Download T-Cell Response to B-Cells and Epstein-Barr

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts

Phagocyte wikipedia , lookup

T cell wikipedia , lookup

Psychoneuroimmunology wikipedia , lookup

Lymphopoiesis wikipedia , lookup

Hepatitis B wikipedia , lookup

Adaptive immune system wikipedia , lookup

Polyclonal B cell response wikipedia , lookup

Innate immune system wikipedia , lookup

Molecular mimicry wikipedia , lookup

Sjögren syndrome wikipedia , lookup

Immunomics wikipedia , lookup

X-linked severe combined immunodeficiency wikipedia , lookup

Cancer immunotherapy wikipedia , lookup

Adoptive cell transfer wikipedia , lookup

Immunosuppressive drug wikipedia , lookup

Transcript
[CANCER RESEARCH 41, 4210-4215,
0008-5472/81
/0041-OOOOS02.00
November 1981]
T-Cell Response to B-Cells and Epstein-Barr Virus Antigens in
Infectious Mononucleosis1
E. Klein,2 I. Ernberg, M. G. Masucci,3 R. Szigeti,4 Y. T. Wu,5 G. Masucci, and E. Svedmyr
Department of Tumor Biology, Karolinska Institute!, S-104 01 Stockholm, Sweden
Abstract
After Epstein-Barr virus (EBV) infection in vivo, B-cells with
latent virus infection persist indefinitely through life. These cells
grow in vitro on explantation and can be established as immor
tal B-cell lines.
To reconcile the unlimited growth potential in vitro with the
maintenance of a low proportion of B-cells infected by EBV in
vivo, a strict in vivo control mechanism has to be postulated.
Certain aspects of this control are apparent when the primary
infection is followed by infectious mononucleosis. This is char
acterized by lymphocytosis and the presence of activated Tcells. The T-cell proliferation is probably the manifestation of
the immune response against EBV antigens. However, the
reaction of T-cells upon encounter of B-blasts is also likely to
contribute to the events. At present, it is difficult to detect an
EBV-specific component in the action of the T-cells in the acute
phase of mononucleosis exerted on B-cells. However, for the
clinical course of the disease the activation of T-cells is impor
tant. The activated T-cells may control and also eliminate the
B-cells infected by EBV. In addition to the immunity which
develops during the disease, the immunoregulatory mechanism
is likely to have a role in the inhibition of B-cell proliferation.
(12). Depending on the geographic area, approximately 60 to
95% of a population has been exposed to the virus by the age
of 10 years and up to 100% by the age of 40 (11, 13, 38). In
young adolescents, the primary infection elicits a clinically
acute syndrome in nearly one-half of persons which is char
acterized by a massive lymphocyte proliferation, acute IM.
Independently of the clinical picture, the primary infection
has 2 lasting consequences: (a) persisting antibody titers (IgG
class) against at least 3 virus-associated antigens (viral capsid
antigens, membrane antigens, and EBNA); and (b) persistence
of the virus in a nonlytic latent form in a proportion of Blymphocytes. Most probably, the virus also persists in cells
around the oral cavity since virus was isolated from the saliva
of seropositive individuals and during acute IM (3, 22). Epithe
lial cells of the oropharynx or salivary glands could be a source
of the virus (18, 25).
Characteristics of BEBV-Cells
The spontaneous cell lines established from BEBv-cells in the
peripheral blood resemble those derived from B-cells trans
formed in vitro by EBV (26, 27). These explanted cells are
"immortal" and divide about every 24 to 48 hr.
Uncontrolled proliferation of BEBv-cells in vivo would lead to
considerable lymphocytosis. Consequently, either the charac
The majority of individuals of the human species harbors BEBv- teristics of the BEBv-cell change upon explantation, or in culture
cells. Such cells have an unlimited proliferative potential in the cells are released from the in vivo control mechanisms.
vitro. BEBv-Cells are kept under rigorous control in vivo, and
Rickinson ef al. (31) proposed that EBV is harbored in a
their uncontrolled proliferation is very rare. Such "accidents"
latent form in nontransformed cells, i.e., in cells which do not
are due to either an escape from or an impairment of the
have proliferative capacity. Upon in vitro explantation of the
control mechanisms. The former event may occur in Burkitt's
lymphocytes, virus production and infection of yet uninfected
lymphoma and the latter in the X-linked lymphoproliferative
B-cells occurs. This hypothesis was based on the finding that
syndrome (30).
phosphonoacetic acid and neutralizing antibodies reduced the
spontaneous outgrowth of cell lines from the blood of acute IM
Primary EBV Infection in Vivo and its Consequences
