आऋऑऎऊࣽईࣜऋंࣜ उँऀअࣿࣽईࣜ ࣿऋईईँःँएࣜऋंࣜऌईࣽ Journal of Medical Colleges of PLA 24 (2009) 249–258
www.elsevier.com/locate/jmcpla
ICOS-Ig combined with CsA induces long term survival of cardiac allografts in mouse¯ Zhang Peng1, Wang Zhenmeng2, Qin Qin1, Tang Yi3, Wang Quanxing3, Shen Qian1* 1
Department of Clinical Diagnosis, Changhai Hospital, Second Military Medical University, Shanghai, 200433
2
Department of Anesthesiology, East Hepatobiliary Surgery Hospita, Second Military Medical University, Shanghai, 200438 3
Department of Immunology, Second Military Medical University, Shanghai, 200433 Received 9 April 2009; accepted 26 June 2009
Abstract Objective: To study the synergistic effect of ICOS-Ig combined with cyclosporine (CsA) on mouse heart transplantation and explore its therapeutic potential. Methods: ICOS-Ig fusion protein was generated by fusing the extracellular portion of human ICOS and Fc portion of human IgG. To investigate the effect of ICOS-Ig on T-cell proliferation in vitro, ICOS-Ig or IgG was added to the primary MLR cultures (BALB/c spleen T cells as responder cells and irradiated C57BL/6 spleen cells as stimulator cells). The cells responsiveness rates were detected by 3H-TdR methods. Then the T cells of each group in primary MLR were cultured as responder cells for secondary MLR, and irradiated C57BL/6 (donor) or C3H (third party) spleen cells as stimulator cells. To study the effect of ICOS-Ig on T-cell proliferation in vivo, CFSE-labeled C57BL/6 spleen cells were transferred to irradiated BALB/c mice. Mice were then treated with IgG, ICOS-Ig or CsA. Seventy two hours after transfer, the spleen cells of the mice were harvested for the detection of CD4+CFSE+ and CD8+CFSE+ by FACS. C57BL/6 mouse underwent transplantation of the hearts of BALB/c mouse and were then randomly divided into five equal groups: no treatment group, control IgG treated group (250 μg i.p. d2, 4, 6), ICOS-Ig treated group (250 μg i.p. d2, 4, 6), CsA treated group (10 mg/kg i.p. d0-6), ICOS-Ig combined with CsA group. The cardiac allograft survival was monitored by daily palpation. Results: In primary MLR, ICOS-Ig inhibited T-cell proliferation, (inhibition ratio 58±8.2% in 50 ȝg/ml). In secondary MLR, ICOS-Ig specifically inhibited donor spleen cells, which suggested ICOS-Ig could induce donor-specific hyporesponsiveness. In the CFSE dye assay, CD4+CFSE+ and CD8+CFSE+ in ICOS-Ig and CsA group was stronger than those in control group, which showed ICOS-Ig and CsA could inhibit
ƿ
Supported by Research Grants from the Ministry of Science and Technology of People’s Republic of China (National 863 Plan, 2002AA214091) and the National Science Foundation for Young Scholars of China (30500501).
