18.06.2013 Views

Glycodelin-A interferes with IL-2/IL-2R signalling to induce cell ...

Glycodelin-A interferes with IL-2/IL-2R signalling to induce cell ...

Glycodelin-A interferes with IL-2/IL-2R signalling to induce cell ...

SHOW MORE
SHOW LESS

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

Human Reproduction, Vol.0, No.0 pp. 1–11, 2012<br />

doi:10.1093/humrep/der477<br />

ORIGINAL ARTICLE Early pregnancy<br />

<strong>Glycodelin</strong>-A <strong>interferes</strong> <strong>with</strong> <strong>IL</strong>-2/<strong>IL</strong>-<strong>2R</strong><br />

<strong>signalling</strong> <strong>to</strong> <strong>induce</strong> <strong>cell</strong> growth arrest,<br />

loss of effec<strong>to</strong>r functions and apop<strong>to</strong>sis<br />

in T-lymphocytes<br />

Chetna Soni and Anjali A. Karande*<br />

Department of Biochemistry, Indian Institute of Science, Bangalore 560012, India<br />

*Correspondence address. Tel: +91-80-2293-2306; Fax: +91-80-2360-0814; E-mail: anjali@biochem.iisc.ernet.in<br />

Submitted on September 5, 2011; resubmitted on November 14, 2011; accepted on December 19, 2011<br />

background: The progesterone-regulated glycoprotein glycodelin-A (GdA), secreted by the decidualized endometrium at high concentrations<br />

in primates, inhibits the maternal immune response against fetal antigens and thereby contributes <strong>to</strong> the <strong>to</strong>lerance of the semiallogenic<br />

fetus during a normal pregnancy. Our earlier studies demonstrated the ability of GdA <strong>to</strong> <strong>induce</strong> an intrinsic apop<strong>to</strong>tic cascade in<br />

CD4 + T-lymphocytes and suppress the cy<strong>to</strong>lytic effec<strong>to</strong>r function of CD8 + T-lymphocytes. In this report, we investigated further in<strong>to</strong> the<br />

mechanism of action of GdA controlling perforin and granzyme B expression in CD8 + T-lymphocytes and the mechanism of action of GdA<br />

leading <strong>to</strong> lymphocyte death.<br />

methods: Flow cy<strong>to</strong>metry analysis was performed <strong>to</strong> check for the surface expression of interleukin-2 recep<strong>to</strong>r a (<strong>IL</strong>-<strong>2R</strong>a) and intra<strong>cell</strong>ular<br />

eomesodermin (Eomes) in activated T-lymphocytes, whereas quantitative RT–PCR analysis was used <strong>to</strong> find out their mRNA profile<br />

upon GdA treatment. Western analysis was carried out <strong>to</strong> confirm the protein level of Bax and Bcl-2.<br />

results: GdA reduces the surface expression of the high-affinity <strong>IL</strong>-<strong>2R</strong> complex by down-regulating the synthesis of <strong>IL</strong>-<strong>2R</strong>a (CD25). This<br />

disturbs the optimal <strong>IL</strong>-2 <strong>signalling</strong> and decreases the Eomes expression, which along <strong>with</strong> <strong>IL</strong>-2 directly regulates perforin and granzymes<br />

expression. Consequently, the CD8 + T-lymphocytes undergo growth arrest and are unable <strong>to</strong> mature in<strong>to</strong> competent cy<strong>to</strong><strong>to</strong>xic<br />

T-lymphocytes. In the CD4 + T-lymphocytes, growth fac<strong>to</strong>r <strong>IL</strong>-2 deprivation leads <strong>to</strong> proliferation inhibition, decreased Bcl-2/enhanced<br />

Bax expression, culminating in mi<strong>to</strong>chondrial stress and <strong>cell</strong> death.<br />

conclusions: GdA spurs <strong>cell</strong> cycle arrest, loss of effec<strong>to</strong>r functions and apop<strong>to</strong>sis in different T-<strong>cell</strong> subsets by making T-lymphocytes<br />

unable <strong>to</strong> respond <strong>to</strong> <strong>IL</strong>-2.<br />

Key words: glycodelin-A / CD25 / eomesodermin / Bcl-2 / Bax<br />

Introduction<br />

Hum. Reprod. Advance Access published February 7, 2012<br />

The ability of the maternal immune <strong>cell</strong>s <strong>to</strong> <strong>to</strong>lerate the semi-allogenic<br />

fetus is conferred by mechanisms which either suppress fetal allogenicity<br />

or spatio-temporally inhibit maternal immune <strong>cell</strong>s’ growth and<br />

functions (Pearson, 2002; Veenstra van Nieuwenhoven et al., 2003;<br />

Niederkorn, 2006; Trowsdale and Betz, 2006; Terness et al., 2007).<br />

<strong>Glycodelin</strong>-A (GdA) or placental protein 14, a glycoprotein found specifically<br />

in primates, is synthesized by the luminal and glandular epithelium<br />

of the decidua and its concentration in the uterine compartment<br />

reaches up <strong>to</strong> 2.5 mM between 14 and 16 weeks of pregnancy<br />

(Seppala et al., 2002). It is one of the immunomodula<strong>to</strong>ry fac<strong>to</strong>rs<br />

that augment fetal <strong>to</strong>lerance (Bol<strong>to</strong>n et al., 1987; Tulppala et al.,<br />

1995). GdA, a 162 amino acid secreted protein, is spatio-temporally<br />

regulated by progesterone (Joshi, 1983; Taylor et al., 1998) and functions<br />

as a galac<strong>to</strong>se-specific lectin (SundarRaj et al., 2009) <strong>to</strong> bring<br />

about its inhibi<strong>to</strong>ry effects on maternal immune <strong>cell</strong>s which are activated<br />

by the trophoblastic antigens at the fetal–maternal interface<br />

during pregnancy (van Kampen et al., 2001).<br />

The placenta, which is comprised of both fetal and maternal tissues,<br />

is the site of rigorous immune <strong>cell</strong> activity. It contains a diverse population<br />

of maternal immune <strong>cell</strong>s; uterine natural killer (NK) <strong>cell</strong>s, dendritic<br />