patients or seropositive donors (32, 33). Although there are
indications that the "2-step model" for spontaneous in vitro
As a rule, EBV infection in early childhood is clinically silent
transformation may be valid, cells with the characteristics of
the in w'fro-established EBV cells probably circulate in the
The relationship between EBV6 and its natural host is unique.
' This project was supported by Contract No. NO1 CP 33316 to Dr. G. Klein
from the Division of Cancer Cause and Prevention. National Cancer Institute, and
by the Swedish Cancer Society.
2 Recipient of Grant 5 R01 CA 25250-02 awarded by the National Cancer
Institute.
3 Recipient of a fellowship from the Blanceflor Boncompagni-Ludovici
dation, Stockholm, Sweden.
' Recipient of the Guest Scholarship
Foun
of the Swedish Institute, on leave from
the Second Department of Paediatrics, Semmelweis Medical University, Buda
pest, Hungary.
5 Recipient of a fellowship from the Ministry of Education of the People's
Republic of China.
6 The abbreviations used are: EBV, Epstein-Barr virus; BEBv-cells. B-cells
(lymphocytes) infected by Epstein-Barr virus; IM, infectious mononucleosis;
EBNA, Epstein-Barr virus-determined nuclear antigen; LMI, leukocyte migration
inhibition; UP, leukocyte migration inhibition factor.
Received January 5, 1981 : accepted June 23, 1981.
4210
blood.
When blood lymphocytes or tissues from seropositive donors
are explanted, BEBv-cells grow and can be established as
permanent lines. Limiting dilution experiments estimate that
there are 500 to 5000 B-cells with proliferative potential in the
blood (7). In acute IM, the number is 10" times higher and up
to 18% of the B-cells are EBNA positive (4, 17, 35). Moreover,
in fatal IM cases, EBNA-positive cells were detected even in
the spleen, liver, and thymus (2).
We have only limited knowledge about the dynamics of the
BEBv-cell population in vivo. Recruitment, proliferation, differ
entiation, and elimination are probably the events which deter
mine the production and persistence of these cells.
CANCER
RESEARCH
VOL. 41
Downloaded from cancerres.aacrjournals.org on June 18, 2017. © 1981 American Association for Cancer Research.
Control of BEBV-Cells
Possible Mechanisms
Stable Level in Vivo
for Maintaining
the BEBv-Cells at a
The EBV-specific immune responses have been studied ex
tensively. Antibodies directed against cell surface antigens
have been demonstrated by various methods (6, 15, 16, 39).
EBV-specific T-cell-mediated growth inhibition and cytotoxicity have been demonstrated by Moss ef al. (24), ThorleyLawson ef al. (47), and Rickinson et al. (34). In their assay,
autologous T-cells suppressed the outgrowth of BEBv-cells in
the in vitro transformation system. Subsequently, Misko ef al.
(23) demonstrated T-cell-mediated
recognition in cytotoxic
tests. These phenomena are discussed elsewhere in this issue.
The LMI assay was also used for detection of cellular memory
to EBV-determined antigens (42). Healthy seropositive individ
uals reacted to antigen extracts from EBV genome-carrying
cells and to partially purified EBNA, while seronegatives did
not. Mock EBNA had no such effect (41). In addition, mem
brane preparations from EBV-positive non-virus-producing cell
lines induced LIF production in lymphocytes of seropositive
individuals. Membranes from EBV-negative lines did not induce
LIF (43). The LMI response of lymphocytes derived from sero
positive and seronegative donors differed when exposed to 10
fig of partially purified EBNA or 25 jug membrane preparations
per ml. Lower concentrations of the antigens had no effect,
while with higher concentrations nonspecific LMI responses
were found; i.e., even mock preparations caused LMI and this
occurred even with the lymphocytes of seronegative persons
(Table 1). Since lymphocytes readily produce LIF in vitro, the
conditions for any particular assay have to be well controlled,
and evaluation of the results has to take in account the dose
responses to the particular inducing substance.
In addition to EBV-specific immune responses, 2 phenomena
observed in vitro on cell lines may also contribute to the
elimination of BEBv-cells and to counteract the increase of the
virus load.
(a) The BEBv-cells harbor the virus in a latent form. In the
majority of cell lines, only a small proportion of cells enter the
"productive" virus cycle [the first sign of this is the appearance
of the early antigen (EA)]. The cells entering the productive
virus cycle die within 3 to 4 days (8).
(b) The sensitivity of lymphoid lines to natural killer cells was
LMI assay with EBV-seronegative