* Corresponding author. Tel: +86 021 81873612; fax: +86 021 81873612; E-mail address:
[email protected] Abbreviations: ICOS, inducible costimulatory molecule; CsA, cyclosporine; MHC, major histocompatibility complex; MLR, mixed lymphocyte reaction; CTL, cytotoxic lymphocyte reaction
250
Zhang Peng et al. / Journal of Medical Colleges of PLA 24 (2009) 249–258
the proliferation of allo-reactive T cells in vivo. In mouse heart transplantation model, survival was significantly prolonged in animals treated with ICOS-Ig or CsA as compared with controls. Moreover, ICOS-Ig combined with CsA group had even longer engraftment (>100 d) than ICOS-Ig or CsA used alone. In histological examination, it was found that there were congestions and edemas in no treatment and IgG treated recipients, together with a lot of inflammatory cells infiltrated. Allogeneic hearts from ICOS-Ig and/or CsA immunized recipients revealed milder histological changes. It was revealed in mechanical analysis that splenic T cells from recipients also exhibited depressed mixed leukocyte reactions (MLR) and cytotoxic lymphocyte reactions (CTL). Conclusion: These data suggest that ICOS-Ig combined with CsA induces a long-term survival of mouse cardiac allografts, whereas monotherapy is less effective in this regard. Thus, ICOS-Ig combined with CsA treatment may be a novel regimen to combat allograft rejection. Keywords: Inducible costimulator-Ig fusion protein; Cyclosporine; Donor-specific hyporesponsiveness; Mouse cardiac transplantation
cyclosporine (CsA) in mouse heart transplantation and
1. Introduction
explored its therapeutic potential. Many studies indicate that the blockage of CD28:B7 and CD40:CD40L costimulatory molecules is
2. Materials and methods
likely to serve as important targets for the prevention of allograft rejection and for the induction of tolerance in
2.1. Animals
animal models [1-3]. Inducible costimulatory (ICOS), with its ligand B7h, is a recently discovered
Male C57BL/6 mice (H-2b), BALB/c mice (H-2d)
costimulatory molecule of the CD28 family [4,5]. Unlike
and C3H (H-2k), 8–12 weeks of age, were purchased
some other costimulatory molecules, which are
from Shanghai Joint Venture SIPPR BK Experimental
expressed on lymphocytes constitutively, ICOS is
Animal Co. The animals were maintained under
induced only after T-cell activation, which can enhance
standard conditions and fed rodent food and water,
T-cell proliferation, cytokine production, and CD154
according to the principle of laboratory animal care and
expression and provides help for Ig production by B
the guide for the care and use of laboratory animals in
cells [6, 7]. With anti-ICOS mAb or an ICOS-Ig fusion
our institution.
-/-
protein, or transfer of grafts ICOS
recipient mice,
prolonged graft survival, although to a lesser degree
2.2. Preparation of ICOS-Ig
than CTLA4-Ig or anti-CD40L mAb therapy, chronic rejection developed with each of these treatments
The soluble form of ICOS (ICOS-Ig) was prepared
[8-11]. These findings suggest that blockade of ICOS
by constructing an mammalian expression vector
signaling can be a new therapeutic target in clinic
containing an extracellular portion of human ICOS
transplantation.
cDNA and Fc portion of human IgG. The extracellular
Because of the low efficacy of conventional
portion of human ICOS cDNA was generated from the
immunosuppressive regimens, and the consequences of
PMA(20 ng/ml)+ionomycin(1 ȝg/ml)-stimulated human
uncontrolled rejection, the costimulatory blockade is
peripheral blood leukocytes, and was amplified with
being studied in clinic as monotherapy. In this study,
primers (sense, 5’-GCGGCCCAGCCGGCCGA AATC
we investigated the effect of ICOS-Ig combined with
AATGGTTCTGCC-3’; antisense, 5’-GCTCTAGACT
Zhang Peng et al. / Journal of Medical Colleges of PLA 24 (2009) 249–258
251
TCAGCTGGCAAAG- 3’) containing Sfi I (sense) and
thymidine/well, harvested and thymidine incorporation
Xba I (antisense) restriction enzyme site sequences. The
determined.
pIGplus plasmid containing Fc portion of human IgG1 was amplified with primers (sense, 5’-TCTAGAGATCC
2.3.2. Secondary mixed leukocyte reaction (2o MLR)
CAAATCTTGTGAC-3’; antisense, 5’- GCGGC CGCT
2×106 BALB/c responder cells were co-cultured in
CATTTACCCGGAGAC AG-3’) containing Xba I
24-well plates with 2×106 irradiated (3000 rad)
(sense) and Not I (antisense) restriction enzyme site
C57BL/6 stimulator cells for 3 d in the presence of
sequences. PCR products were digested with the
ICOS-Ig or IgG1 Fc. Then, the viable cells were
appropriate restriction enzymes and cloned into a
separated from the debris by LSM and restimulated
mammalian expression vector pSecTag2. The plasmid
with the same allogeneic (C57BL/6) cells or third-party
with the correct insert was stably transfected by
(C3H) cells in 96-well round-bottom plates (for seeing
electroporation into CHO cells. The ICOS-Ig protein
primary response). No reagents were added to the
was purified from the supernatant by protein A-
second culture. Secondary response was pulsed 3 d
sepharose
later as primary response.
