You are on page 1of 9

Carbon Monoxide Produced by Heme

Oxygenase-1 Suppresses T Cell Proliferation


via Inhibition of IL-2 Production
This information is current as Hyun-Ock Pae, Gi-Su Oh, Byung-Min Choi, Soo-Cheon
of December 27, 2016. Chae, Young-Myeong Kim, Khee-Rhin Chung and
Hun-Taeg Chung
J Immunol 2004; 172:4744-4751; ;
doi: 10.4049/jimmunol.172.8.4744
http://www.jimmunol.org/content/172/8/4744

Downloaded from http://www.jimmunol.org/ by guest on December 27, 2016


References This article cites 32 articles, 9 of which you can access for free at:
http://www.jimmunol.org/content/172/8/4744.full#ref-list-1
Subscriptions Information about subscribing to The Journal of Immunology is online at:
http://jimmunol.org/subscriptions
Permissions Submit copyright permission requests at:
http://www.aai.org/ji/copyright.html
Email Alerts Receive free email-alerts when new articles cite this article. Sign up at:
http://jimmunol.org/cgi/alerts/etoc

The Journal of Immunology is published twice each month by


The American Association of Immunologists, Inc.,
9650 Rockville Pike, Bethesda, MD 20814-3994.
Copyright 2004 by The American Association of
Immunologists All rights reserved.
Print ISSN: 0022-1767 Online ISSN: 1550-6606.
The Journal of Immunology

Carbon Monoxide Produced by Heme Oxygenase-1 Suppresses


T Cell Proliferation via Inhibition of IL-2 Production1

Hyun-Ock Pae,* Gi-Su Oh,* Byung-Min Choi, Soo-Cheon Chae, Young-Myeong Kim,
Khee-Rhin Chung, and Hun-Taeg Chung2*
Heme oxygenase-1 (HO-1) catabolizes heme into CO, biliverdin, and free iron and serves as a protective enzyme by virtue of its
anti-inflammatory, antiapoptotic, and antiproliferative actions. Previously, we have demonstrated that human CD4 T cells
express HO-1 and that HO-1-overexpressing Jurkat T cells tend to display lower proliferative response. The aim of this study is
to elucidate the mechanism(s) by which HO-1 can mediate its antiproliferative effect on CD4 T cells. Among the three HO-1
byproducts, only CO showed suppressive effect on T cell proliferation in response to anti-CD3 plus anti-CD28 Abs, mimicking the
antiproliferative action of HO-1. CO blocked the cell cycle entry of T cells, which was independent of the guanylate cyclase/cGMP
pathway. CO also suppressed the secretion of IL-2, and this suppressive effect of CO on IL-2 secretion mediated the antiprolif-

Downloaded from http://www.jimmunol.org/ by guest on December 27, 2016


erative action of CO. CO selectively inhibited the extracellular signal-regulated kinase pathway, which could explain the sup-
pressive effects of CO on T cell proliferation and IL-2 secretion. Based on these findings, we suggest that HO-1/CO suppresses T
cell proliferation and IL-2 secretion, possibly via its inhibition of extracellular signal-regulated kinase activation. The Journal of
Immunology, 2004, 172: 4744 4751.

H eme oxygenase-1 (HO-1)3 is the rate-limiting enzyme in processes in the T cell-mediated immune response and that they
the conversion of heme to CO, free iron, and biliverdin, are associated with increased release of IL-2 (7).
the latter being reduced to bilirubin by biliverdin reduc- The mitogen-activated protein (MAP) kinase cascade is one of
tase. HO-1 is induced by a variety of stimuli in many types of cells, the most ancient and evolutionarily conserved signaling pathways,
including human CD4 T cells (1). Several studies have demon- which is also important for many processes occurring in immune
strated anti-inflammatory properties of HO-1 (2, 3), the existence responses. Three major groups of MAP kinases have been de-
of which is further supported by the fact that HO-1-deficient mice scribed in mammalian cells: they are the extracellular signal-reg-
develop a progressive chronic inflammatory state (4, 5) and that a ulated kinase (ERK), the Jun NH2-terminal kinase (JNK), and the
human lacking HO-1 enzymatic activity died of an inflammatory p38 kinase (8). ERK, JNK, and p38 pathways are rapidly up-reg-
syndrome (6).
ulated by engagement of the TCR in T cells and play a critical role
After the receipt of signals from APCs through TCR and CD28
in the events leading to activation and increased IL-2 secretion (8).
costimulator, CD4 T cells are triggered to produce IL-2 and enter
There are two isoforms of ERK, ERK-1 and -2. They can be ac-
the cell cycle. During or after several days of rapid cell division,
tivated by MAP/ERK kinase (MEK). ERK activation is dependent
these cells differentiate into one of two classes of effector CD4 T
cells (Th1 and Th2 cells). It is generally accepted that activation on p56lck and coupling of the TCR/CD3 complex to p21ras, with
and subsequent proliferation of resting CD4 T cells are essential subsequent activation of the Raf-1/MEK/ERK kinase cascade (8,
9). JNK activation also requires p21ras and signals generated by
the CD28 costimulatory receptor (10). The activation of the Raf-
1/MEK/ERK pathway is essential for induction of IL-2 transcrip-
*Department of Microbiology and Immunology, School of Medicine, and Genomic tion in T cells (11). After phosphorylation of c-jun by JNK, acti-
Research Center for Immune Disorders, Wonkwang University, Iksan, Chonbuk, Re-
public of Korea; Vascular System Research Center and Department of Molecular and
vated c-fos and c-jun combine to form the AP-1 protein required
Cellular Biochemistry, Kangwon National University School of Medicine, Chunchon, for IL-2 synthesis (12). Interestingly, deficient ERK and JNK ac-
Kangwon-Do, Republic of Korea; and Seoul National University Medical School, tivations have been reported to exist in clones that are anergized
Seoul, Republic of Korea
(13). However, there is also evidence that ERK inhibition alone
Received for publication October 13, 2003. Accepted for publication January
23, 2004. suppressed T cell proliferation, but did not induce anergy (14).
The costs of publication of this article were defrayed in part by the payment of page CO, a reaction product of HO-1 activity, has been shown to be
charges. This article must therefore be hereby marked advertisement in accordance highly protective in several rodent disease models (15, 16). It has
with 18 U.S.C. Section 1734 solely to indicate this fact.
anti-inflammatory, antiapoptotic, and antiproliferative effects (17),
1
This work was supported by a grant from the Korea Health 21 R&D Project of the
Ministry of Health and Welfare (01-PJ3-PG6-01GN09-003).
thereby conferring, at least in part, the protective effects of HO-1.
2 Furthermore, the MAP kinase pathway has been shown to mediate
Address correspondence and reprint requests to Dr. Hun-Taeg Chung, Department
of Microbiology and Immunology, Wonkwang University Medical School, 344-2 the biological effects of CO: the p38 MAP kinase activation me-
Shinyong-Dong, Iksan, Chonbuk 570-749, Republic of Korea. E-mail address: diates the cytoprotective effect of CO on ischemia-reperfusion lung
htchung@wonkwang.ac.kr
3
injury (18), CO prevents glucose deprivation-induced cytotoxicity
Abbreviations used in this paper: HO-1, heme oxygenase-1; MAP, mitogen-acti-
vated protein; ERK, extracellular signal-regulated kinase; JNK, Jun NH2-terminal through ERK MAP kinase inactivation in rat hepatocytes (19), and
kinase; MEK, MAP/ERK kinase; CoPP, cobalt protoporphyrin; RuCO, tricarbonyl- the antiproliferative effect of CO on smooth muscle cells and hu-
dichlororuthenium (II) dimer; Hb, hemoglobin; ODQ, 1H-[1,2,4]oxadiazolo-[4,3-
a]quinoxalin-1-one; DN-MEK1, dominant-negative MEK1; CA-MEK1, constitu- man airway smooth muscle cells requires p38 MAP kinase acti-
tively active MEK1. vation (20) and ERK MAP kinase inactivation (21), respectively.