shown to be increased after superinfection with EBV (P3HR1), which induces the viral cycle. If natural killer cells attack
BeBv-cells in a similar state in vivo, this mechanism would help
to reduce the amount of virus produced because it damages
the cells before virus may be released (28).
T-Cell-mediated
Control of BFBV-Cellsin IM
Studies with lymphocytes of patients during the acute phase
of IM suggested the existence of immunological factors which
control the growth or eliminate the BEBV-lymphocytes. Since
the majority of atypical lymphocytes in the blood of the patients
are T-cells, it was assumed that the T-cell proliferation repre
sents an immune response against viral and/or virally deter
mined antigens (5, 37). The T-cells were found to exert a
cytotoxic effect against lymphoblastoid cell lines, and the sen
sitive panel suggested that recognition of EBV-determined
antigen(s) on the target surface is responsible for the lytic
interaction (40). The experiments were performed with effector
populations from which the Fc receptor-positive
cells were
removed. This was an important measure since a part of the
lymphocytes responsible for the natural nonspecific killer effect
of healthy persons carry easily detectable Fc receptors (1 ). For
testing the nonspecific component of cytotoxicity, the proto
type of natural killer-sensitive cells, K562, was used as the
target. This is a non-B-line and does not express HLA antigens.
It is important to note that the strategy of the experiments and
the evaluation of the results was based on the experience with
freshly isolated blood lymphocytes of healthy individuals. Re
cent experiments indicate, however, that the characteristics of
activated lymphocyte populations are different from those of
freshly harvested unmanipulated lymphocytes. In view of the
accumulating knowledge concerning lymphocyte-mediated cy
totoxicity, it seems that the effects of the IM blood lymphocytes
have to be reevaluated. As will be discussed below, elimination
of Fc receptor-positive cells from an activated population does
not ensure the elimination of nonspecific cytotoxicity, and
depletion of such cells may have a different impact on the
effects against different targets.
Compared to freshly harvested lymphocytes of healthy per
sons, the activated T-cell populations: (a) are cytotoxic for a
Table 1
and -seropositive healthy individuals to partially purified EBNA and plasma
membranes of lymphoblastoid cell lines
% of LMI
Protein concentration
Donorsa+_+_+_+AntigenMock
EBNA0Mock
pig/ml8121119000810
fig/ml916153554111925
fig/ml20272559713153750
jig/ml3635436313191941100
/ig/mlND"NDNOND18252151
EBNA"EBNACEBNACRamos
membraneRamos
membraneRaji
membraneRaji
membrane2.5ng/ml08a1300008
a -, EBV seronegative; + , EBV seropositive
0 Prepared from Ramos.
c Prepared from Raji.
d ND, not done.
NOVEMBER
1981
Downloaded from cancerres.aacrjournals.org on June 18, 2017. © 1981 American Association for Cancer Research.
4211
E. Klein et al.
Table 2
Characterization
of blood lymphocytes of one donor previous to and at the time
of the acute phase of IM
Reactivity with the various monoclonal reagents was tested simultaneously in
indirect immunofluorescence on frozen preserved samples (in collaboration with
J. Seeley and E. Svedmyr).
Days in relation t
the clinical symp
toms-32-20
0DOKT3a55
cellsOKT818
of positive
78
89%
2880OKT42831
224OKU1050
6OKM114
OKT3, OKT8. OKT4 react with various categories of T-cells, OKM1 with
monocytes, granulocytes and a subset of lymphocytes, and OKU defines the la
antigen. The monoclonal reagents were supplied by P. Kung and G. Goldstein
(Ortho Pharmaceutical Corp.. Raritan, N. J.).
broader target panel (20); (b) express fewer Fey receptors (29,
36); and (c) have a lower proportion of cells defined with the
OKM1 monoclonal reagent.7 In these respects, the activated
lymphocytes are thus similar to the atypical populations in the
acute phase of IM (10).
We have followed one EBV-seronegative individual known to
have been in contact with an IM patient just preceding the
outbreak of clinical symptoms.8 The proportion of cells which
reacted with the reagents which characterize mature T-cells in
the blood (OKT3) and with OKT8 which reacts with thymocytes
and part of the T-cells (suppressor T) increased while the
OKT4-positive (helper T) and OKM1-positive cells decreased
(Table 2). The increase of cells with the suppressor phenotype
that we observed corroborates the results of functional studies
of these cells (48). In accordance with the presence of activated
T-cells, the proportion of la-positive cells increased (44). The
lytic effect of lymphocytes on K562 cells decreased. This
correlates well with the low level of OKM1-positive cells since