4FF
(Pharmacia,
Sweden)
affinity
chromatography. Recovered proteins were analyzed by SDS-PAGE in reducing and nonreductig conditions and
2.4. Effect of ICOS-Ig on T-cell proliferation in vivo
Western blotting with HRP-goat anti-human IgG1 Fc antibody (Pierce, USA). For all experiments involving
C57BL/6 spleen T cells purified by nylon wool
fusion proteins, IgG1 Fc in the absence of ICOS portion,
passage were incubated with 10 mmol/L of the tracking
a construct produced by the same cell line as ICOS-Ig,
fluorochrome CFSE (Molecular Probes, USA) [13].
was used as control.
Then, 20×106 CFSE-labeled cells were adoptively transferred to lethally irradiated (1800 rad) BALB/c
2.3. Effect of ICOS-Ig on T-cell proliferation in
recipients. Mice were subsequently allocated into four
vitro [12]
groups: (a) no treatment, (b)control IgG (0.1 mg/d), (c) ICOS-Ig (0.1 mg/d), (d) CsA (0.1 mg/d) (Novartis, o
2.3.1. Primary mixed leukocyte reaction (1 MLR) Spleens
were
major
C57BL/6 control mice also received CFSE-labeled cells.
histocompatibility complex (MHC)-mismatched mice
Three days after cell transfer, mice were sacrificed and
and prepared
by
the splenocytes were harvested and stained with either
density-gradient centrifugation using Ficoll-Hypaque
PE-anti-CD4 or PE-anti-CD8 (eBioscience, USA). By
(Pharmacia, Sweden). MLR were performed in
gating onto CD4+CFSE+ cells and CD8+CFSE+ cells,
round-bottom 96-well plates using 2×105 BALB/c
the proliferation of CD4+ and CD8+ T cells in each
responder cells and 2×105 gamma-irradiated (3000 rad)
separate generation of dividing cells could be
C57BL/6 stimulator cells in a total volume of 0.2 ml.
determined according to the CFSE profiles.
as
obtained
a single
cell
from
Switzerland). A fifth group consisting of syngeneic
suspension
ICOS-Ig or IgG1 Fc were added to the culture wells at the start of the MLR. Cultures were performed in
2.5. Cardiac transplantation procedures
triplicate and incubated at 37°C with 5% CO2 for up to 3 d. Cultures were then pulsed for 18 h with 1 PCi 3H
C57BL/6 mice were used as recipients and
252
Zhang Peng et al. / Journal of Medical Colleges of PLA 24 (2009) 249–258
MHC-mismatched BALB/c mice as donors. Heterotopic
2.8. Histological examinations
cardiac transplantation to a cervical site was performed under a cuff technique as described previously [14].
The cardiac allografts were harvested 7 d after
The function of the heart was monitored daily after
transplantation. All specimens were fixed in 10%
transplantation by cervical palpation. Rejection was
buffered formalin and embedded in paraffin. Five
defined as total cessation of cardiac muscle contraction.
sections were stained with hematoxylin and eosin (HE)
To evaluate the role of ICOS-Ig and CsA in cardiac
for histological examinations.
allograft rejection, recipient mice were assigned to five experimental groups (n=15/group): (a) no treatment, (b)
2.9. Statistical analysis
treated with control IgG (250 Pg, days 0, 2, 4, 6), (c) treated with ICOS- Ig (250 Pg, days 0, 2, 4, 6), (d) CsA
All data were
expressed as mean ± SD
and
tested using the Chi square or Mann-Whitney U test. P
(10 mg/kg/d ×7 d) , and (e) ICOS- Ig and CsA combined.
value of <0.05 was considered significant. Survival was
2.6. Mixed leukocyte reactions (MLR)
evaluated by Kaplan-Meier analysis using the LogRank test.
Recipient C57BL/6 mice were sacrificed 7 d after transplantation, and the spleen T cells purified
3. Results
by nylon wool passage were used as responders. To assess the T-cell hyporesponsiveness of the recipient
3.1. Construction and expression of ICOS-Ig
mice against donor-derived cells, gamma-irradiated (3000 rad) BALB/c cells were used as stimulator
For cloning ICOS cDNA, mRNA was prepared
cells. After being cultured at 37°C for 3 d, cells were
from PMA + ionomycin-activated human peripheral
pulsed for 18 h with 1 PCi
3
H thymidine/well,
blood leukocytes. The extracellular domain (EC) of
harvested and thymidine incorporation made the
ICOS was amplified so that it terminated after Lys140,
determination.