Copyright 2004 by The American Association of Immunologists, Inc. 0022-1767/04/$02.00


The Journal of Immunology 4745

We have recently demonstrated that human CD4 T cells ex- were activated in flat-bottom, 96-well plates precoated with the anti-CD3
press HO-1 and that HO-1-overexpressing Jurkat T cells tend to Ab (1 g/ml) in the presence of soluble anti-CD28 Ab (1 g/ml). CO gas,
display lower proliferative response (1). The aim of this study, CO donor, CO scavenger, and ERK inhibitor at the appropriate concen-
trations were preincubated for 1 h before being added to plates and stim-
therefore, was to elucidate the mechanism(s) by which HO-1 could ulated with anti-CD3 plus anti-CD28 Abs. Culture supernatants were har-
mediate its antiproliferative effect on CD4 T cells, and we dem- vested after 48 h of incubation for measurements of cytokine
onstrated that CO, a reaction product of HO-1, could suppress IL-2 concentrations. Cytokine analysis was performed by analysis of super-
secretion, probably by inhibiting ERK activation, and thereby re- natants with commercially available ELISA kits for human IL-2, IFN-,
and IL-10 (R&D Systems). After 4 days of culture, proliferation was
sulted in suppression of T cell proliferation in response to anti- assessed by [3H]TdR (0.5 Ci/well; Amersham Pharmacia Biotech,
CD3 plus anti-CD28 Abs. Piscataway, NJ) uptake for the next 16 h.

Materials and Methods Evaluation of cGMP


Reagents, cytokines, and Abs Different pools of CD4 T cell samples were incubated for 1 h at 37C in
the presence of 20 M CO or air. To inhibit phosphodiesterase activity,
RPMI 1640 supplemented with 2 mM L-glutamine, 1% nonessential amino 31-isobutyl-1-methyl xanthine (10 M) was added to the cell suspension.
acids, 1% pyruvate, 100 U/ml penicillin, 100 g/ml streptomycin (Life The concentration of cGMP was determined by a radioimmunoassay kit
Technologies, Grand Island, NY), and 10% FBS (HyClone Laboratories, using 125I-labeled cGMP (Amersham Pharmacia Biotech). Briefly, after
Logan, UT) was used as complete medium in all cultures. Anti-CD3 (clone incubation, 500 l of 10% TCA was added to the cell suspensions. The
UCHT1) and anti-CD28 (clone CD28.2) Abs were purchased from Immu- samples were then centrifuged and TCA was extracted with 0.5 M tri-n-
notech (Westbrook, ME) and BD PharMingen (San Diego, CA), respec- octylamine dissolved in TCA, and the samples were then acetylated with
tively. Phospho (p)-specific rabbit Abs to p-ERK-1/2, p-JNK-1/2, and p- acetic anhydride and the amount of cGMP in the aqueous phase was
p38 were obtained from Cell Signaling Technology (Beverly, MA), and

Downloaded from http://www.jimmunol.org/ by guest on December 27, 2016


measured.
ERK-1/2, JNK-1/2, and p38 Abs were from Santa Cruz Biotechnology
(Santa Cruz, CA). PE- and FITC-conjugated and HRP-conjugated second- Western blotting analysis
ary Abs were purchased from BD PharMingen and Santa Cruz Biotech-
nology, respectively, and U0126 and cobalt protoporphyrin (CoPP) were Cells were solubilized in ice-cold 1% Triton X-100 lysis buffer supple-
from Promega (Madison, WI) and Porphyrin Products (Logan, UT), re- mented with protease and phosphatase inhibitors as described previously
spectively. SP600125 was purchased from Calbiochem (San Diego, CA). (21). After 30 min on ice, the lysates were clarified by centrifugation and
CO gas (99%), tricarbonyldichlororuthenium (II) dimer (RuCO), the protein concentration was determined with the Pierce bicinchoninic
SB203580, hemoglobin (Hb), and 1H-[1,2,4]oxadiazolo-[4,3-a]quinoxalin- acid protein assay reagent. Proteins (20 g) were resolved by SDS-PAGE
1-one (ODQ) were purchased from Sigma-Aldrich (St. Louis, MO), and (10% acrylamide), transferred to nitrocellulose membranes, and probed
anti-HO-1 and anti--actin Abs were from Santa Cruz Biotechnology. with specific Abs (diluted 1/1,000), followed by incubation with secondary
cDNAs encoding constitutively active (S218E/S222E) and dominant neg- HRP-conjugated Ab (1/100,000). Bands were detected using the Luminol
ative (S218A/S222A) mutants of MEK1 were kindly provided by Dr. K. Y. chemiluminescent detection reagents (New England Biolabs, Beverly,
Choi (Yonsei University, Seoul, Korea). HO-1 cDNA was a kind gift from MA). Blots were exposed to autoradiographic film (DuPont Merk Chem-
Dr. A. M. K. Choi (University of Pittsburgh, Pittsburgh, PA). Recombinant istry Department, Wilmington, DE) for 12 min for detection.
human IL-2, IL-10, IFN-, and anti-IL-10 neutralizing Ab were obtained
from R&D Systems (Minneapolis, MN). CFSE was purchased from Mo- Flow cytometry
lecular Probes (Eugene, OR). The other reagents were from Sigma-Aldrich. Cells were suspended in HBSS containing 5% FBS, fixed by the drop-wise