in the blood of healthy donors this population is cytotoxic for
K562 (52).
We have shown previously that the natural cytotoxic function
of lymphocyte subsets differs depending on the target cells
used (21). The effect of subsets rich in OKM1-positive cells
acted strongly on K562 but not on Daudi. The OKM1-positive
cells are found in the subsets isolated on the basis of Fc-y
receptor expression.
These results prompted us to compare the cytotoxicities of
lymphocytes activated by various measures against a panel of
cell lines including the natural killer-sensitive prototype K562,
EBNA-positive and -negative B-lines, the EBV-negative Burkitt
line BJAB, and the in wiro-infected EBV-positive subline BJABB95-8 (Table 3). This cell line panel was killed by lymphocytes
from patients with IM, as well as by the various activated
populations. Comparison with the fresh effectors shows that
activation enhanced the cytotoxic activity. Since different do
nors were used in the experiments (except for the series with
untreated and interferon pretreated effectors), the significance
of small differences between the various activated populations
cannot be judged. In general, cells activated in vitro by various
measures or in vivo during the acute phase of IM show en
hanced killing and no apparent EBV relatedness. The activation
achieved in vitro was strongest when the lymphocytes were
cultured in T-cell growth factor. Brief treatment with interferon
potentiated the cytotoxicity. Noteworthy was the finding that
' M. G. Masucci and E. Klein, to be published
* E. Svedmyr, I. Ernberg. G. Masucci. R. Szigeti, G. M Masucci. J. Seeley. E.
Klein. G. Klein, and O. Weiland, to be published.
4212
the EBV-negative
line U698 was killed by both the in vitro-
activated and the cells from patients with IM.
Comparison of the results with 2 BJAB lines shows that the
EBV-positive BJAB subline is less sensitive to the lytic effects.
This difference may be related to the alteration of the plasma
membrane properties by the virus. Membrane IgM and concanavalin A receptor redistribution was found to be reduced when
the EBV-negative lymphoma lines (BJAB and Ramos) were
converted to the genome-positive state by in vitro infection
(51).
These results support our hypothesis (14, 21) that the lytic
function of the T-lymphocytes in the acute phase of IM is not
determined by the recognition of EBV-related antigen on the
target but is rather a consequence of their activation. Cytotoxic
function of lymphocyte populations exerted against certain cell
lines is a measure of their activation.
As proposed earlier (19), it is likely that the recognition of
cell surface antigens can be detected more regularly by looking
for the signs for activation of the lymphocyte population than
for lytic effects on the specific target.
The appearance of activated T-cells in the blood of patients
with IM may be the consequence of the host response against
the antigens of EBV or antigens induced by EBV in B-cells. TCells respond with blastogenesis when exposed to UV-irradiated EBV (45) or to EBV absorbed on the surface of autologous
B-cells (9). In experiments published previously (49), T-cells
from healthy donors were confronted with autologous B-cells
infected with EBV. The B-cells were taken from cultures kept
for various times up to 13 days; thus, the B-cell cultures were
in various stages of transformation. The blastogenic response
of T-cells elicited by these B-cells increased with time which
elapsed after their infection. In parallel with the blast transfor
mation, the lymphocytes acquired cytotoxic potential which
damaged the EBV-negative K562 and 2 EBV-positive B-lines
(Chart 1). Thus, the effectors did not act specifically against
EBV-related surface antigens. However, the trigger for activa
tion was provided by the EBV-infected B-cells.
Since it is known that T-cells are stimulated in vitro by
autologous B-cells and that this reaction has the classical
attributes of an immune phenomenon, i.e., memory and
specificity (50), it is important to distinguish the activation event
due to the EBV-induced transformation state of B-cells from
the possibility that B-blasts irrespective of how they were
induced activate the T-cells.
The results of the experiment shown in Table 4 reveal that Tcells are stimulated in vitro by autologous B-cells and to a
higher extent if the B-cells have been exposed to mitogen or
EBV. These events also occurred with cells of the EBV-sero
negative donor, although the stimulation was slightly weaker.
This indicates that the T-cell stimulation by B-blasts is unrelated
to EBV-determined antigens.
The response of T-cells of seronegative donors to BEBv-cells