just upstream of two cysteines near the junction with the transmembrane domain. ICOS EC domain with the
2.7. Cytotoxic lymphocyte reactions (CTL)
correct sequence was subcloned upstream of a human IgG1 Fc domain inserted into the mammalian
Cytotoxic responses were assayed by the JAM 6
expression vector pSecTag2 (Fig. 1A). Plasmid DNA
Test as previously described [15]. 5 × 10 spleen T cells
was stably transfected to CHO cells. The ICOS-Ig
from
were
protein was purified from the cell culture supernatants
stimulated for 5 d with 1×10 irradiated allogeneic
by protein A affinity chromatography. Similar to the
BALB/c spleen cells. Con A blast targets were set up
natural monovalent dimmer, ICOS-Ig seemed to be a
6
40 h prior to the CTL assay by culturing 1.5 × 10
dimeric protein about 100 kDa in molecular size, as
syngeneic and BALB/c spleen cells with ConA (1.25
judged by SDS-PAGE and Western blotting in reducing
recipient
C57BL/6
mice
(effectors)
6
3
Pg/ml), then labeling with H thymidine. Lysis of target cells was tested at various effector to target ratios.
versus nonreducing conditions (Fig. 1B and 1C).
Zhang Peng et al. / Journal of Medical Colleges of PLA 24 (2009) 249–258
253
Fig. 1. A. Structure of the ICOS-Ig fusion protein. The cDNA fragment encoding the ICOS-Ig fusion protein is diagrammed. The extracellular domain of ICOS was fused immediately
Fig. 2. A. ICOS-Ig inhibits proliferation of one-way 1° MLR.
after Lys140 to the human IgG1 Fc domain. The restriction
BALB/c spleen cells were isolated as responder cells, and
sites incorporated into the fusion gene cassette are also
gamma-irradiated C57BL/6 cells as stimulator cells. Different
indicated. B. SDS-PAGE analysis of ICOS-Ig fusion proteins
dosages of ICOS-Ig were added to primary MLR cultures
in reducing versus nonreducing conditions. Similar to the
(termed ICOS-Ig group)ēand in control group(no treatment or
natural monovalent dimmer, ICOS-Ig seems to be a dimeric
IgG). Allogeneic MLRs of cells treated with graded doses of
protein about 100 kDa in molecular size. C. Western blot analysis
ICOS-Ig or control IgG. Proliferative T cell responses can be
of ICOS-Ig fusion protein under reducing condition.
significantly inhibited by ICOS-Ig compared with control IgG. B. ICOS-Ig induces hyporesponsiveness to alloantigen
3.2. ICOS-Ig inhibits alloreactive T cell activity in
restimulation. 1° MLR cultures were incubated for 3 d in the
vitro
presence in the presence of ICOS-Ig or IgG. Then, the viable cells were separated from the debris by LSM and restimulated
To study the potential role of ICOS as a
with the same donor (C57BL/6) cells or third-party (C3H)
costimulatory molecule, we performed allogeneic
cells. 3H thymidine incorporation was determined at d3. Each
MLRs in the presence of ICOS-Ig and control IgG. To
group restimulated with third-party cells showed the maximal
monitor primary MLR, BALB/c spleen cells were
responses. IgG group restimulated with donor cells showed
isolated as responder cells, and gamma-irradiated
similar responses.The proliferation in ICOS-Ig group with
C57BL/6 cells as stimulator cells. Different dosages of
donor spleen cells was specifically inhibited in comparison
ICOS-Ig were added to primary MLR cultures (termed
with that in other groups. Error bars represent SD.
ICOS-Ig group)ēand in control group(no treatment or IgG). Incubated for 3 dēthe cells responsiveness rates
primary MLR were cultured as responder cells for
were detected by 3H thymidine methods. ICOS-Ig
secondary MLR, and irradiated C57BL/6 (donor) or
significantly inhibited T-cell proliferation, which
C3H (third-party) spleen cells as stimulator cells. The
dependedon the dosage of ICOS-Ig (inhibition ratio
proliferation of ICOS-Ig group restimulated with donor
~60% in 50 ȝg/ml) (Fig. 2A). T cells of each group in
spleen cells was specifically inhibited compared with
254
Zhang Peng et al. / Journal of Medical Colleges of PLA 24 (2009) 249–258
that restimulated with third-party spleen cells, which
combined. Mice were sacrificed 3 d after cell transfer,
suggested
donor-specific
and the splenocytes were harvested and analysed by
hyporesponsiveness, that was to say, induce donor-
FACS. The results showed that CD4+ and CD8+
specific tolerance (Fig. 2B).