Isolations of resting CD4 T cells and CD8 T cells from addition of ice-cold 70% ethanol to a final 50% concentration, and held on
ice for 1 h. After extensive washing, the cells were suspended in HBSS
peripheral blood containing 50 g/ml propidium iodide (Sigma-Aldrich) and 50 g/ml
PBMCs were isolated from healthy blood by Ficoll-Paque density gradient RNase A (Boehringer Mannheim, Indianapolis, IN) and were incubated for
centrifugation. After three washes in HBSS, CD4 T cells were isolated 1 h at room temperature. In some experiments, the states of ERK activation
from PBMCs using the MACS negative depletion system (Miltenyi Biotec, and HO-1 expression were determined by intracellular staining with either
Auburn, CA). No contamination with CD8 T cells, B cells, monocytes, or anti-p-ERK or anti-HO-1 Abs labeling either FITC- or PE-conjugated sec-
NK cells was detected. Isolation of CD8 cells was performed using a ondary Ab, as described previously (23). Stained cells were analyzed by
negative CD8 T cell isolation kit (Miltenyi Biotec). flow cytometry on a FACSVantage with CellQuest software (BD Bio-
sciences, Franklin Lakes, New Jersey). G0/G1, S, and G2/M populations
Cell transfection were quantified using the ModFiT program (BD Biosciences).
HO-1, dominant-negative MEK1 (DN-MEK1), and constitutively active CFSE dilution assay
MEK1 (CA-MEK1) were cloned into pcDNA3 (Invitrogen, San Diego,
CA). Jurkat T cells (5 106) were transfected with 10 g of constructs by Naive CD4 T cells were labeled with 1.5 M CFSE for 10 min at 37C
electroporation at 270V, 950 F in serum-free RPMI 1640 using a Gene in serum-free RPMI 1640. Cells were washed twice in RPMI 1640 con-
Pulser (Bio-Rad, Richmond, CA) followed by culture in RPMI 1640 sup- taining 10% FBS and were stimulated as indicated. Cell division was as-
plemented with 10% FBS for 48 h and plating on 96-well plates at 5 105 sessed at 4 days by determining the pattern of CFSE dilution using flow
cells/well in the presence of 1.25 mg/ml G418. Single stable clones of each cytometry.
transfectant were isolated and expanded.
Statistics
CO exposure Data were expressed as mean SEM of the individual titer. Levels of
T cells were exposed to compressed air or varying concentrations of CO significant differences between groups were determined by the Student t
(200 ppm), as previously described (22). Briefly, 1% CO in compressed air test. Values of p 0.01 were considered statistically significant.
was mixed with compressed air with or without 5% CO2 in a stainless steel
mixing cylinder before being delivered into the exposure chamber. The cell Results
culture chamber was humidified and maintained at 37C. A CO analyzer HO-1 expression in T cells is antiproliferative
(CM-525HB; Gastec, Kanagawa, Japan) was used to monitor CO levels
continuously in the chambers. T cells were grown in RPMI 1640 medium We found that pharmacological expression of HO-1 by the HO-1
containing 10% FBS in a humidified atmosphere of 5% CO2 in air or 200 inducer CoPP in PBMCs containing monocytes and lymphocytes
ppm CO and 5% CO2 in air. was antiproliferative in response to anti-CD3 plus anti-CD28 Abs
T cell activation, cytokine analysis, and proliferation (Fig. 1A). This suggested that HO-1 expression in monocytes
and/or lymphocytes could suppress T cell proliferation. Therefore,
The human acute T cell leukemia Jurkat clone E6-1 was obtained from the
American Type Culture Collection (Rockville, MD). Purified CD4 T cells
we examined whether CoPP could induce HO-1 expression in hu-
or Jurkat T cells were cultured in RPMI 1640 supplemented with 10% man CD4 T cells purified from PBMCs and also whether HO-1
heat-inactivated FBS at 37C in 5% CO2. CD4 T cells (2 105/well) expression by CoPP could be antiproliferative. Preincubation of
4746 CO SUPPRESSES T CELL PROLIFERATION VIA IL-2 INHIBITION

FIGURE 1. Effects of HO-1 expression on T


cell proliferation. PBMCs (A) and CD4 T cells
(B) purified from PBMCs were incubated for 12 h
with CoPP, and Jurkat T cells (C) were transfected
with human HO-1 gene (Jurkat/HO-1) or empty
vector (Jurkat/pcDNA). HO-1 expressions (top
panels) were confirmed by Western blot analysis
after 12-h incubation of either PBMCs or CD4 T
cells with CoPP (20 M) or after stable transfec-
tion of Jurkat T cells with either HO-1 gene or
empty vector. One of three experiments is shown.
T cell proliferation (bottom panels) was deter-
mined after 4-day stimulation of T cells with anti-
CD3 plus anti-CD28 Abs. Values are the mean
SEM of six triplicate experiments. , p 0.01.

