi.asm.org/ on November 21, 2018 by guest


cellular DNA. However, neither cell line is susceptible to
CAy, in contrast to MDCC-MSBi and HDll cells. It might
well be that VP3 can cause degradation of cellular DNA only
in cells that are susceptible to CAy. An alternative explanation
might be that VP3 induces a cascade of events, resulting in
single-strand modification, i.e., the generation of alkaline-
sensitive sites in the cellular DNA. Apparently, in lymphocytes
E F and monocytes these modifications lead to double-stranded
cleavage, whereas HeLa cells and CEFs remain arrested at the
stage of single-strand modification. This would account for the
observation that lymphocytes and monocytes fragment their
DNA within hours after treatment with VP3. This explanation
was offered by Tomei (30) for the glucocorticoid-induced
apoptosis observed in lymphocytes but not in fibroblasts and
HeLa cells.
We conclude that a single CAV protein can induce apoptosis
in chicken lymphoblastoid T and myeloid cells. Experiments
that will unravel the mechanism by which VP3 induces cell
death are under way.

ACKNOWLEDGMENTS
FIG. 7. Indirect immunofluorescence of LSCC-HDI I cells and
We thank Hans van Ormondt for critically reading the manuscript,
CEFs which were transfected with plasmid pRSV-VP3, which encodes
VP3. The LSCC-HDII
Alt Zantema for helpful discussions, and Frans van Bussel for his
cells (A to D) were harvested 48 h after
transfection, and the CEFs
excellent drawing work.
(E and F) were harvested 72 h after
transfection. The cells fixed and stained with MAb 85.1 directed
This research was made possible partially with research grants from
were
The Netherlands Ministry of Economy Affairs and Aesculaap By,
against VP3 (A, C, and E) and with PI (B3, D, and F).
Boxtel, The Netherlands.
REFERENCES
the membrane-bound receptors. However, this be
seems to
1. Akiyama, Y., and S. Kato. 1974. Two cell lines from lymphomas of
unlikely, as VP3 is located in the nucleus. VP3 could induce Mareks disease. Biken J. 17:105-117.
apoptosis by regulating genes involved in apoptosis either 2. Ameisen, J. C., and A. Capron. 1991. Cell dysfunction and
directly, via the induction of endonucleases, or indirectly, via depletion in AIDS: the programmed cell death hypothesis. Immu-
the induction of, e.g., the oncogene c-myc or p53 or repression nol. Today 12:102-105.
of the putative oncogene bcl-2. 3. Arends, M. J., R. G. Morris, and A. H. Wyllie. 1990. Apoptosis: the
The results of the immunoassays revealed that VP3 is strictly role of the endonucleases. Am. J. Pathol. 136:593-608.
located within the cellular chromatin structures. The basic
4. Beug, H., A. Von Kirkbach, G. Doiderlein, J. F. Conscience, and T.
character of VP3 and the C-terminal
Graf. 1979. Chicken hematopoietic cells transformed by seven
region in particular might strains of defective avian leukemia viruses display three different
be the that VP3 has binding properties to the cellular
reason
phenotypes of differentiation. Cell 18:375-390.
DNA. The results obtained with the VP3-tr mutant showed
5. Cohen, J. J. 1993. Apoptosis. Immunol. Today 14:126-130.
that deletion of the C-terminal basicregion significantly re- 6. Ellis, R. E., J. Yuan, and H. R. Horvitz. 1991. Mechanisms and
duced the
apoptotic activity of VP3. Does VP3 act as an functions of cell death. Annu. Rev. Cell Biol. 7:663-698.
endonuclease? Findings with CEFs argue against this idea, and 7. Evan, G. I., A. H. Wyllie, C. S. Gilbert, T. D. Littlewood, H. Land,
computer analyses did not reveal any sequence homology of M. Brooks, C. M. Waters, L. Z. Penn, and D. C. Hancock. 1992.
VP3 with one of the known endonucleases. Induction of apoptosis in fibroblasts by c-myc protein. Cell 69:119-
The small size of VP3 and its rather basic character may
128.
allow it to interact with histone and/or nonhistone
8. Graham, F. L., and A. J. Van der Eb. 1973. A new technique for
proteins the assay of infectivity of human adenovirus 5 DNA. Virology
within the chromatin structure. Thus, the presence of VP3 in
52:456-467.
the chromatin result in breakdown of the
structure may a
9. Jeurissen, S. H. M., E. M. Janse, S. Ekino, P. Nieuwenhuis, G.
supercoiling organization. Eventually, these modifications of Koch, and G. F. de Boer. 1988. Monoclonal antibodies as probes
the chromatin structure may lead to fragmentation and con- for defining cellular subsets in the bone marrow, thymus, bursa of
VOL. 68, 1994 APOPTOSIS INDUCED BY A SINGLE CAV PROTEIN 351