C57BL/6 T cells in untreated mice demonstrated strong
ICOS-Ig
could
induce
proliferative responses to BALB/c hosts. In contrast,
3.3. ICOS-Ig and/or CsA blocks allogeneic CD4+
cells transferred into syngeneic hosts underwent
and CD8+ T cell proliferation in vivo
negligible proliferation. Compared with untreated recipients, mice treated with ICOS-Ig or CsA alone had
An in vivo assay was also set up to evaluate the
a significant reduction in allo-reactive CD4+ and CD8+
impact of ICOS-Ig on allogeneic T cell proliferation.
T-cell proliferation, respectively. The most striking
CFSE-labeling C57BL/6 spleen T cells were adoptively
decay in response is illustrated in mice treated with
transferred to lethally irradiated BALB/c recipients,
ICOS-Ig and CsA combined(Fig. 3).
which were then treated by ICOS-Ig, CsA or the two
Fig. 3. ICOS-Ig and/or CsA blocks allogeneic CD4+ and CD8+ T cell proliferation in vivo. C57BL/6 spleen T cells were incubated with the tracking fluorochrome CFSE and adoptively transferred to lethally irradiated C57BL/6 (A) or BALB/c (B) recipients. Mice were subsequently allocated into five groups: (a) no treatment, (b)control IgG, (c) ICOS-Ig, (d) CsA, (e)syngeneic C57BL/6 control. 3 d after cell transfer, mice were sacrificed and the splenocytes were harvested and stained with either PE-anti-CD4 or PE-anti-CD8. By gating onto CD4+CFSE+ cells and CD8+CFSE+ cells, the proliferation of CD4+ and CD8+ T cells in each separate generation of divided cells could be determined according to the CFSE profiles.
255
Zhang Peng et al. / Journal of Medical Colleges of PLA 24 (2009) 249–258
Fig. 4. A. Survival graph of cardiac allografts. Treatment with ICOS-Ig (29.5r7.7 d), CsA (21±4.5 d) significantly prolongs cardiac allograft survival as compared with untreated mice (8.5±1 d) (P<0.05). ICOS-Ig combined with CsA group induces even longer engraftment (>100 d) than ICOS-Ig or CsA used alone(P<0.01). B. Histology of murine cardiac allografts harvested 7 d after transplantation. Rejected grafts from untreated and control IgG recipients demonstrates diffuse inflammatory cell infiltration, myocyte destruction, and interstitial hemorrhage, whereas scattered inflammatory cells and relatively preserved myocytes are seen in the grafts treated with ICOS-Ig or CsA. Moreover, in ICOS-Ig combined with CsA treated group little inflammatory infiltrate are seen, and myocytes undamaged. Magnification, 400.
3.4. ICOS-Ig and/or CsA prolonged allograft
induced even longer engraftment (>100 d) than
survival
ICOS-Ig and CsA alone (P<0.01). (Fig. 4A). Histologic examination at 7 d after transplantation revealed that
Treatment with ICOS-Ig significantly prolonged
acute
rejection
was
significantly
suppressed
in
cardiac allograft survival (29.5r7.7 d) in comparison
allografts treated with ICOS-Ig or CsA in comparison
with that in untreated mice (8.5±1 d) (P<0.05). CsA
with that in untreated allografts. Rejected grafts from
group also similarly prolonged cardiac allograft
untreated recipients demonstrated diffuse inflammatory
survival (21±4.5 d). ICOS-Ig combined with CsA group
cell infiltration, myocyte destruction, and interstitial
256
Zhang Peng et al. / Journal of Medical Colleges of PLA 24 (2009) 249–258
hemorrhage, whereas scattered inflammatory cells and relatively preserved myocytes were seen in the grafts treated with ICOS-Ig or CsA. Moreover, ICOS-Ig combined with CsA treatment almost completely suppressed lymphocytes infiltration of the grafts, and myocytes were undamaged. (Fig. 4B)
3.5. ICOS-Ig and CsA induced donor-specific hyporesponsiveness The authors performed donor-specific MLR using sensitized splenocytes from the recipients 7 d after transplantation. T-cell proliferation was significantly inhibited by ICOS-Ig or CsA compared with that in the untreated or IgG group (P<0.05). ICOS-Ig and CsA combined
synergistically
and
more
efficaciously
suppressed T-cell proliferation as compared with their respective treatment alone (P<0.05) (Fig. 5). Moreover, CTL assays were completed to evaluate anti-donor T-cell
cytotoxic
responses
demonstrating
donor-
Fig. 6. CTL responses of splenic T cells from the recipients 7
specific graft acceptance. In this assay, effector T cells
d after transplantation. A. Effector T cells from all groups has
from untreated and control IgG groups had strong CTL
similar low CTL ability to syngeneic C57BL/6 targets. B.