<strong>cell</strong>s, T-lymphocytes, monocytes and B <strong>cell</strong>s (Starkey et al., 1988;<br />

Williams et al., 2009). GdA exerts its inhibi<strong>to</strong>ry effect on both innate<br />

and adaptive immune responses (Seppala et al., 2002). Subjects <strong>with</strong><br />

subnormal levels of GdA face unexplained infertility (Mackenna<br />

et al., 1993), habitual abortions (Tulppala et al., 1995) and recurrent<br />

miscarriages (Dal<strong>to</strong>n et al., 1998). GdA inhibits the proliferation of<br />

& The Author 2012. Published by Oxford University Press on behalf of the European Society of Human Reproduction and Embryology. All rights reserved.<br />

For Permissions, please email: journals.permissions@oup.com<br />

Downloaded from<br />

http://humrep.oxfordjournals.org/ by guest on June 17, 2013


2 Soni and Karande<br />

B <strong>cell</strong>s, along <strong>with</strong> reduced immunoglobulin M (IgM) secretion and<br />

major his<strong>to</strong>compatibility complex class II surface expression upon activation<br />

(Yaniv et al., 2003; Alok and Karande, 2009). Experiments<br />

carried out on peripheral NK <strong>cell</strong>s and decidual large granular lymphocytes<br />

confirm the ability of GdA <strong>to</strong> inhibit their cy<strong>to</strong>lytic activity<br />

(Okamo<strong>to</strong> et al., 1991) and <strong>to</strong> enhance interleukin-6 (<strong>IL</strong>-6), <strong>IL</strong>-13<br />

and granulocyte-macrophage colony-stimulating fac<strong>to</strong>r production<br />

from them, signifying its role in trophoblast invasion (Lee et al.,<br />

2010). Survival and proliferation of monocytes are inhibited (Tee<br />

et al., 2008; Alok et al., 2009) and dendritic <strong>cell</strong>s derived from monocytes<br />

acquire a <strong>to</strong>lerogenic phenotype on GdA treatment (Scholz<br />

et al., 2008).<br />

The most extensively studied effect of GdA is that on the<br />

T-lymphocytes. GdA inhibits the proliferation of mi<strong>to</strong>gen stimulated<br />

T-<strong>cell</strong>s (Bol<strong>to</strong>n et al., 1987; Pockley and Bol<strong>to</strong>n, 1989; Rachmilewitz<br />

et al., 1999; Mukhopadhyay et al., 2001). Additionally, it <strong>induce</strong>s apop<strong>to</strong>sis<br />

in activated T-lymphocytes (Mukhopadhyay et al., 2001) and<br />

the protein backbone is necessary and sufficient for this activity<br />

(Jayachandran et al., 2004). Importantly, the N-linked glycosylation<br />

on GdA modulates accessibility of the active site on the protein<br />

(Mukhopadhyay et al., 2004; Jayachandran et al., 2006; Poornima<br />

and Karande, 2007). GdA also shifts the Th1–Th2 cy<strong>to</strong>kine balance<br />

<strong>to</strong>wards Th2 cy<strong>to</strong>kines (Mishan-Eisenberg et al., 2004) by selectively<br />

triggering apop<strong>to</strong>sis in Th1 <strong>cell</strong>s (Lee et al., 2011). Interestingly, like<br />

the Th2 <strong>cell</strong>s, B <strong>cell</strong>s (Alok and Karande, 2009) as well as CD8 + Tlymphocytes<br />

are resistant <strong>to</strong> GdA-<strong>induce</strong>d apop<strong>to</strong>sis (Soni and<br />

Karande, 2010), though the proliferation (SundarRaj et al., 2009)<br />

and cy<strong>to</strong>lytic potential of CD8 + CTLs (cy<strong>to</strong><strong>to</strong>xic T-lymphocytes) are<br />

compromised upon GdA treatment (Soni and Karande, 2010).<br />

Further exploring the regula<strong>to</strong>ry effect of GdA on the effec<strong>to</strong>r functions<br />

of CD8 + T-<strong>cell</strong>s, which predominate the uterine T-lymphocyte<br />

population during pregnancy (Williams et al., 2009), we reported<br />

that GdA exerts its inhibi<strong>to</strong>ry effect on the cy<strong>to</strong>lytic activity of CTLs<br />

by down-modulating the synthesis and release of key cy<strong>to</strong>lytic<br />

fac<strong>to</strong>rs perforin and granzyme B from them (Soni and Karande, 2010).<br />

In this study, we have probed further in<strong>to</strong> the mechanistic details of<br />

the regulation of perforin and granzymes by GdA. In agreement <strong>with</strong><br />

the report by Pockley and Bol<strong>to</strong>n (1989), we find that GdA <strong>induce</strong>s<br />

suboptimal <strong>IL</strong>-2 <strong>signalling</strong>, by down-regulating the surface expression<br />

of an <strong>IL</strong>-<strong>2R</strong>a subunit (CD25), which <strong>to</strong>gether <strong>with</strong> reduced eomesodermin<br />

(Eomes) expression result in<strong>to</strong> abated perforin and granzyme<br />

B contributing <strong>to</strong> the debilitated cy<strong>to</strong>lytic activity of CTLs. Interestingly,<br />

our data suggest that the aberrant <strong>IL</strong>-2 <strong>signalling</strong> in the presence of<br />

GdA translates in<strong>to</strong> a dis-balance between the pro- and anti-apop<strong>to</strong>tic<br />

proteins <strong>with</strong>in T-<strong>cell</strong>s. Thus, data reported in this study indicate that<br />

growth fac<strong>to</strong>r depletion may be a potential mechanism not only for<br />

the inhibition of proliferation but also for apop<strong>to</strong>sis and loss of effec<strong>to</strong>r<br />

functions <strong>induce</strong>d by GdA in T-lymphocytes at the fe<strong>to</strong>-maternal<br />

interface.<br />

Materials and Methods<br />

Cells and <strong>cell</strong> lines<br />

Peripheral blood mononuclear <strong>cell</strong>s (PBMCs) were isolated using ‘Ficoll’<br />

density gradient centrifugation (His<strong>to</strong>paque, Sigma-Aldrich, MO, USA),<br />

from fresh whole blood of normal healthy donors (males and non-pregnant<br />

females of the age group 25–30 years; Boyum, 1964). PBMCs were cultured<br />

in Roswell Park Memorial Institute (RPMI)-1640 medium (Sigma-Aldrich)<br />

supplemented <strong>with</strong> 10% fetal bovine serum (FBS; Gibco, NY, USA),<br />