was also shown by enumerating the sheep RBC-rosetting cells
which attach to surface immunoglobulin-positive
B-cells in lym
phocyte cultures infected with EBV. Recognition of the B-cells
seemed to occur because the proportion of attaching T-cells
increased during the first 2-3 days. In 6 experiments in which
seropositive and seronegative donors were tested concomitantly, the samples of the positive donors had only slightly
higher proportions, their number varying between 20 and 50%,
of the T-cells included in the B-cell groups. Comparing the
CANCER
RESEARCH
VOL. 41
Downloaded from cancerres.aacrjournals.org on June 18, 2017. © 1981 American Association for Cancer Research.
Control of BEBV-Cells
Cytotoxic effect of activated lymphocytes
Table 3
towards a panel of EBV genome-positive and -negative lines and comparison
the acute phase of IM
with the cytotoxicity
of lymphocytes
from
release*U698ÕEBV)
of specific 5'Cr
)"Healthy
(EBV*)n544
K562 (EBV
)Mean13
25-9029-78 3433 15-5417-52
544%324
536167Range19-90
54
Patientsn5
31-80n5
4Mean23
58Range10-45
4Mean43 58-78Daudi
(interferoni
One-way mixed
lymphocyte cul
ture
T-cell growth
factor"
IMe
52
29-65
4
52
34-70
2
51
14-75
5
53
34
Kaplan (EBV)
BJAB (EBV
Range0-40
n Mean Range n Mean Range n Mean
2
0-5
5
17
4-25 6
17
10-29
17-57
19-270-810-30BJAB-B95-8
2
10-10 4
16
0-40 2
<fresh)c
Healthy
(Interferon)0
Patients (fresh)0
IM 241 (EBV)
(EBV*)n
Range4
Mean
9
0-19
21Range
29-75
5
20
12-41
5
15
0-42
2
60-68
3
40
35-47
3
14
5-22
3
28
26-30
20-55
7
28
14-40
5
28
8
54
22-70
a The percentage of specific 5'Cr release at 25:1 effectortarget
20-40
1-9
3
60
44-72
3
15
10-18
30
17-47
3
31
30-32
ratio was tested as described previously in a 4-hr assay (40). Except for the interferon-activated
effectors which were always compared to untreated lymphocytes from the same donor, the various experiments were performed with different donors.
b EBV , EBV genome negative; EBV , EBV genome positive.
0 Freshly separated macrophage-depleted
lymphocytes from healthy donors and Hodgkin's disease and lymphoma patients with high anti-VGA titer (>5120)
without and with interferon pretreatment.
d Seven-day culture with T-cell growth factor-containing supernatants from phytohemagglutinin-stimulated
lymphocytes.
e Lymphocytes from IM patients were obtained during the acute phase of the disease. The diagnosis was done on the basis of the clinical picture and serological
data.
100.
2 80.
<
u I6
Table 4
Stimulation of T-cells by autologous B-cells
I4
Purified B-cells were left untreated, treated with protein A (10 /ig/ml), or
infected with EBV (5 x 105 infectious units/ml). After 2 days, the cells were
ni
,12 o
washed and treated with 30 ¡igof mitomycin C per ml for 30 min. Subsequently,
the B-cells were incubated with freshly separated autologous T-cells in microwells. 105 B-cells with 105 T-cells. On Day 6, 1 /iCi [3HJthymidine per well was
-IO u
1C
added, and after 18 hr of incubation the incorporation was determined.
(cpm)Cultures
[3H]Thymidine incorporation
z
tal
_J
UJ
a- 60
8 o!
_o
Ë-40J
e
u
2
O
.4
J
X
"
I- 2 i—i
20
a.
m
a«
Z
4
DAYS
6
AFTER
8
EBV
IO
I2
INFECTION
Chart 1. Autologous lymphocyte-BEBv-cell stimulation system. BEBv-lymphocytes cultured for various times after EBV infection were irradiated (3000 R) and
used to stimulate autologous nylon-passed T-enriched lymphocytes. After 6 days,
the different cultures were tested for [3H]thymidine uptake and for cytotoxicity
against a panel of EBV genome-positive and -negative lines. Stimulation index
(X) was calculated as:
mean of counts/min
mean of counts/min
in stimulated cultures
in cultures of effectors alone
The means were obtained from triplicated cultures. Cytotoxic activity against
K562 (EBV genome negative) (A), Kaplan (EBV genome positive) (O), and S118
(EBV genome positive) (O) at a 45:1 effectortarget
ratio is shown. The curves
are based on results published previously.
pairs in each experiment, this proportion was 1.5 times higher
in the seropositives. The sheep RBC-rosetting T-cells were
separated after dispersion from the clumps and mixed with
autologous lymphoblastoid cell lines established by transfor
mation with EBV. The proportion of conjugate-forming T-cells
was 20 to 30%, but there was no difference between sero
positive and seronegative donors.
The experiments of Thorley-Lawson (46) showed prompt
recognition of Beav-cells by autologous T-cells. Their prolifer-
donors1,150
donors600
±120s
alone
903,050±
T-Cells with B-cells cultured
23010.650
4.900 ±
2006.500
±
for 2 days
T-Cells with B-cells exposed
±370
±380
to protein A for 2 days
T-Cells
520" with B-cells exposed
12,250 ±540EBV-seronegative
1 1.050 ±
to EBV for 2 daysEBV-seropositive