Downloaded from http://www.jimmunol.org/ by guest on December 27, 2016


CD4 T cells with CoPP for 12 h resulted in an apparent increase liferative response, whereas a 12-h preincubation of the cells with
in HO-1 expression and a significant decrease in T cell prolifera- CoPP in the presence of Hb showed no suppressive effect (Fig.
tion (Fig. 1B). CoPP preincubation also suppressed the prolifera- 2C). Similarly, CoPP in the presence of Hb did not suppress the
tive response of CD8 T cells (data not shown). To further inves- proliferation of either CD8 T cells or Jurkat T cells (data not
tigate the contribution of HO-1 expression to T cell proliferation, shown). CO gas, RuCO, CoPP, or Hb at concentrations used in
we transfected the HO-1 gene into human Jurkat T cells (Fig. 1C). these experiments showed no effect on T cell viability (data not
In agreement with the CoPP data, overexpression of HO-1 sup- shown). These results suggest that exogenous and endogenous CO
pressed T cell proliferation (Fig. 1C). These data demonstrate that can mimic the antiproliferative action of HO-1.
HO-1 expression in T cells is antiproliferative.
CO blocks the cell cycle entry of human T cells, independent of
Exogenous and endogenous CO can suppress human T cell the guanylate cyclase/cGMP pathway
proliferation, mimicking antiproliferative action of HO-1 We observed that addition of CO to CD4 T cells 24 h after stimu-
We next determined which of the HO-1 metabolites could mediate lation with anti-CD3 plus anti-CD28 Abs had no effect on prolifera-
the antiproliferative effect of HO-1 on human CD4 T cells. Thus, tion (Fig. 3A). This suggested that CO was blocking early events
we preincubated CD4 T cells with CO gas, bilirubin, or free iron leading to T cell activation. Therefore, we examined the effect of
(Fe2) for 1 h before stimulation of the T cells with anti-CD3 plus RuCO on the cell cycle and could show that CD4 T cells did not
anti-CD28 Abs. Only CO gas had an antiproliferative effect on progress past G0/G1 phase, if the cells were stimulated after preincu-
CD4 T cells (Fig. 2A). Similarly, the CO-releasing compound bation with RuCO for 1 h (Fig. 3B, lower left panel). We could also
RuCO suppressed the proliferative response of CD4 T cells in a show that CO was not inhibitory to cell cycle progression, if the ac-
dose-dependent manner (IC50 20 M), but not in the presence tivation was already progressed beyond TCR signaling (Fig. 3B,
of the CO scavenger Hb (Fig. 2B). In addition, a 12-h preincuba- lower right panel). Furthermore, a kinetic CFSE analysis with in-
tion of CD4 T cells with CoPP significantly suppressed the pro- tracellular HO-1 staining revealed that CO was not inhibitory to T

FIGURE 2. Suppressive effects of CO on the proliferation of CD4 T cells. A, CD4 T cells were preincubated for 1 h with CO gas (200 ppm), bilirubin
(20 M), or Fe2 (20 M) before stimulation. B, CD4 T cells were preincubated for 1 h without (control) or with indicated concentrations of RuCO in
the absence or presence of Hb (80 M) before stimulation. C, CD4 T cells were preincubated for 12 h with CoPP (20 M) in the absence or presence
of Hb (80 M). The CD4 T cells were then stimulated for 4 days with anti-CD3 plus anti-CD28 Abs. Proliferation was measured, as described in Materials
and Methods. Data (B) are expressed as percent of control. Values are the mean SEM of six triplicate experiments. , p 0.01.
The Journal of Immunology 4747

FIGURE 3. Blocking effects of CO on the cell cy-


cle progression of human T cells. A, CD4 T cells
labeled with CFSE were cultured for 1 h with medium
(upper left panel) or were stimulated for 4 days with
anti-CD3 plus anti-CD28 Abs (upper right panel).
CD4 T cells labeled with CFSE were preincubated
for 1 h with RuCO (20 M) and stimulated for 4 days
with anti-CD3 plus anti-CD28 Abs (lower left panel).
RuCO (20 M) was added after 24-h stimulation of
CD4 T cells with anti-CD3 plus anti-CD28 Abs
(lower right panel), and the T cells were further cul-
tured for 3 days. CFSE fluorescence was measured by
flow cytometry. B, CD4 T cells were cultured for
48 h with medium (upper left panel) or were stimu-
lated with anti-CD3 plus anti-CD28 Abs (upper right
panel). CD4 T cells were preincubated for 1 h with
RuCO (20 M) and were stimulated for 48 h with

Downloaded from http://www.jimmunol.org/ by guest on December 27, 2016


anti-CD3 plus anti-CD28 Abs (lower left panel).
RuCO (20 M) was added after 24-h stimulation of
CD4 T cells with anti-CD3 plus anti-CD28 Abs
(lower right panel), and the T cells were further cul-
tured for 24 h. Cell cycle analysis was performed, as
described in Materials and Methods. Cells with
sub-2N amounts of DNA were not gated and com-
prised from 10 to 25% of the population. C, CD4 T
cells labeled with CFSE were stimulated for 4 days
with anti-CD3 plus anti-CD28 Abs. Cells were har-
vested and stained with anti-HO-1-PE. Dotted box in
one of three representative dot plots of HO-1 expres-
sion vs CFSE represents the population of HO-1-pos-
itive cells. D, CD4 T cells were preincubated for 1 h
with medium or RuCO (20 M) in the presence or
absence of ODQ (30 M) and were stimulated for 48 h
with anti-CD3 plus anti-CD28 Abs. Each bar repre-
sents the percentage of cells belonging to each cell
cycle in flow cytometric analysis. Values are the
mean SEM of six triplicate experiments.