100 U/ml penicillin, 100 mg/ml strep<strong>to</strong>mycin and 2 mM L-glutamine,<br />

unless mentioned otherwise. Ovarian carcinoma <strong>cell</strong> line OVCAR-3<br />

(obtained from Prof. J. Hilgers, Vrije University, Amsterdam) was cultured<br />

in Delbecco’s modified Eagle’s medium (Sigma-Aldrich) supplemented<br />

<strong>with</strong> 10% FBS, 2 mM L-glutamine and antibiotics.<br />

Antibodies and reagents<br />

Anti-CD4 and CD8 monoclonal antibodies (mAbs) were purified from<br />

culture supernatants of OKT4 and OKT8 hybridoma <strong>cell</strong>s [National Facility<br />

for Animal Tissue and Cell Culture (NFATCC), Pune, India] cultured in<br />

Iscove’s modified Dulbecco’s medium (IMDM) <strong>with</strong> 10% FBS, using<br />

protein-A affinity chroma<strong>to</strong>graphy. Purified Abs were labelled <strong>with</strong> FITC<br />

using the standard pro<strong>to</strong>cols (Bioconjugate Techniques by Greg<br />

T. Hermanson, second edition). Anti-Eomes Alexa Fluor R 647, anti-human<br />

CD25-PE, anti-human Bcl-2 and anti-human Bax antibodies were purchased<br />

from eBiosciences (USA). Anti-human beta-actin horse-radish peroxidase<br />

(HRP) conjugate, anti-human interferon (IFN)-g-PE conjugate, HISTOPA-<br />

QUE, PHA (phy<strong>to</strong>haemagglutinin) and RNA isolation reagent (TRI reagent)<br />

was purchased from Sigma-Aldrich. Rabbit anti-mouse HRP-conjugated antibody<br />

was procured from Dako (Denmark). RevertAid reverse transcriptase,<br />

Oligo-dT and dNTP mix was obtained from Thermo Fisher Scientific<br />

(Canada). For real-time PCR analysis, iQ SYBR Green sensimix was procured<br />

from Bio-Rad (CA, USA). CD4 + and CD8 + T-<strong>cell</strong>s were isolated using<br />

magnetic-activated <strong>cell</strong> sorting (MACS—Meltenyi Biotec, Germany), by the<br />

MS-MACS columns using streptavidin–phycoerythrin microbeads.<br />

Purification of GdA<br />

GdA was purified from amniotic fluid by immunoaffinity chroma<strong>to</strong>graphy,<br />

using a Gd-specific mAb, D9D4 using the standard lab pro<strong>to</strong>col (Mukhopadhyay<br />

et al., 2001). Briefly, GdA was eluted <strong>with</strong> 100 mM gly–HCl, pH<br />

2.5, and neutralized <strong>with</strong> 1 M Tris–HCl, pH 8.0, thereafter dialysed against<br />

50 mM phosphate-buffered saline (PBS). The purity of the protein was<br />

determined using sodium dodecyl sulphate–polyacrylamide gel electrophoresis<br />

(SDS–PAGE) followed by silver staining and specificity ascertained<br />

by western blotting. The protein preparations were checked for<br />

LPS (lipopolysaccharide) contamination using E-Toxate, LPS detection kit<br />

(Sigma-Aldrich). Protein preparations free of LPS contamination were<br />

used in all the assays. Also, the activity of GdA was ascertained by <strong>cell</strong> proliferation<br />

and apop<strong>to</strong>sis assays (Soni and Karande, 2010). One of the most<br />

abundant proteins of the amniotic fluid, namely human serum albumin<br />

(HSA), was purified from amniotic fluid (Mukhopadhyay et al., 2001)<br />

and has been used as the negative control throughout the study.<br />

Alloactivation and T-<strong>cell</strong> stimulation<br />

To activate PBMCs and <strong>to</strong> generate CTLs in vitro, a previously standardized<br />

method of alloactivation was followed (Soni and Karande, 2010). Briefly,<br />

PBMCs (responders) were alloactivated, using gOVCAR-3 (OVCAR-3<br />

<strong>cell</strong>s irradiated <strong>with</strong> 30 Gy of gamma rays using the BI-2000 gamma irradia<strong>to</strong>r),<br />

as stimula<strong>to</strong>rs. Stimula<strong>to</strong>rs were co-cultured <strong>with</strong> responders in a<br />

ratio of 1:10 in IMDM supplemented <strong>with</strong> 10% FBS, 10% normal human<br />

serum, 2 mM sodium pyruvate and 2 mM L-glutamine in 60 mm culture<br />

grade Petri plates, at a <strong>cell</strong> density of 6 × 10 6 /ml for 96 h at 378C in<br />

5% CO 2. About 5 U/ml of recombinant <strong>IL</strong>-2 (Sigma-Aldrich) was supplemented<br />

at 0 and 48 h of the co-culture. Depending on the requirement of<br />

the experiment, the alloactivation was either terminated after 18 h of<br />

co-culture or continued for 96 h or both and has been mentioned wherever<br />

necessary. Cells were treated <strong>with</strong> GdA for 24 h post-alloactivation.<br />

Alternatively, purified PBMCs (2.5 × 10 6 <strong>cell</strong>s/ml) were activated <strong>with</strong><br />

Downloaded from<br />

http://humrep.oxfordjournals.org/ by guest on June 17, 2013


<strong>Glycodelin</strong>-A dampens CD25 expression in T-<strong>cell</strong>s 3<br />

5 mg/ml of PHA for 18 h (or as mentioned) in RPMI supplemented <strong>with</strong><br />