Mean ±S.E. of triplicate determinations.
testedT-Cells
ation was suppressed if they were exposed to the T-cells for 3
hr immediately after virus infection. This suppressive effect was
as efficient as after 48 hr of contact.
During the first week of IM, the EBV-induced polyclonal Bcell activation is reflected by hypergammaglobulinemia.
During
the second week of the disease, T-cells were found to suppress
B-cell activation, as measured by immunoglobulin production
in vitro (48). The effector part of this interaction was not specific
for the EBV-induced B-cell activation, since the same T-population suppressed also the pokeweed mitogen-induced
re
sponse of autologous and allogeneic B-cells. At present, it is
difficult to detect an EBV-specific component in the action of
the lymphocytes from patients with IM. It may not be present
since the evolution of the EBV-specific T-cell memory during
the disease develops slowly compared to most of the antibody
responses to the EBV-determined antigens (45).
For the clinical course of the disease, the important factor is
the activated state of T-cells. Activated T-cells have also been
shown to be cytotoxic to autologous lymphoblastoid cell lines
established by transformation with EBV (Table 5). They may
NOVEMBER 1981
Downloaded from cancerres.aacrjournals.org on June 18, 2017. © 1981 American Association for Cancer Research.
4213
E. Klein et al.
Table 5
Killing of autologous lymphoblastoid cell lines established by transformation
with EBV by activated T-ce/ls
T-Cells were first cultured in the presence of autologous B>,,.,-colls and then
after 5 days transferred to medium containing 25% human T-cell growth factor.
These cells were tested for cytotoxicity against a panel of target cells after 14
days of culture (in addition to the targets shown in the table. K562, Daudi, and
one other lymphoblastoid cell line established by transformation with EBV were
also killed).
% of specific 51Cr release
NPC-45"Effector
1"30
donorsNPC-45YTW-5725:
cell
Ie9±
±18:114
" Target lines.
6 Effectontarget
c Mean ±S.E.
13±
223 ±
±2YTW-5725:147
±18:123
18±
±1
ratios.
therefore be instrumental in the elimination or control of the
BEBv-cells. However, in certain individuals such as males with
X-linked lymphoproliferative syndrome, the autoreactivity may
lead to complications and long-lasting or fatal phenotypes (30).
Given the complexity of the immune system with interacting
T- and B-cells, the consequences of EBV infection of B-cells
are intriguing. While the virus is a potent activator and mitogen
for the B-subset, the T-subset responds to changes which
occur in the B-subset. The phenomenon of "autostimulation"
is an in vitro corollary of this (50). The events in IM may
represent an amplified picture of the interaction between Tand B-lymphocytes in the immune response. It thus follows that
in addition to the specific immunity to EBV which develops
during the disease, the immunoregulatory mechanisms also act
to inhibit B-cell proliferation.
References
1. Bakács. T.. Gergely. P., and Klein, E. Characterization of cytotoxic human
lymphocyte subpopulations. The role of Fc receptor carrying cells. Cell.
Immunol., 32: 317-328. 1977.
2. Britton, S.. Andersson-Anvret. M.. Gergely. P.. Henle, W.. Jondal, M., Klein.
G., Sandstedt, B.. and Svedmyr, E. EB virus immunity and tissue distribution
in a fatal case of infectious mononucleosis. N. Engl. J. Med., 298. 89-92.
1978.
3. Chang. R. S., Lewis. J. P., and Abildgaard. L. F. Prevalence of oropharyngeal
excretors of leukocyte transforming agents among a human population. N.
Engl. J. Med.. 289: 1325-1329, 1973.
4. Diehl. V., Henle, G.. Henle, W., and Kohn, G. Demonstration of a herpes
group virus in cultures of peripheral leukocytes from patients with infectious
mononucleosis. J. Virol., 2. 663-669, 1968.
5. Enberg, R. N.. Eberle, B. J., and Williams, C. H. T- and B-cells in peripheral
blood during infectious mononucleosis. J. Infect. Dis., 130: 104-111, 1971.
6. Ernberg, I., Klein. G., Kourilsky, F. M., and Silvestre, D. Differentiation
between early and late membrane antigen on human lymphoblastoid cell
lines infected with Epstein-Barr virus. I. Immunofluorescence. J. Nati. Cancer
Inst, 53. 61-65. 1974.
7. Gergely, L.. Czeglédy, J., Vaczi, L., Szalka, A., and Berényi, E. Cells
containing Epstein-Barr nuclear antigen (EBNA) in peripheral blood. Acta
Microbiol. Acad. Sci. Hung., 26 41-45, 1979.
8. Gergely, L., Klein. G.. and Ernberg, I. Effect of EBV-induced early antigens
on host cell macromolecular synthesis, studied by combined immunofluorescence and radioautography. Virology, 45: 22-29, 1971.
9. Gergely, P.. Ernberg, I., Klein, G., and Steinitz. M. Blastogenic responses of