cell cycle progression after a certain stage of activation because cretion levels of IFN- and IL-10 (Table I). Moreover, preincu-
HO-1/CO was induced during T cell activation (Fig. 3C). bation of CD4 T cells with CoPP inhibited IL-2 secretion,
Most of the biological effects of CO are attributed to its abilities whereas preincubation with CoPP in the presence of Hb showed no
to modulate the activity of guanylate cyclase and to increase the inhibitory effect (Figs. 4C).
cellular cGMP levels (17). Therefore, we examined whether the To test whether the observed CO-induced suppression of T cell
blockage of cell cycle progression by RuCO could be due to ele- proliferation could be due to reduction of secretion levels of IL-2,
vation of cGMP and found that ODQ, a potent inhibitor of the we added recombinant human IL-2 to the cultures of T cells stim-
soluble form of guanylyl cyclase, had no effect on RuCO-induced ulated with anti-CD3 plus anti-CD28 Abs in the presence of
blockage of the cell cycle (Fig. 3D). However, ODQ significantly RuCO. The exogenous IL-2 effectively reversed the antiprolifera-
blocked RuCO-induced cGMP synthesis in T cells (data not tive effects of CO in T cell culture (Fig. 4D). Unlike IL-2, neither
shown). IFN- nor IL-10 reversed the antiproliferative effects of CO (Fig.
4D). These data indicate that the suppression of T cell proliferation
CO inhibits IL-2 secretion by activated T cells, contributing by CO could be due to its inhibition of IL-2 secretion.
antiproliferative effect of CO
IL-2 is an important regulator of T cell proliferation and is released CO inhibits ERK phosphorylation in activated T cells, probably
by activated T cells (8 14). We observed that, among three HO-1 mediating the inhibitory effect of CO on IL-2 secretion
reaction products, only CO gas inhibited IL-2 secretion by acti- The ERK MAP kinases, which have been implicated as an intra-
vated CD4 T cells (Fig. 4A). Similarly to CO gas, RuCO also cellular target to contribute to certain CO-induced biological ac-
inhibited the secretion of IL-2 by CD4 T cells in a dose-depen- tions (19, 21), also play critical roles in T cell proliferation and
dent manner (IC50 20 M), but not in the presence of Hb (Fig. IL-2 secretion (13, 14). Therefore, we examined the effects of CO
4B). Additionally, RuCO at higher concentrations reduced the se- gas and RuCO on ERK-1/ERK-2 activation in CD4 T cells. CO
4748 CO SUPPRESSES T CELL PROLIFERATION VIA IL-2 INHIBITION

Table I. Effects of RuCO on IFN- and IL-10 secretions in CD4 T


cellsa

Treatment IFN- (pg/ml) IL-10 (pg/ml)

Medium 300 20 350 10


RuCO (10 M) 320 10 330 9
RuCO (20 M) 290 12 300 13
RuCO (40 M) 210 5b 240 5b
RuCO (80 M) 150 7b 170 12b
a
Cells were preincubated for 1 h with medium (control for either IFN- or IL-10)
or RuCO at indicated concentrations and were stimulated for 48 h with anti-CD3 plus
anti-CD28 Abs. IFN- and IL-10 secretions were measured as described in Materials
and Methods. Results are the means SEM of six experiments.
b
Significance of difference from each control, p 0.01.

did not further reduce T cell proliferative response when ERK


pathway was effectively blocked by U0126 (Fig. 5C). These results
further suggest that the ERK pathway might be involved in the
suppressive effects of CO on T cell proliferation.

Downloaded from http://www.jimmunol.org/ by guest on December 27, 2016


CO may block upstream signaling cascades of MEK1 in
activated T cells, thereby resulting in ERK inactivation
Several studies have brought evidence that the intracellular signals
that mediate activation of transcription factors regulating IL-2
gene transcription in human T cells involve p21ras-mediated sig-
naling pathways (24 27). These studies obtained with T cell lines
collectively suggest that IL-2 gene transcription might require the
p21ras/Raf-1/MEK/ERK phosphorylation cascade.
To further evaluate the role of the ERK pathway in CO-induced
inhibition of IL-2 secretion, we transfected DN-MEK1 and CA-
MEK1 genes into Jurkat T cells to selectively inhibit or activate
the ERK1 pathway. DN-MEK1 expression suppressed ERK1 ac-
FIGURE 4. Inhibitory effects of CO on IL-2 secretion. A, CD4 T cells tivation and IL-2 secretion by the simultaneous ligation of CD3
were preincubated for 1 h with CO gas (200 ppm), bilirubin (20 M), or and CD28 Abs (Fig. 6), which was similar to RuCO effects on IL-2
Fe2 (20 M) before stimulation. B, CD4 T cells were preincubated for secretion. Conversely, CA-MEK1 expression enhanced ERK1 ac-
1 h without (control) or with indicated concentrations of RuCO in the tivation and IL-2 secretion by the same stimuli (Fig. 6). No sig-
absence or presence of Hb (80 M) before stimulation. C, CD4 T cells nificant inhibition of IL-2 secretion by RuCO was observed in
were preincubated for 12 h with CoPP (20 M) in the absence or presence DN-MEK1- or CA-MEK1-transfected cells (Fig. 6). These results
of Hb (80 M). D, CD4 T cells were preincubated for 1 h with RuCO (20 suggest that CO might block or inactivate upstream signaling cas-
M) in the presence or absence of IL-2 (50 U/ml), IFN- (50 U/ml), IL-10
cades of MEK1, thereby inhibiting ERK pathway.
(50 U/ml), or anti-IL-10 neutralizing Ab (2 g/ml) before stimulation. The
CD4 T cells were then stimulated for 2 days (AC) or for 4 days (D) with IL-10 may be involved in HO-1 expression in activated CD4 T
anti-CD3 plus anti-CD28 Abs. IL-2 secretion (AC) and proliferation (D)
cells
were measured, as described in Materials and Methods. Data (B) are ex-
pressed as percent of control. Values are the mean SEM of six triplicate It has been reported that IL-10 is able to induce HO-1 expression
experiments. , p 0.01. and to exert its anti-inflammatory effects via the HO-1-dependent
pathway in monocytes (28). In human CD4 T cells, IL-10 secre-
tion (see Table I) as well as HO-1 expression (see Fig. 3C) was
gas and RuCO significantly inhibited ERK phosphorylation in the induced by the simultaneous ligation of CD3 and CD28 Abs. Thus,
activated CD4 T cells (Figs. 5, A and B). In contrast, RuCO had one may ask about whether IL-10 could be involved in HO-1 ex-
no effect on either p38 phosphorylation or JNK phosphorylation pression in CD4 T cells. Although IL-10 itself did not induce
(Fig. 5B). In Jurkat T cells, RuCO also inhibited ERK phosphor- HO-1 expression in naive CD4 T cells, this cytokine further en-
ylation in a dose-dependent manner (data not shown). Next, we hanced HO-1 expression in CD4 T cells stimulated with CD3
were interested in determining the effects of blocking the ERK, plus CD28 Abs (Fig. 7). Moreover, anti-IL-10 neutralizing Ab
p38, and JNK pathways on T cell proliferation. U0126, a selective reduced the level of HO-1 expression in activated CD4 T cells
inhibitor of ERK pathway, SB20358, a selective inhibitor of p38 (Fig. 7). It is most likely that there are many complex signals,
pathway, and SP600125, a selective inhibitor of JNK pathway, including an IL-10 signal, to sufficiently induce HO-1 expression
were used to block each pathway. U0126 significantly suppressed in human CD4 T cells.
T cell proliferation, but SB20358 and SP600125 did not (Fig. 5C).
Suppression of T cell proliferation by U0126 was most likely due Discussion
to reduction of IL-2 levels, because U0126-treated CD4 T cells The antiproliferative and anti-inflammatory HO-1 may play im-
could proliferate when IL-2 was exogenously added to the cultures portant roles in regulating T cell responses. It has been reported
(Fig. 5C). In addition, RuCO at 20 M reduced proliferative re- that HO-1-deficient mice contract a progressive chronic inflamma-
sponse even when p38 and JNK pathways were blocked by tory disease, demonstrated by enlarged spleen and lymph nodes,
SB20358 and SP600125, respectively (Fig. 5C). However, RuCO high peripheral white blood cell counts, and high splenic and
The Journal of Immunology 4749