10% FBS. PHA-stimulated <strong>cell</strong>s were simultaneously treated <strong>with</strong> GdA<br />

unless mentioned otherwise.<br />

Cell surface/intra<strong>cell</strong>ular staining and flow<br />

cy<strong>to</strong>metry<br />

For <strong>cell</strong> surface staining, activated <strong>cell</strong>s (1 × 10 6 ) were stained <strong>with</strong><br />

anti-CD4-FITC/CD8-FITC and/CD25-PE direct conjugates (as indicated)<br />

or anti-CD28 for 1 h on ice. Direct conjugates were washed twice <strong>with</strong><br />

fluorescence-activated <strong>cell</strong> sorting (FACS) wash buffer [0.2% bovine<br />

serum albumin (BSA) (w/v) and 0.1% (w/v) sodium azide in 50 mM<br />

PBS], fixed <strong>with</strong> 2% paraformaldehyde and s<strong>to</strong>red at 48C until acquisition<br />

on FACSCalibur/Can<strong>to</strong> II flow cy<strong>to</strong>meter (Bec<strong>to</strong>n Dickenson, NJ, USA).<br />

Acquired data were analysed using WinMDI 2.9 software/Cell Quest<br />

Pro and plotted using Graph Pad Prism Ver 5.0. Anti-CD28 surfacelabelled<br />

<strong>cell</strong>s were washed <strong>with</strong> FACS wash buffer and then probed<br />

<strong>with</strong> Rabbit-anti-mouse FITC conjugate (Dako), washed <strong>to</strong> remove<br />

unbound secondary antibody and then fixed <strong>with</strong> 2% paraformaldehyde.<br />

Data were acquired and analysed as aforementioned. In all cases, at<br />

least 10 000 events were acquired per sample for analysis.<br />

Intra<strong>cell</strong>ular staining: for staining secreted proteins like IFN-g, PBMCs<br />

were cultured <strong>with</strong> 2 mM monensin (Sigma-Aldrich) for the last 12 h<br />

during the 18-h alloactivation and also during 24 h of GdA treatment.<br />

Thereafter, the same pro<strong>to</strong>col was followed for secreted or intra<strong>cell</strong>ular<br />

proteins. Cells (up <strong>to</strong> 1 × 10 6 ) were surface stained <strong>with</strong> desired antibodies<br />

against various surface markers, fixed <strong>with</strong> 2% paraformaldehyde<br />

(pHCHO), for 15 min at RT, washed <strong>to</strong> remove pHCHO followed by permeabilization<br />

<strong>with</strong> 1 ml of 90% methanol in 50 mM PBS (pH 7.2) for<br />

30 min on ice. Cells were then centrifuged at 1957 g for 15 min <strong>to</strong><br />

remove methanol and washed twice <strong>with</strong> FACS wash buffer, resuspended<br />

in 100 ml of FACS wash buffer and incubated <strong>with</strong> either anti-IFN-g-PE or<br />

anti-Eomes-Alexa Fluor for 1 h on ice. Washed <strong>to</strong> remove the excess antibody<br />

and s<strong>to</strong>red at 48C until acquisition by FACS Can<strong>to</strong> II (Bec<strong>to</strong>n<br />

Dickenson). Data were analysed using BD FACSDiva 6.0/Cell Quest<br />

Pro software. At least 20 000 events were acquired per sample.<br />

RNA preparation and real-time quantitative<br />

RT–PCR<br />

Total RNA was isolated using TRI reagent (Sigma-Aldrich) from unactivated<br />

PBMCs, alloactivated PBMCs (18 or 96 h) treated <strong>with</strong> control<br />

protein (HSA) and alloactivated GdA-treated (24 h) PBMCs. 2.5 mg of<br />

<strong>to</strong>tal RNA was reverse transcribed in a vol of 20 ml using random hexamers<br />

(olig-dT) and RevertAid M-MuLV reverse transcriptase as per the manufacturer’s<br />

instructions. Real-time quantitative PCR was performed using<br />

the DNA-binding dye, the SYBR Green method (Morrison et al., 1998).<br />

Reaction was carried out in triplicates in 20 ml on a BioRad iCycler iQ5.<br />

The primers used for real-time PCR are listed in Table I. The amplification<br />

conditions for housekeeping and test genes were: initial denaturation at<br />

958C for 3 min followed by 40 cycles of denaturation at 948C for 10 s,<br />

primer annealing at 638C for 30 s and extension at 728C for 45 s. The<br />

final extension was carried out at 728C for 5 min. The fluorescence<br />

emitted at each cycle was collected for the entire period of 45 s during<br />

the extension step of each cycle. The homogeneity/specificity of the<br />

PCR amplicons was verified by running 2% agarose gels and also by melt<br />

curve analysis. Mean Ct values generated in each experiment using the<br />

iCycler software (Bio-Rad) were used <strong>to</strong> calculate the relative fold change<br />

in gene expression upon activation and GdA treatment. Briefly, data for<br />

the test genes were normalized <strong>with</strong> reference gene (h18S rRNA) and calculated<br />

manually as the fold change in test gene expression above control (unactivated<br />

PBMCs) by the 2 2△△Ct method. Final fold change in gene expression<br />

Table I List of primers for real time RT-PCR.<br />

S.<br />

No.<br />

Gene Primer sequence<br />

........................................................................................<br />