Fabricius, and spleen of the chicken. Vet. Immunol. Immuno- chicken anemia virus by DNA hybridization and polymerase chain
pathol. 19:225-238. reaction. Avian Pathol. 21:107-118.
10. Jeurissen, S. H. M., J. M. A. Pol, and G. F. de Boer. 1989. 24. Pinching, A. J., and K. E. Nye. 1990. Defective signal transduc-
Transient depletion of cortical thymocytes induced by chicken tion-a common pathway for cellular dysfunction in HIV infec-
anaemia agent. Thymus 14:115-123. tion? Immunol. Today 11:256-259.
11. Jeurissen, S. H. M., F. Wagenaar, J. M. A. Pol, A. J. van der Eb, 25. Smith, C. A., G. T. Williams, R. Kingston, E. J. Jenkinson, and
and M. H. M. Noteborn. 1992. Chicken anemia virus causes J. J. T. Owen. 1989. Antibodies to CD3/TCR complex induce
apoptosis of thymocytes after in vivo infection and of cell lines death by apoptosis in immature T cells in thymic culture. Nature
after in vitro infection. J. Virol. 66:7383-7388. (London) 337:181-183.
12. Kyprianou, N., and J. T. Isaacs. 1988. Activation of programmed 26. Telford, W. G., L. E. King, and P. J. Fraker. 1992. Comparative
cell death in the rat ventral prostate after castration. Endocrinol- evaluation of several DNA binding dyes in the detection of
ogy 122:552-662. apoptosis-associated chromatin degradation by flow cytometry.
13. Luthman, H., and G. Magnusson. 1983. High efficiency polyoma Cytometry 13:137-143.
DNA transfection of chloroquine treated cells. Nucleic Acids Res. 27. Todd, D., J. L. Creelan, D. P. Mackie, F. Rixon, and M. S.
11:1295-1308. McNulty. 1990. Purification and biochemical characterisation of
14. Maniatis, T., E. F. Fritsch, and J. Sambrook. 1982. Molecular chicken anaemia agent. J. Gen. Virol. 71:819-823.