Downloaded from http://www.jimmunol.org/ by guest on December 27, 2016


FIGURE 6. Effects of RuCO on IL-2 secretion in Jurkat T cells trans-
fected with DN-MEK1 or CA-MEK1. Jurkat T cells were transfected with
empty vector (Vector), DN-MEK1, or CA-MEK1. The effects of DN-
MEK1 and CA-MEK1 expression on ERK phosphorylation were deter-
mined by Western blot (top panel). Transfected cells were preincubated for
1 h with RuCO (20 M) and stimulated for 48 h with anti-CD3 plus
anti-CD28 Abs (bottom panel). IL-2 secretion was measured, as described
in Materials and Methods. Values are the mean SEM of six triplicate
experiments.

lymph node CD4:CD8 T cell ratios with numerous activated


CD4 T cells (4, 5). It has also been reported that splenocytes
isolated from HO-1-overexpressing mice tend to display lower
proliferation indices against allogeneic stimulation in both CD4
and CD8 subsets and markedly increased allotransplant survival
by inhibiting infiltrations of inflammatory cells and CD4 T cells
(29). The administration of a HO-1 inducer to normal mice results
in suppressions of T cell-mediated cytotoxicity and Th1-mediated
cytokine production and decreases in the lymphoproliferative al-
loresponse and differentiation of CTLs (30). HO-1 inducer also
prevents the induction of T cell-mediated experimental auto-
immune encephalomyelitis in rats (31). In accord with these ob-
servations, we recently demonstrated that human CD4 T cells
express HO-1 and that HO-1-overexpressing Jurkat T cells tend to
FIGURE 5. Inhibitory effects of CO on ERK phosphorylation in CD4 display lower proliferative response. In the present study, we ex-
T cells. A, CD4 T cells were preincubated for 1 h with either medium or plored the mechanism(s) by which HO-1 mediated its antiprolif-
CO gas (200 ppm) and were stimulated for 10 min with anti-CD3 plus
erative effect on CD4 T cells and found that the HO-1/CO system
anti-CD28 Abs. The cells were harvested, stained with either isotype con-
trol (open histogram) or anti-p-ERK Abs (filled histogram) and were an-
was an important regulator of T cell responses. CO suppressed
alyzed by flow cytometry. B, CD4 T cells were preincubated for 1 h with proliferation and IL-2 secretion of CD4 T cells, most likely by
RuCO (20 M) and stimulated for 5 or 10 min with anti-CD3 plus anti- inhibiting ERK activation.
CD28 Abs. Whole-cell lysates were prepared and the amounts of phos- The exact mechanisms of HO-1 expression in activated T cells
phorylated ERK1/2 (first panel), p38 (third panel), and JNK (fifth panel) are currently unknown. However, the antiproliferative and anti-
were assessed by Western blot. C, CD4 T cells were preincubated for 1 h inflammatory IL-10 produced by activated CD4 T cells was in-
with medium, the ERK inhibitor U0126 (20 M), the p38 inhibitor volved at least in part in HO-1 expression in T cells (Fig. 7),
SB20358 (20 M), the JNK inhibitor SP600125 (20 M), RuCO (20 M), raising an interesting question of whether HO-1 expression could
or inhibitors plus RuCO and were stimulated for 4 days with anti-CD3 plus be involved in modulating T cell responses. Pharmacological in-
anti-CD28 Abs in the presence or absence of IL-2 (50 U/ml). Proliferation
duction or gene transfer of HO-1 in human T cells was antiprolif-
assay was performed, as described in Materials and Methods. Values are
the mean SEM of six triplicate experiments.
erative (Fig. 1). Among the three HO-1 byproducts, only both ex-
ogenously added and endogenously generated CO-suppressed T
cell proliferation (Fig. 2), mimicking the antiproliferative action of
4750 CO SUPPRESSES T CELL PROLIFERATION VIA IL-2 INHIBITION