1 h18s rRNA F: 5 ′ -CGC CGC TAG AGG TGA AAT TC-3 ′<br />

R: 5 ′ -TTG GCA AAT GCT TTC GCT C-3 ′<br />

2 hEOMES F: 5 ′ -GGG GAG GTC GAG GTT CTT ACC AGA-3 ′<br />

R: 5 ′ -CTT GAA CAC AGT GGG GCT TGT TCT-3 ′<br />

3 hT-Bet F: 5 ′ -GCC TGG GGT CTC CCT ACC CG-3 ′<br />

R: 5 ′ -GGC TCC AAG GAA GCG GCT C-3 ′<br />

4 hCD25 F: 5 ′ -TCG GCC TGG AGT GGT GTG TC-3 ′<br />

R: 5 ′ -GGG CCC CTC CTT TTG GGG GA-3 ′<br />

5 hBcl-2 F: 5 ′ -CCT GTC GAT GAC TGA GTA CC-3 ′<br />

R: 5 ′ -GAG ACA GCC AGG AGA AAT CA-3 ′<br />

6 hBax F: 5 ′ -ATG CGT CCA CCA AGA AGC TGA GC-3 ′<br />

R: 5 ′ -CCC CGA TGC GCT TGA GAC ACT-3 ′<br />

7 hCD28 F: 5 ′ -AGC AGG CTC CTG CAC AGT GAC-3 ′<br />

R: 5 ′ -TAG GCT GCG AAG TCG CGT GG-3 ′<br />

8 hCTLA4 F: 5 ′ -GCA TCG CCA GCT TTG TGT GTG AGT-3 ′<br />

R: 5 ′ -TCC GAAGCA CTG TCA CCC GGA CC-3 ′<br />

was plotted using Graph Pad Prism 5.0 software. Statistical analysis was performed<br />

using the paired Student’s t-test.<br />

Cell lysate preparation and immunoblotting<br />

Resting or PHA-activated PBMCs, treated <strong>with</strong> control protein or GdA,<br />

were washed <strong>with</strong> 50 mM PBS and pelleted at 2000 rpm/5 min. Cell<br />

pellets were blotted <strong>to</strong> remove any remaining PBS. Pellets were tapped<br />

and <strong>cell</strong>s lysed for 30 min on ice in RIPA buffer (100 ml for 1 × 10 7<br />

PBMCs) consisting of 50 mM Tris–HCl (pH 7.4), 1% Nonidet-P-40,<br />

0.25% sodium deoxycholate, 150 mM NaCl, 1 mM EDTA, 1 mM phenylmethylsulphonyl<br />

fluoride, 1 mg/ml each of aprotinin, leupeptin and pepstatin<br />

(added freshly), 1 mM Na3VO 4 and 1 mM NaF. Samples were then<br />

vigorously vortexed and centrifuged at 14 000 rpm for 10 min at 48C.<br />

Pellet was discarded and protein estimated using Bradford’s method of<br />

protein estimation. Equal protein from each sample was boiled in SDS<br />

sample buffer and subjected <strong>to</strong> SDS–PAGE along <strong>with</strong> a molecular<br />

weight marker. After equilibration in transfer buffer (25 mM Tris-base,<br />

192 mM glycine and 20% methanol) gels were transferred on<strong>to</strong> nitro<strong>cell</strong>ulose<br />

membrane (Millipore, MA, USA) by semi-dry western blotting<br />

(Bio-Rad). Non-specific binding was blocked <strong>with</strong> 5% non-fat dry milk<br />

powder in Tris-buffered saline Tween-20 (TBS-T; 20 mM Tris–HCl, pH<br />

7.4, 137 mM NaCl and 0.1% Tween-20) for 2–3 h at 48C <strong>with</strong> constant<br />

rocking. The blots were thereafter incubated overnight <strong>with</strong> constant<br />

rocking at 48C, <strong>with</strong> primary antibodies diluted in TBS-T <strong>with</strong> 5% BSA.<br />

Thereafter, blots were thoroughly washed <strong>with</strong> TBS-T and then incubated<br />

<strong>with</strong> anti-mouse IgG secondary antibodies conjugated <strong>with</strong> HRP for 1 h at<br />

48C. After further washing in TBS-T, the immunoblots were developed<br />

<strong>with</strong> an enhanced chemiluminiscence detection system (Millipore) as per<br />

instructions. Data acquired using Fujifilm Las 3000 Imager and densi<strong>to</strong>metric<br />

analysis was done using Fujifilm Multigauge software.<br />

Statistical analysis<br />

All the data were evaluated using the paired Student’s t-test. P-value of<br />

,0.05 was considered as statistically significant.<br />

Downloaded from<br />

http://humrep.oxfordjournals.org/ by guest on June 17, 2013


4 Soni and Karande<br />

Results<br />

Activated PBMCs upon GdA treatment<br />

express lower level of Eomes<br />

In our previous study, we reported that GdA suppresses the cy<strong>to</strong>lytic<br />

activity of CTLs generated in vitro by impeding the transcription of<br />

cy<strong>to</strong>lytic genes viz. perforin and granzyme B (Soni and Karande,<br />

2010). We went further <strong>to</strong> find out how GdA controls the expression<br />

of perforin and granzyme B. T-box transcription fac<strong>to</strong>rs T-Bet and<br />

Eomes <strong>induce</strong>d upon the CD8 + <strong>cell</strong> activation act as key regula<strong>to</strong>ry<br />

proteins in the development of competent effec<strong>to</strong>r and memory<br />

CD8 + T-<strong>cell</strong>s (Szabo et al., 2000, 2002; Pearce et al., 2003; Araki<br />

et al., 2008). To test the possibility, if GdA was able <strong>to</strong> downmodulate<br />

the cy<strong>to</strong>lytic effec<strong>to</strong>r genes by affecting the induction of<br />

Eomes and T-bet, we ascertained the transcriptional status of T-Bet<br />

and Eomes by real-time RT–PCR analysis.<br />

PBMCs alloactivated <strong>with</strong> gOVCAR-3 for 18 or 96 h were treated<br />

<strong>with</strong> 1 mM GdA for 24 h (which is the time required for GdA <strong>to</strong> have<br />

its inhibi<strong>to</strong>ry effect on CTLs). Two time points were chosen so as <strong>to</strong><br />

pick the message at the time of its optimal synthesis. RNA was isolated<br />

and real-time RT–PCR was performed. Figure 1a and b depicts the<br />

expression of T-Bet and Eomes mRNA, respectively, in alloactivated<br />

PBMCs, expressed as fold change relative <strong>to</strong> the unactivated<br />

PBMCs. As is evident from the graph, there was no significant<br />

change in T-bet mRNA expression upon GdA treatment, but<br />

Figure 1 GdA does not affect T-bet but down-modulates Eomes expression. Quantitative RT–PCR of the expression of T-Bet (a) and Eomes (b)in<br />