Downloaded from http://jvi.asm.org/ on November 21, 2018 by guest


cloning: a laboratory manual. Cold Spring Harbor Laboratory, 28. Todd, D., J. Hull, and J. McNair. 1987. Antigenically important
Cold Spring Harbor, N.Y. proteins of Aujeszky's disease (pseudorabies) virus identified by
15. Martz, E., and D. M. Howell. 1989. CTL: virus control cells first immunoblotting. Arch. Virol. 96:215-224.
and cytolytic cells second? Immunol. Today 10:79-86.
16. McNulty, M. S., T. J. Connor, F. McNeilly, M. F. McLoughlin, and 29. Todd, D., K. A. Mawhinney, and M. S. McNulty. 1992. Detection
K. S. Kirkpatrick. 1990. Preliminary characterization of isolates of and differentiation of chicken anemia virus isolates by using the
chicken anemia agent from the United Kingdom. Avian Pathol. polymerase chain reaction. J. Clin. Microbiol. 30:1661-1666.
19:67-73. 30. Tomei, L. D. 1991. Apoptosis: a program for death or survival, p.
17. McNulty, M. S., W. L. Curran, D. Todd, and D. P. Mackie. 1990. 279. In L. D. Tomei and F. 0. Cope (ed.), Apoptosis: the
Chicken anemia agent: an electron microscopic study. Avian Dis. molecular basis of cell death. Cold Spring Harbor Laboratory,
34:736-743. Cold Spring Harbor, N.Y.
18. Meehan, B. M., D. Todd, J. L. Creelan, J. A. P. Earle, E. M. Hoey, 31. Umansky, S. R., B. A. Korol, and P. A. Nelipovich. 1981. In vivo
and M. S. McNulty. 1992. Characterization of viral DNAs from DNA degradation in thymocytes of gamma-irradiated or hydro-
cells infected with chicken anemia agent: sequence analysis of the cortisone-treated rats. Biochim. Biophys. Acta 655:9-17.
cloned replicative form and transfection capabilities of cloned 32. Van Bleck, G. M., and S. G. Natheson. 1990. Isolation of an
genome fragments. Arch. Virol. 124:301-309. endogenously processed immunodominant viral peptide from the
19. Morey, A. L., D. J. P. Ferguson, and K. A. Fleming. 1993. class I H-2K molecule. Nature (London) 348:213-216.
Ultrastructural features of fetal erythroid precursors infected with 33. Vaux, D. L., H. L. Aguila, and I. L. Weissmann. 1992. Bcl-2
parvovirus B19 in vitro: evidence of cell death by apoptosis. J. prevents death of deprived cells but fails to prevent apoptosis in
Pathol. 169:213-220. targets of cell-mediated killing. Int. Immunol. 4:821-824.
19a.Noteborn, M. H. M. Unpublished results. 34. Von Bulow, V., B. Fuchs, B. Viclitz, and H. Landgraf. 1983.
20. Noteborn, M. H. M., G. F. de Boer, A. Kant, G. Koch, J. L. Bos, A. Fruhsterblichkeitssyndrom bei Kuken nach Doppelinfektion mit
Zantema, and A. J. van der Eb. 1990. Expression of avian dem Virus des Marekschen Krankheit (MDV) und einen Anamie-
leukemia virus env-gp85 in Spodoptera frugiperda cells by use of Erreger (CAA). J. Vet. Med. B 30:742-750.
a baculovirus expression vector. J. Gen. Virol. 71:2641-2648. 35. Watanabe-Fukunaga, R., C. I. Brannan, N. G. Copeland, N. A.
21. Noteborn, M. H. M., G. F. de Boer, D. J. Van Roozelaar, C. Jenkins, and S. Nagata. 1992. Lymphoproliferative disorder in
Karreman, 0. Kranenburg, J. G. Vos, S. H. M. Jeurissen, R. C. mice explained by defects in Fas antigen that mediates apoptosis.
Hoeben, A. Zantema, G. Koch, H. van Ormondt, and A. J. van der Nature (London) 356:314-317.
Eb. 1991. Characterization of cloned chicken anemia virus DNA 36. Wyllie, A. H., J. F. R. Kerr, and A. R. Currie. 1980. Cell death: the
that contains all elements for the infectious replication cycle. J. significance of apoptosis. Int. Rev. Cytol. 68:251-306.
Virol. 65:3131-3139. 37. Yonish-Rouach, E., D. Resnitzky, J. Lottem, L. Sachs, A. Kimchi,
22. Noteborn, M. H. M., 0. Kranenburg, A. Zantema, G. Koch, G. F. and M. Oren. 1991. Wild-type p53 induces apoptosis of myeloid
de Boer, and A. J. van der Eb. 1992. Transcription of the chicken leukaemic cells that is inhibited by interleukin-6. Nature (London)
anemia virus (CAV) genome and synthesis of its 52-kDa protein. 352:345-347.
Gene 118:267-271. 38. Yuasa, N., T. Taniguchi, and I. Yoshida. 1979. Isolation and some
23. Noteborn, M. H. M., C. A. J. Verschueren, D. J. van Roozelaar, S. properties of an agent inducing anaemia in chicks. Avian Dis.
Veldkamp, A. J. van der Eb, and G. F. de Boer. 1992. Detection of 23:366-385.

You might also like