ability to allogeneic BALB/c target cells,
Effector T cells from untreated and control IgG groups has strong CTL ability to allogeneic BALB/c target cells, while the CTL ability of ICOS-Ig and CsA combined treated mice is significantly decreased.
while the CTL ability of ICOS-Ig and CsA treated mice was significantly decreased (Fig. 6). These results suggested that graft survival was a consequence of a long-term donor-specific hyporesponsiveness.
4. Discussion Fig. 5. The proliferation of splenic T cells from the recipients 7 d after transplantation. T-cell proliferation was significantly
Inducible costimulatory molecule (ICOS), with its
inhibited by ICOS-Ig or CsA ( P<0.05 as compared with those in
ligand B7h, is a recently discovered costimulatory
the untreated or IgG group). ICOS-Ig and CsA combined
molecule of the CD28 family [4,5]. ICOS signal
a
synergistically suppressed T-cell proliferation ( P<0.05 compared
blockade has been shown to be an effective strategy to
with their respective treatment alone).
prevent allogeneic graft rejection [8-11]. Whether
b
257
Zhang Peng et al. / Journal of Medical Colleges of PLA 24 (2009) 249–258
concomitant use of ICOS blockade and conventional
response is illustrated in mice treated with combined
immunosuppressive drugs has a beneficial or adverse
ICOS-Ig and CsA.
effect on preventing rejection and promoting long-term
Then, we addressed the question as to whether the
allograft survival or transplant tolerance has not been
concomitant use of ICOS-Ig and CsA could induce the
clear so far. The present study was designed to analyse
prolongation of cardiac allograft survival. It was found
the influence of ICOS-Ig combined with CsA on
that the combined treatment resulted in long-time
cardiac allograft survival. The rational for this
survival (>100 d) of cardiac allograft, whereas ICOS-Ig
combination was based on several previous findings.
or CsA used alone was less effective. In addition to the
CsA was known to inhibit primary T-cell activation at
survival data, histological examination showed that the
the level of intracellular calcium mobilization, and thus
ICOS-Ig
downregulated
other
completely suppressed T-cell infiltration of the graft
cytokines[16]. While, ICOS signal blockade by
and cytotoxic responses in comparison with their
ICOS-Ig was effective on activated T cells because
respective treatment alone (P<0.05). We also tested that
ICOS was expressed on T cells only after stimulation,
MLR and CTL using sensitized splenocytes from the
and ICOS signaling might act synergistically with
recipients 7 d after transplantation. These results
IL-2-mediated signal transduction in T-cell activation
suggested that graft survival was a consequence of a
[17], strengthening the rationale for blockade of both
long-term donor-specific hyporesponsiveness.
expression
of
IL-2
and
and
Csa
combined
treatment
almost
pathways. Moreover, this combination was chosen
In summary, ICOS-Ig combined with CsA
based on CsA's proven efficacy clinically in transplant
treatment may be a novel post-transplantation regimen
patients [18].
to establish a donor-specific tolerant state by biological
In this study, we generated an ICOS-Ig fusion
rather than pharmacological mechanisms.
protein consisting of an extracellular portion of human ICOS and Fc portion of human IgG. We first used an
References
MLR assay here to confirm the effect of ICOS-Ig on T-cell proliferation. Addition of ICOS-Ig exhibited
1.
Clarkson MR, Sayegh MH. T-cell costimulatory pathways
dose-dependent inhibition to T-cell proliferation, similar
in allograft rejection and tolerance. Transplantation 2005;
to previous study [19]. Furthermore, we also found that
80(5):555-563.
the lymphocytes collected from MLR in the presence of
2.