18 or 96 h alloactivated PBMCs subsequently treated <strong>with</strong> GdA/HSA (Ctrl) for 24 h. Data normalized <strong>to</strong> 18 s rRNA and presented as fold change<br />

relative <strong>to</strong> the gene expression in unstimulated PBMCs. Box plot shows median, interquartile range and range of four observations. (c) Flow cy<strong>to</strong>metry<br />

of the expression of Eomes in; unstimulated PBMCs (UA); alloactivated PBMCs treated <strong>with</strong> HSA [A (Ctrl)] and alloactivated PBMCs treated <strong>with</strong><br />

GdA [A (GdA)]. Unstained PBMCs (US) were used <strong>to</strong> define the P3 region. Numbers <strong>with</strong>in the region P3 indicate the percent Eomes positive<br />

<strong>cell</strong>s among <strong>to</strong>tal PBMCs. At least 20 000 events were acquired per sample. Lymphocytes were gated on FSC/SSC. Data are representative of<br />

five independent experiments. (d) Percentage of Eomes positive unactivated or activated PBMCs upon Ctrl or GdA treatment were quantified<br />

using WinMDI and plotted using Graph Pad Prism 5.0, Box plot shows median, interquartile range and range of five observations *P ¼ 0.0139 and<br />

**P ¼ 0.0171.<br />

Downloaded from<br />

http://humrep.oxfordjournals.org/ by guest on June 17, 2013


<strong>Glycodelin</strong>-A dampens CD25 expression in T-<strong>cell</strong>s 5<br />

transcription of Eomes was significantly impeded by GdA (P ¼ 0.03).<br />

The result prompted us <strong>to</strong> look at the intra<strong>cell</strong>ular protein profile of<br />

Eomes. Unstimulated PBMCs, alloactivated PBMCs (24 h) followed<br />

by control/1 mM GdA treatment for 24 h, were stained for Eomes.<br />

Figure 1c shows representative his<strong>to</strong>grams <strong>with</strong> the expression level<br />

of Eomes in resting and alloactivated PBMCs. Figure 1d depicts the<br />

% Eomes positive <strong>cell</strong>s from five independent experiments. As is<br />

evident from the plots, GdA treatment reduced the intra<strong>cell</strong>ular<br />

protein level of Eomes. Interestingly, we did not observe any change<br />

in the intra<strong>cell</strong>ular IFN-g protein expression in activated CD8 +<br />

T-<strong>cell</strong>s upon GdA treatment (Supplementary data, Fig. SI).<br />

GdA dampens CD25 expression in<br />

T-lymphocytes<br />

We observed in stimulated CD8 + T-<strong>cell</strong>s that upon GdA treatment,<br />

they were cy<strong>to</strong>lytically disabled and expressed relatively low levels<br />

of perforin and granzyme B (Soni and Karande, 2010). Furthermore,<br />

our observation that upon GdA treatment, Eomes expression in activated<br />

T-<strong>cell</strong>s was reduced (Fig. 1) indicated that GdA <strong>induce</strong>d a lowperforin,<br />

low-granzyme, low-Eomes profile, which was indicative of<br />

weak <strong>IL</strong>-2 <strong>signalling</strong> (Belz and Masson, 2010). Most importantly, very<br />

early in glycodelin research, it was reported that GdA suppresses <strong>IL</strong>-2<br />

synthesis in mi<strong>to</strong>gen-stimulated T-<strong>cell</strong>s and that exogenous supplementation<br />

of <strong>IL</strong>-2 can rescue T-<strong>cell</strong>s from the GdA-<strong>induce</strong>d inhibition of<br />

proliferation (Pockley and Bol<strong>to</strong>n, 1989) which was further confirmed<br />

by a recent study (Lee et al., 2009). Building up on the available information,<br />

we looked at the mRNA level (at an early and late time point of<br />

activation) and surface expression of CD25 in stimulated PBMCs.<br />

As expected, we observed a significant decrease in CD25 mRNA<br />

expression in PBMCs alloactivated for 18 and 96 h (Fig. 2a), plate<br />

bound anti-CD3-activated and PHA-activated PBMCs (data not<br />

shown) upon treatment for 24 h <strong>with</strong> 1 mM GdA. Figure 2b shows<br />

the kinetics of CD25 surface expression upon PHA stimulation. Its expression<br />

started at around 6 h post-stimulation, peaked at around 18 h<br />

(at 24–30 h in alloactivated PBMCs). Clearly a CD25 hi/bright and a<br />

CD25 lo/dim population could be seen, as indicated in the figure. The<br />

expression was fairly sustained until 48 h of stimulation.<br />

To ascertain the effect of GdA on CD25 surface expression,<br />

PBMCs were stimulated <strong>with</strong> PHA for 24 h and simultaneously<br />

treated <strong>with</strong> GdA and surface stained for CD4/CD8 and CD4/<br />

CD25. As can be seen in the representative dot plot in Fig. 2c,<br />

GdA treatment resulted in a modest decrease in surface CD25 hi<br />

population, <strong>with</strong> a concomitant increase in CD25 lo and CD25 neg<br />

T-<strong>cell</strong>s. The type of stimulation did not alter the result, only the percentage<br />

of <strong>cell</strong>s expressing CD25 was higher upon PHA stimulation<br />

than upon alloactivation(data not shown). Data from six such independent<br />

experiments have been compiled and presented in Fig. 2d.<br />

There was ≏41.5% reduction in CD25 hi CD4 + T-<strong>cell</strong>s, whereas a<br />

≏32.3% reduction in CD25 hi CD8 + T-<strong>cell</strong>s was observed in GdAtreated<br />

samples.<br />

GdA does not affect the expression<br />

of CD28 or CTLA4<br />

CD28 and CTLA4 (CTL-associated protein 4) are immunoglobulin<br />

superfamily proteins expressed on the T-<strong>cell</strong> surface and are ligands<br />

for the B7 molecules (B7-1 and B7-2), which are constitutively<br />

expressed on antigen presenting <strong>cell</strong>s and act antagonistically. CD28<br />