T cell proliferative response, but not in the presence of exog-


enously added IL-2 (Fig. 5C). Similarly, an expression of DN-
MEK1 inhibited IL-2 secretion (Fig. 6). These findings led us to
suggest that CO might be able to inhibit the ERK activation, which
leads to inhibition of IL-2 secretion, eventually resulting in sup-
pression of T cell proliferation in response to the simultaneous
ligation of CD3 and CD28 Abs.
Naive CD4 T cells could be potentially exposed to endogenous
CO produced by CD4CD25 regulatory T cells, which consti-
tutively express HO-1 (1), as well as activated CD4CD25 re-
sponder T cells, which can express HO-1 after stimulation. Thus,
we speculate that CO may directly and/or indirectly affect T cell
responses in vivo. It is of interest that CO shows cGMP-indepen-
dent suppressive effects on T cell responses and it can inhibit ERK
activation in activated CD4 T cells. CO could indirectly inhibit
ERK phosphorylation, probably by blocking upstream signals of
MEK (Fig. 6). It could be possible that CO activates the small G
protein, Rap1, which has been shown to inhibit ERK activation by
blocking Ras-dependent activation of the MAP kinase kinase ki-

Downloaded from http://www.jimmunol.org/ by guest on December 27, 2016


nase, Raf-1 (32). Further studies are in progress to explore poten-
tial mechanisms of CO effects on ERK signaling pathways.
In summary, our findings suggest that HO-1/CO induces sup-
pressive effects on T cell proliferation and IL-2 secretion, possibly
via its inhibition of the ERK MAP kinase pathway, which is cur-
rently believed to be an important signaling pathway for mediating
T cell activation. Our findings may contribute not only to our
deeper understanding of the basic roles of HO-1 in the immune
system, but also to our search for novel targets for new therapeutic
approaches to modulate T cell-mediated immune responses.

Acknowledgments
We thank Dr. W. K. Paik for critical reading of the manuscript,
Dr. Augustine M. K. Choi for helpful discussions on a CO incubator, and
Drs. K. Y. Choi and J. S. Chun for the kind gifts of DN-MEK1 and CA-
MEK1 genes.

FIGURE 7. Effects of IL-10 on HO-1 expression in CD4 T cells. References


CD4 T cells were preincubated with medium, IL-10 (50 U/ml), or anti- 1. Pae, H. O., G. S. Oh, B. M. Choi, S. C. Chae, and H. T. Chung. 2003. Differential
IL-10 neutralizing Ab (2 g/ml) and were stimulated for 48 h with anti- expressions of heme oxygenase-1 gene in CD25 and CD25 subsets of human
CD3 plus anti-CD28 Abs. The cells were harvested, stained with either CD4 T cells. Biochem. Biophys. Res. Commun. 306:701.
isotype control (dotted histogram) or anti-HO-1 Abs (solid histogram), and 2. Song, R., M. Kubo, D. Morse, Z. Zhou, X. Zhang, J. H. Dauber, J. Fabisiak,
S. M. Alber, S. C. Watkins, B. S. Zuckerbraun, et al. 2003. Carbon monoxide
analyzed by flow cytometry. One of three experiments is shown. induces cytoprotection in rat orthotopic lung transplantation via anti-
inflammatory and anti-apoptotic effects. Am. J. Pathol. 163:231.
3. Lee, T. S., H. L. Tsai, and L. Y. Chau. 2003. Induction of heme oxygenase-1
HO-1. Neither bilirubin nor free iron showed any effect on prolif- expression in murine macrophages is essential for the anti-inflammatory effect of
erative response under our experimental conditions. CO blocked low dose 15-deoxy-12,14-prostaglandin J2. J. Biol. Chem. 278:19325.
4. Poss, K. D., and S. Tonegawa. 1997. Reduced stress defense in heme oxygenase
the cell cycle entry of T cells, which was independent of guanylate 1-deficient cells. Proc. Natl. Acad. Sci. USA 94:10925.
cyclase/cGMP pathway (Fig. 3). However, it was of great interest 5. Poss, K. D., and S. Tonegawa. 1997. Heme oxygenase 1 is required for mam-
to observe that CO was not inhibitory to cell cycle progression if malian iron reutilization. Proc. Natl. Acad. Sci. USA 94:10919.
6. Kawashima, A., Y. Oda, A. Yachie, S. Koizumi, and I. Nakanishi. 2002. Heme
the T cell activation already progressed beyond TCR signaling oxygenase-1 deficiency: the first autopsy case. Hum. Pathol. 33:125.
(Fig. 3, AC). This suggests that CO blocks early events in T cell 7. Santana, M. A., and Y. Rosenstein. 2003. What it takes to become an effector T
activation. CO also inhibited IL-2 secretion in CD4 T cells stim- cell: the process, the cells involved, and the mechanisms. J. Cell. Physiol.
195:392.
ulated with anti-CD3 plus anti-CD28 Abs (Fig. 4). This inhibitory 8. Dong, C., R. J. Davis, and R. A. Flavell. 2002. MAP kinases in the immune
effect of CO on the IL-2 secretion appears to be responsible for the response. Annu. Rev. Immunol. 20:55.
9. Alberola-Ila, J., and G. Hernandez-Hoyos. 2003. The Ras/MAPK cascade and the
antiproliferative action of CO, because T cell proliferation oc- control of positive selection. Immunol. Rev. 191:79.
curred when IL-2 was exogenously added to the culture (Fig. 4D). 10. Schwartz, R. H. 1977. T cell clonal anergy. Curr. Opin. Immunol. 9:351.
At high doses, CO also inhibited both IFN- and IL-10 secretions 11. Izquierdo, M., S. J. Leevers, C. J. Marshall, and D. Cantrell. 1993. p21ras couples
the T cell antigen receptor to extra signal regulated kinase in T cells. J. Exp. Med.
(Table I), but the additional inhibitions of these cytokines by CO 78:1199.
were not associated with the antiproliferative effect of CO (Fig. 12. Angel, P., and M. Karin. 1991. The role of Jun, Fos and the AP-1 complex in cell
4D). MAP kinase plays an important role in IL-2 secretion, and proliferation and transformation. Biochim. Biophys. Acta 1072:129.
13. Li, W., C. D. Whaley, A. Mondino, and D. L. Mueller. 1996. Blocked signal
there exists a strong correlation between decreased IL-2 secretion transduction to the ERK and JNK protein kinases in anergic CD4 T cells.
and the inhibition of ERK activation in T cells (13, 14). In support Science 271:1272.
of these findings, our data clearly showed that CO selectively in- 14. DeSilva, D. R., E. A. Jones, M. F. Favata, B. D. Jaffee, R. L. Magolda,
J. M. Trzaskos, and P. A. Scherle. 1998. Inhibition of mitogen-activated protein
hibited the ERK pathway in the activated T cells (Fig. 5). Further- kinase kinase blocks T cell proliferation but does not induce or prevent anergy.
more, U0126, a selective inhibitor of ERK activation, suppressed J. Immunol. 160:4175.
The Journal of Immunology 4751