ICOS-Ig revealed suppressed responsiveness to same donor lymphocytes restimulation. It seemed that ICOS-Ig
Greenwald RJ, Freeman GJ, Sharpe AH. The B7 family revisited. Annu Rev Immunol 2005; 23:515-548.
3.
Quzada SA, Jarvinen LZ, Lind FF, et al. CD40/CD154
could induce donor-specific hyporesponsiveness, that was
interactions at the interface of tolerance and immunity.
to say, induce donor-specific tolerance. Next, we tested
Annu Rev Immunol 2004; 22:307-328.
in our in vivo system whether the combination of
4.
Hutloff A, Dittrich AM, Beier KC, et al. ICOS is an
ICOS-Ig with CsA had a beneficial or adverse effect on
inducible
allogeneic T cell proliferation inhibition. The results
functionally related to CD28. Nature 1999; 397:263–66.
showed that mice treated with ICOS-Ig or CsA alone +
had a significant reduction in allo-reactive CD4 and +
CD8 T-cell proliferation. The most striking decay in
5.
T-cell
co-stimulator
structurally
and
Yoshinaga SK, Whoriskey JS, Khare SD, et al. T-cell costimulation through B7RP-1 and ICOS. Nature 1999; 402:827–832.
258
6.
Zhang Peng et al. / Journal of Medical Colleges of PLA 24 (2009) 249–258
Coyle AJ, Lehar S, Lloyd C, et al. The CD28-related molecule ICOS is required for effective T cell-dependent
7.
frequency of alloreactive T cells in vivo: new answers to an
McAdam AJ, Chang TT, Lumelsky AE, et al. Mouse
old question. J Immunol 2001; 166(2):973-981.
enhanced
by
CD28
costimulation
and
regulates
differentiation of CD4+ T cells. J Immunol 2000;
14. Yukihiro T, Zhang QW, Masahiro Y, et al. Improved technique of heterotopic cervical heart transplantation. Transplantation 1997; 64: 1598-1601. 15. Matzinger P. The JAM test. A simple assay for DNA
165:5035–5040. Ozkaynak E, Gao W, Shemmeri N, et al. Importance of
fragmentation and cell death. J Immunol Methods 1991;
ICOS-B7RP-1 costimulation in acute and chronic
145: 185– 192. 16. Kapturczak MH, Meier-Kriesche HU, Kaplan B.
allograft rejection. Nat Immunol 2001; 2:591–596. 9.
13. Suchin EJ, Langmuir PB, Palmer E, et al. Quantifying the
immune responses. Immunity 2000; 13:95–105. inducible costimulatory molecule (ICOS) expression is
8.
Immunol 2003; 170(12):5842-5850.
Kosuge H, Suzuki J, Gotoh R, et al. Induction of
Pharmacology of calcineurin antagonists. Transplant
immunologic
Proc 2004; 36(2 Suppl):25S-32S.
tolerance
to
cardiac
allograft
by
simultaneous blockade of inducible co-stimulator and cytotoxic
T-lymphocyte
antigen
4
pathway.
17. Riley JL, Blair PJ, Musser JT, et al. ICOS costimulation requires IL-2 and can be prevented by CTLA-4 engagement. J Immunol 2001; 166: 4943- 4948.
Transplantation 2003; 75:1374–1379. 10. Nanji SA, Hancock WW, Anderson CC, et al. Multiple
18. Banner NR, Yacoub MH. Cyclosporine in thoracic organ
combination therapies involving blockade of ICOS/
transplantation. Transplant Proc 2004; 36(2 Suppl):
B7RP-1 costimulation facilitate long-term islet allograft
302S-308S. 19. Aicher A, Hayden-Ledbetter M, Brady WA, et al.
survival. Am J Transplant 2004; 4:526–536. 11. Hara Y, Kitazawa Y, Funeshima N, et al. Anergic
Characterization of human inducible costimulator ligand
lymphocytes generated by blocking CD28 and ICOS
expression and function. J Immunol 2000; 164:
pathways in vitro prolong rat cardiac graft survival. Int
4689–4696.
Immunopharmacol 2006; 6(7):1143-1151. 12. Elhalel MD, Huang JH, Schmidt W, et al. CTLA-4. FasL induces
alloantigen-specific
hyporesponsiveness.
J
ďEdior Lu RenminĐ