(present on resting and activated <strong>cell</strong>s) delivers a positive<br />

co-stimula<strong>to</strong>ry signal, while <strong>signalling</strong> through CTLA4 (present only<br />

on activated T-<strong>cell</strong>s) is inhibi<strong>to</strong>ry. We reasoned that GdA might<br />

exert its inhibi<strong>to</strong>ry effect on T-<strong>cell</strong>s by affecting CD28 or CTLA4<br />

levels; hence, we looked at the mRNA and protein profile of CD28<br />

and the mRNA expression of CTLA4 in 18 or 96 h alloactivated<br />

PBMCs treated <strong>with</strong> GdA for 24 h. We did not observe any significant<br />

variation in the CD28 mRNA/protein upon GdA treatment (Supplementary<br />

data, Fig. S2a and b) Similarly, CTLA4 mRNA expression was<br />

also found <strong>to</strong> be unaffected by GdA (Supplementary data, Fig. S3).<br />

GdA signals <strong>to</strong> down-regulate Bcl-2 and<br />

up-regulate Bax in activated PBMCs<br />

Three distinct <strong>IL</strong>-<strong>2R</strong> <strong>signalling</strong> pathways are reported <strong>to</strong> exist,<br />

mediated by c-myc, lck and Bcl-2 (Miyazaki et al., 1995). Resting<br />

T-<strong>cell</strong>s gain metabolic activity following the recognition of cognate<br />

Ag by the T <strong>cell</strong> recep<strong>to</strong>r (TCR); yet, they do not progress <strong>to</strong><br />

S-phase and instead undergo apop<strong>to</strong>sis, unless additionally stimulated<br />

by <strong>IL</strong>-2 or other mi<strong>to</strong>genic cy<strong>to</strong>kines (Akbar et al., 1996; Van Parijs<br />

et al., 1997). Bcl-2 and Bcl-x have been proposed <strong>to</strong> augment <strong>cell</strong> viability<br />

because constitutive expression of these genes in cy<strong>to</strong>kinedependent<br />

<strong>cell</strong> lines significantly delays the onset of the apop<strong>to</strong>tic<br />

program <strong>induce</strong>d by growth fac<strong>to</strong>r deprivation (Deng and Podack,<br />

1993).<br />

In the light of the above literature, we probed in<strong>to</strong> the mRNA and<br />

protein expression status of important anti-apop<strong>to</strong>tic protein Bcl-2 in<br />

activated PBMCs upon GdA treatment. Supplementary data, Fig. S4<br />

shows the fold induction of Bcl-2 mRNA in control-treated and GdAtreated<br />

alloactivated (18/96 h) PBMCs. There was a modest downregulation<br />

of Bcl-2 mRNA upon GdA treatment; however it did not<br />

reach statistical significance (n ¼ 6). Interestingly though, the assessment<br />

of <strong>to</strong>tal Bcl-2 protein levels from the <strong>cell</strong> lysates prepared<br />

after PHA activation and simultaneous GdA treatment of PBMCs for<br />

24 h showed a moderate but a significant decrease (n ¼ 6). Essentially,<br />

the modest increase seen in Bcl-2 expression upon activation was<br />

abrogated in the presence of GdA and the Bcl-2 levels were similar<br />

<strong>to</strong> the resting PBMCs (Fig. 3a and b).<br />

Bcl-2 and Bax proteins are known <strong>to</strong> heterodimerize that prevents<br />

Bax oligomerization and subsequent permeabilization of the outer<br />

mi<strong>to</strong>chondrial membrane by Bax (Oltvai et al., 1993; Hengartner,<br />

2000). Hence, we went ahead <strong>to</strong> check for the expression profile of<br />

the pro-apop<strong>to</strong>tic protein Bax in alloactivated PBMCs upon GdA<br />

treatment. For RNA analysis, PBMCs were allostimulated for 18 h,<br />

treated <strong>with</strong> GdA for 24 h. For protein assessment, PBMCs were<br />

PHA activated along <strong>with</strong> GdA for 24 h and processed for <strong>to</strong>tal<br />

protein extraction. RNA analysis by real-time RT–PCR revealed an<br />

almost 5-fold up-regulation in Bax mRNA in the presence of GdA<br />

(n ¼ 5; Fig. 4a). In concurrence, there was a significant increase in<br />

the <strong>to</strong>tal Bax protein upon GdA treatment of PHA-stimulated<br />

PBMCs (n ¼ 4; Fig. 4b and c).<br />

Discussion<br />

Many studies have put forward mechanisms <strong>to</strong> explain the inhibi<strong>to</strong>ry<br />

effect of GdA on T-lymphocytes. Rachmilewitz et al. show that the<br />

Downloaded from<br />

http://humrep.oxfordjournals.org/ by guest on June 17, 2013


6 Soni and Karande<br />

Figure 2 GdA treatment reduces the mRNA surface expression of CD25 in T-lymphocytes. (a). Total RNA was extracted and cDNA synthesized<br />

from resting PBMCs or PBMCs activated <strong>with</strong> gOVCAR-3 <strong>cell</strong>s for 18 or 96 h followed by treatment <strong>with</strong> HSA (Ctrl) or GdA for 24 h. 18S rRNA and<br />

CD25 mRNA levels were determined by real-time quantitative RT–PCR. Result is expressed as fold change in CD25 mRNA in 18 or 96 h<br />

alloactivated-GdA/Ctrl-treated PBMCs, when compared <strong>with</strong> unactivated PBMCs and normalized <strong>with</strong> 18s rRNA transcription. Samples were run<br />

in triplicates. Box plot show median, interquartile range and range of six observations. (b) Flow cy<strong>to</strong>metric analysis of CD25 surface expression<br />

on resting (i) and PHA stimulated PBMCs at various time points indicated (ii–v). Panel vi represents dot plot for CD4 and CD25 double-stained<br />

PBMCs after 18 h of PHA activation. Regions Q1 and Q2 represent CD25 hi population, Q3 and Q4 represent CD25 lo population and Q5 and<br />