purified human T-lymphocyte populations to Epstein-Barr virus (EBV). Clin.
Exp. Immunol., 30. 347-353. 1977.
10. Haynes. B. T., Schooley. R. T.. Grouse. J. E.. Payling-Wright, C. R., Dolin,
R., and Fauci, A. S. Characterization of thymus-derived lymphocyte subsets
in acute Epstein-Barr virus-induced infectious mononucleosis. J. Immunol.,
/22. 699-702, 1979.
11. Henle, G., and Henle, W. Immunofluorescence. interference and complement
fixation techniques in the detection of the herpes-type virus in Burkitt tumor
cell lines. Cancer Res.. 27 2442-2446,
1967.
4214
12. Henle, G., and Henle, W. Observations on childhood infections with the
Epstein-Barr virus. J. Infect. Dis., 121: 303-310, 1970.
13. Hinuma, Y., Konn, M., Yamaguchi. J., Wudarski, D. J., Blakesler, J. R., Jr.,
and Grace. J. T., Jr. Immunofluorescence and herpes-type virus particles in
theP3HR-1 Burkitt lymphoma cell line. J. Virol., /: 1045-1051.
1967.
14. Klein, E., Masucci, M. G., Berthold, W., and Blazar, B. A. Lymphocytemediated cytotoxicity toward virus-induced tumor cells: natural and activated
killer lymphocytes in man. Cold Spring Harbor Conf. Cell Proliferation. 7:
1187-1197,
1980.
15. Klein, G., Clifford, P., Klein, E., and Stjernswärd. J. Search for tumor specific
immune reactions in Burkitt lymphoma patients by the membrane immunofluorescence reaction. Proc. Nati. Acad. Sei. U. S. A., 55: 1628-1635,
1966.
16. Klein, G., Gergely, L., and Goldstein. G. Two colour immunofluorescence
studies on EBV determined antigens. Clin. Exp. Immunol., 8. 593-602,
1971.
17. Klein, G.. Svedmyr. E.. Jondal, M., and Persson, P. O. EBV determined
nuclear antigen (EBNA) positive cells in the peripheral blood of infectious
mononucleosis patients. Int. J. Cancer, 17: 21-26, 1976.
18. Lemon, S. M., Hutt, L. M.. Shaw, J. E.. Li, J. L., and Pagano. J. S. Replication
of EBV in epithelial cells during infectious mononucleosis. Nature (Lond.).
268. 268-270, 1977.
19. Martin Chandon, M. R., Vanky. F.. Carnaud, C.. and Klein, E. In vitro
education of autologous human sarcomas generates non specific killer cells.
Int. J. Cancer, 15: 342-350, 1975.
20. Masucci, M. G., Klein. E., and Argov, S. Disappearance of the NK effect
after explantation of lymphocytes correlated to the level of blastogenesis in
activated cultures. J. Immunol.. 124: 2458-2463,
1980.
21. Masucci, M. G., Masucci, G., Klein, E., and Berthold, W. Target selectivity
of interferon-induced human killer lymphocyte related to their Fc receptor
expression. Proc. Nati. Acad. Sei. U. S. A., 77: 3620-3624,
1980.
22. Miller. G.. Niederman, J. C.. and Andrews. L. Prolonged oropharyngeal
excretion of EB virus following infectious mononucleosis. N. Engl. J. Med.,
»37:140-147, 1973.
23. Misko, I. S., Moss, D. J., and Pope, J. H. HLA-antigen-related restriction of
T-lymphocyte cytotoxicity to Epstein-Barr virus. Proc. Nati. Acad. Sei. U. S.
A., 77. 1247-1250.
1980.
24. Moss, D. J., Scott. W., and Pope, J. H. An immunological basis for inhibition
of transformation of human lymphocytes by EB virus. Nature (Lond.), 268:
735-736. 1977.
25. Niederman, J. C., Miller. G.. Pearson, H. A., Pagano, J. G.. and Dowalibi, J.
M. Infectious mononucleosis. Epstein-Barr virus shedding in the saliva and
the oropharynx. N. Engl. J. Med., 294: 1355-1359,
1976.
26 Nilsson. K. The nature of lymphoid cell lines and their relationship to the
virus. In: M. A. Epstein and B. G. Achong (eds.). The Epstein-Barr Virus, pp.
225-281. New York: Springer Verlag, 1979.
27. Nilsson. K.. Giovanella, B. C., Stehlin, J. S.. and Klein. G. Tumorigenicity of
human hematopoietic cell lines in athymic nude mice. Int. J. Cancer, Õ9:
337-344, 1977.
28. Patarroyo, M., Blazar, B., Pearson, G., Klein, E., and Klein, G. Induction of
the EBV cycle in B-lymphocyte-derived
lines is accompanied by increased
natural killer (NK) sensitivity and the expression of EBV-related antigen(s)
deleted by the ADCC reaction. Int. J. Cancer, 26: 365-371, 1980.
29. Poros, A., and Klein, E. Cultivation with K562 cells leads to blastogenesis
and increased cytotoxicity with changed properties of the active cells when
compared to fresh lymphocytes. Cell. Immunol., 41: 240-255. 1978.
30. Purtllo, D. T., De Floria. D., Hutt. L. M., Bhawan. J.. Yang, J. P. S., Otto, R.,
and Edwards, W. Variable phenotypic expression of an X-linked recessive
lymphoproliferative syndrome. N. Engl. J. Med.. 297: 1077-1081,
1977.
31. Rickinson, A. B., Epstein, M. A., and Crawford, D. H. Absence of EpsteinBarr virus in blood in acute infectious mononucleosis. Nature (Lond.), 258:
236-238. 1975.
32. Rickinson, A. B., Finerty, S.. and Epstein. M. A. Comparative studies on