15. Soares, M. P., Y. Lin, J. Anrather, E. Csizmadia, K. Takigami, K. Sato, 24. Lafont, V., B. Rouot, and J. Favero. 1998. The Raf-1/mitogen-activated protein
S. T. Grey, R. B. Colvin, A. M. Choi, K. D. Poss, and F. H. Bach. 1998. Ex- kinase kinase-1/extracellular signal-regulated-2 signaling pathway as prerequisite
pression of heme oxygenase-1 can determine cardiac xenograft survival. Nat. for interleukin-2 gene transcription in lectin-stimulated human primary T lym-
Med. 4:1073. phocytes. Biochem. Pharmacol. 55:319.
16. Sato, K., J. Balla, L. Otterbein, R. N. Smith, S. Brouard, Y. Lin, E. Csizmadia, 25. Franklin, R. A., A. Tordai, H. Patel, A. M. Gardner, G. L. Johnson, and
J. Sevigny, S. C. Robson, G. Vercellotti, et al. 2001. Carbon monoxide generated E. W. Gelfand. 1994. Ligation of the T cell receptor complex results in activation
by heme oxygenase-1 suppresses the rejection of mouse-to-rat cardiac trans- of the Ras/Raf-1/MEK/MAPK cascade in human T lymphocytes. J. Clin. Invest.
plants. J. Immunol. 166:4185. 93:2134.
17. Otterbein, L. E., M. P. Soares, K. Yamashita, and F. H. Bach. 2003. Heme
26. Chen, D., V. Heath, A. OGarra, J. Johnston, and M. McMahon. 1999. Sustained
oxygenase-1: unleashing the protective properties of heme. Trends Immunol.
activation of the raf-MEK-ERK pathway elicits cytokine unresponsiveness in T
24:449.
cells. J. Immunol. 63:5796.
18. Zhang, X., P. Shan, L. E. Otterbein, J. Alam, R. A. Flavell, R. J. Davis,
A. M. Choi, and P. J. Lee. 2003. Carbon monoxide inhibition of apoptosis during 27. Chang, F., L. S. Steelman, J. T. Lee, J. G. Shelton, P. M. Navolanic,
ischemia-reperfusion lung injury is dependent on the p38 mitogen-activated pro- W. L. Blalock, R. A. Franklin, and J. A. McCubrey. 2003. Signal transduction
tein kinase pathway and involves caspase 3. J. Biol. Chem. 278:1248. mediated by the Ras/Raf/MEK/ERK pathway from cytokine receptors to tran-
19. Choi, B. M., H. O. Pae, Y. M. Kim, and H. T. Chung. 2003. Nitric oxide- scription factors: potential targeting for therapeutic intervention. Leukemia
mediated cytoprotection of hepatocytes from glucose deprivation-induced cyto- 17:1263.
toxicity: involvement of heme oxygenase-1. Hepatology 37:810. 28. Lee, T. S., and L. Y. Chau. 2002. Heme oxygenase-1 mediates the anti-
20. Otterbein, L. E., B. S. Zuckerbraun, M. Haga, F. Liu, R. Song, A. Usheva, inflammatory effect of interleukin-10 in mice. Nat. Med. 8:240.
C. Stachulak, N. Bodyak, R. N. Smith, E. Csizmadia, et al. 2003. Carbon mon- 29. Araujo, J. A., L. Meng, A. D. Tward, W. W. Hancock, Y. Zhai, A. Lee,
oxide suppresses arteriosclerotic lesions associated with chronic graft rejection K. Ishikawa, S. Iyer, R. Buelow, R. W. Busuttil, et al. 2003. Systemic rather than
and with balloon injury. Nat. Med. 9:183. local heme oxygenase-1 overexpression improves cardiac allograft outcomes in a
21. Song, R., R. S. Mahidhara, F. Liu, W. Ning, L. E. Otterbein, and A. M. Choi. new transgenic mouse. J. Immunol. 171:1572.
2002. Carbon monoxide inhibits human airway smooth muscle cell proliferation
30. Woo, J., S. Iyer, M. C. Cornejo, N. Mori, L. Gao, I. Sipos, M. Maines, and
via mitogen-activated protein kinase pathway. Am. J. Respir. Cell Mol. Biol.
R. Buelow. 1998. Stress protein-induced immunosuppression: inhibition of cel-
27:603.

Downloaded from http://www.jimmunol.org/ by guest on December 27, 2016


lular immune effector functions following overexpression of haem oxygenase
22. Otterbein, L. E., F. H. Bach, J. Alam, M. Soares, H. Tao Lu, M. Wysk,
(HSP 32). Transplant Immunol. 6:84.
R. J. Davis, R. A. Flavell, and A. M. Choi. 2000. Carbon monoxide has anti-
inflammatory effects involving the mitogen-activated protein kinase pathway. 31. Liu, Y., B. Zhu, L. Luo, P. Li, D. W. Paty, and M. S. Cynader. 2001. Heme
Nat. Med. 6:422. oxygenase-1 plays an important protective role in experimental autoimmune en-
23. Chow, S., H. Patel, and D. W. Hedley. 2001. Measurement of MAP kinase ac- cephalomyelitis. NeuroReport. 12:1841.
tivation by flow cytometry using phospho-specific antibodies to MEK and ERK: 32. Boussiotis, V. A., G. J. Freeman, A. Berezovskaya, D. L. Barber, and
potential for pharmacodynamic monitoring of signal transduction inhibitors. Cy- L. M. Nadler. 1997. Maintenance of human T cell anergy: blocking of IL-2 gene
tometry 46:72. transcription by activated Rap1. Science 278:124.

You might also like