adult donor lymphocytes infected by EB-virus in vivo or in vitro: origin of
transformed cells arising in cocultures with foetal lymphocytes. Int. J.
Cancer, 19: 775-782, 1977.
33. Rickinson, A. B., Finerty, S., and Epstein, M. A. Mechanism of the establish
ment of Epstein-Barr virus genome containing lymphoid cell lines from
infectious mononucleosis patients: studies with phosphonoacetate.
Int. J.
Cancer, 20: 861-868. 1977.
34. Rickinson, A. B., Moss, D. J., and Pope. J. H. Long-term T-cell mediated
immunity to Epstein-Barr virus in man. II. Components necessary for regres
sion in virus-infected leucocyte cultures. Int. J. Cancer, 23:610-617,1979.
35. Robinson, J., Smith, D., and Niederman, J. Mitotic EBNA-positive lympho
cytes in peripheral blood during infectious mononucleosis. Nature (Lond.),
287:334-335,
1980.
36. Seeley, J. K., Masucci. G.. Poros. A.. Klein. E.. and Golub, S. H. Studies on
cytotoxicity generated in human mixed lymphocyte cultures. J. Immunol.,
Õ23. 1303-1311,
1979.
37. Sheldon, P. J., Hemsted, E. H., Papamichail, M., and Holborrow. E. J.
Thymic origin of atypical lymphoid cells in infectious mononucleosis. Lancet,
1: 1153-1155, 1973.
38. Svedmyr, A., and Demissie, A. Age distribution of antibodies to Burkitt cells.
CANCER
RESEARCH
VOL. 41
Downloaded from cancerres.aacrjournals.org on June 18, 2017. © 1981 American Association for Cancer Research.
Control of BEBV-Cells
Acta Pathol. Microbio!. Scand., 73: 653-654, 1968.
39. Svedmyr, A., Demissie, A., Klein, G.. and Clifford, P. Antibody patterns in
different human sera against complexes associated with Epstein-Barr virus.
J. Nati. Cancer Inst., 44: 595-610, 1970.
40. Svedmyr, E.. and Jondal, M. Cytotoxic effector cells specific for B-cell lines
transformed by Epstein-Barr virus are present in patients with infectious
mononucleosis. Proc. Nati. Acad. Sei. U. S. A., 72: 1622-1626,
1975.
41. Szigeti, R., Luka, J., and Klein, G. Leukocyte migration inhibition studies
with Epstein-Barr virus (EBV) determined nuclear antigen (EBNA) in relation
to the EBV-carrier status of the donor. Cell. Immunol., 53. 269-276, 1981.
42. Szigeti, R., Timar. L., and Révész.
T. Leukocyte migration inhibition with
Epstein-Barr virus negative and positive cell extracts. Allergy, 35: 97-103,
1980.
43. Szigeti, R., Volsky, D. J., Luka, J., and Klein, G. Membranes of EBV-carrying
virus nonproducer cells inhibit leukocyte migration of EBV-seropositive but
not seronegative donors. J. Immunol., Õ26: 1676-1679.
1981.
44. Tatsumi, E., Kimura, K., Takinchi. Y.. Fukuhara, S., Sturakawa, S., Uchino,
H., Morikawa, S., and Minowada, J. T lymphocytes expressing human lalike antigens in infectious mononucleosis (IM). Blood. 56: 383-387, 1980.
45. Ten Napel, C. H. H., and The, T. H. Lymphocyte reactivity in infectious
mononucleosis. J. Infect. Dis., 141: 716-7123,
1980.
NOVEMBER
46. Thorley-Lawson, D. A. The suppression of Epstein-Barr virus infection in
vitro occurs after infection but before transformation of the cells. J. Immunol.,
724: 745-751, 1980.
47. Thorley-Lawson, D. A.. Chess, L.. and Strominger, J. Suppression of in vitro
Epstein-Barr virus infection. J. Exp. Med.. 146: 495-508, 1977.
48. Tosato, G., Magrath, I., Koski, I., Dooley, M.. and Blease, M. Activation of
suppressor T cells during Epstein-Barr virus-induced infections mononucle
osis. N. Engl. J. Med., 307. 1133-1137,
1979.
49. Viallat, J., Svedmyr, E.. Yefenof, E., Klein, G., and Weiland, O. Stimulation
of human peripheral blood lymphocytes by autologous EBV-infected B cells.
Cell. Immunol.. 41: 1-8. 1978.
50. Weksler, M. E., and Kozak. R. Lymphocyte transformation
induced by
autologous cells. V. Generation of immunologie memory and specificity
during the autologous mixed lymphocyte reaction. J. Exp. Med.. 746:18331838, 1977.
51. Yefenof, E., and Klein, G. Difference in antibody induced redistribution of
membrane IgM in EBV genome free and EBV positive human lymphoid cells.
Exp. Cell Res., 99: 175-178, 1976.
52. Zarling, I. M., and King, P. C. Monoclonal antibodies which distinguish
between human NK cells and cytotoxic T lymphocytes. Nature (Lond.), 288:
394-396, 1980.
1981
Downloaded from cancerres.aacrjournals.org on June 18, 2017. © 1981 American Association for Cancer Research.
4215
T-Cell Response to B-Cells and Epstein-Barr Virus Antigens in
Infectious Mononucleosis
E. Klein, I. Ernberg, M. G. Masucci, et al.
Cancer Res 1981;41:4210-4215.
Updated version
E-mail alerts
Reprints and
Subscriptions
Permissions
Access the most recent version of this article at:
http://cancerres.aacrjournals.org/content/41/11_Part_1/4210
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 June 18, 2017. © 1981 American Association for Cancer Research.