Q6 are CD25 neg . Numbers in i–v represent the percentage of CD25 hi and CD25 lo <strong>cell</strong>s in the respective dot plot. (c) Resting PBMCs or PBMCs<br />

activated using 5 mg/ml of PHA and simultaneously treated <strong>with</strong> HSA (Ctrl) or GdA were harvested; stained for surface CD4/CD8/CD25 and<br />

data acquired using flow cy<strong>to</strong>metry. Upper three panels show representative two colour dot plots showing FACS analysis of PBMCs stained for<br />

CD4 and CD25; lower three panels show staining <strong>with</strong> CD8/CD25 Abs. CD25-PE acquired in FL-2 channel and CD4/CD8 in FL-1 channel. At<br />

least 10 000 events acquired <strong>with</strong>in the lymphocyte gate. Data analysed using winMDI software, Ver.5.0. (d) Percentage of CD4 + /CD8 + T-<strong>cell</strong>s<br />

<strong>with</strong> CD25 lo or CD25 hi expression upon Ctrl or GdA treatment were quantified using WinMDI and plotted using Graph Pad Prism 5.0. Data<br />

expressed as box plots showing median, interquartile range and range from four independent experiments. *P ¼ 0.0128, **P ¼ 0.0261,<br />

***P ¼ 0.0026 and ****P ¼ 0.007.<br />

inhibi<strong>to</strong>ry function of GdA is dependent on its localization <strong>to</strong> the phosphatase<br />

CD45 at the site of TCR triggering, where it negatively<br />

regulates T-<strong>cell</strong> activation by reducing the half-life of TCR-triggered<br />

phosphotyrosines (Rachmilewitz et al., 2001, 2003). Delineating<br />

the mechanism of induction of apop<strong>to</strong>sis in T-lymphocytes,<br />

SundarRaj et al. (2008) provide compelling evidences of a<br />

TCR-independent pathway, wherein GdA <strong>induce</strong>s a stress signal<br />

which culminates in<strong>to</strong> mi<strong>to</strong>chondrial membrane potential loss and permeabilization,<br />

causing T-lymphocytes <strong>to</strong> apop<strong>to</strong>se via the intrinsic<br />

mi<strong>to</strong>chondrial pathway. Corroborating these data, overexpression of<br />

Downloaded from<br />

http://humrep.oxfordjournals.org/ by guest on June 17, 2013


<strong>Glycodelin</strong>-A dampens CD25 expression in T-<strong>cell</strong>s 7<br />

Figure 3 GdA inhibits the up-regulation of Bcl-2 expression in activated<br />

PBMCs. (a) Immunoblot analysis of test protein Bcl-2 and reference<br />

protein b-actin in resting PBMCs and PBMCs activated for<br />

24 h <strong>with</strong> 5 mg/ml of PHA <strong>with</strong> simultaneous HSA (Ctrl) treatment<br />

or GdA treatment. Proteins detected <strong>with</strong> anti-human Bcl-2 Ab and<br />

anti-b-actin HRP in whole <strong>cell</strong> lysates. Data are representative of<br />

five independent experiments. (b) Quantification of Bcl-2 protein expression.<br />

Bcl-2 protein expression in PHA-activated (HSA-treated/<br />

Ctrl) and PHA-activated (GdA-treated) PBMCs was quantified from<br />

western blots using densi<strong>to</strong>metric analysis <strong>with</strong> multigauge software,<br />

normalized <strong>with</strong> b-actin expression and expressed relative <strong>to</strong> the<br />

level in resting PBMCs. Data compiled from five independent experiments<br />

and represented as box plots showing median, interquartile<br />

range and range.<br />

Bcl-2 and prevention of mi<strong>to</strong>chondrial membrane permeabilization<br />

are sufficient <strong>to</strong> rescue T-<strong>cell</strong>s from death by GdA (SundarRaj et al.,<br />

2008). Pockley and Bol<strong>to</strong>n (1989) and Lee et al. (2009) attribute<br />

the immunosuppressive effect of GdA <strong>to</strong> its ability <strong>to</strong> cause inhibition<br />

of <strong>IL</strong>-2 synthesis in T-lymphocytes. We have reported earlier that<br />

GdA <strong>induce</strong>s functional ineptness in differentiated CD8 + T-<strong>cell</strong>s by<br />

blunting the synthesis and release of cy<strong>to</strong>lytic molecules (Soni and<br />

Karande, 2010).<br />

We began this study <strong>with</strong> the simple objective <strong>to</strong> further unravel the<br />

mechanism regulating the expression of perforin and granzyme B in<br />

alloactivated CD8 + T-<strong>cell</strong>s upon GdA treatment. From the available<br />

literature, we knew that T-Bet, Blimp-1, Eomes and Bcl-6 are the<br />

known transcription fac<strong>to</strong>rs regulating CD8 + T-<strong>cell</strong> effec<strong>to</strong>r functions.<br />

Among them, under antigen-specific stimulation conditions, T-Bet is<br />

required for the differentiation of naïve CD8 + T-<strong>cell</strong>s <strong>to</strong> effec<strong>to</strong>r<br />

CTLs and also for the IFN-g synthesis by these <strong>cell</strong>s (Sullivan et al.,<br />

2003). Also, Eomes complements T-bet functions and plays a major<br />

Figure 4 GdA signals <strong>to</strong> <strong>induce</strong> Bax mRNA and protein expression in<br />

activated PBMCs. (a) Bax mRNA levels were analysed by quantitative<br />

RT–PCR in unstimulated PBMCs or PBMCs stimulated <strong>with</strong><br />

gOVCAR-3 for 18 h followed by HSA (Ctrl) or GdA treatment for<br />

24 h. Expression was normalized <strong>with</strong> 18s rRNA gene as control and<br />

plotted as relative fold change in Bax expression upon alloactivation<br />

<strong>with</strong> or <strong>with</strong>out GdA treatment. Data are represented as a box plot<br />

showing median, interquartile range and range of four individual experiments.<br />

(b) Immunoblot analysis of test protein Bax and reference protein<br />

b-actin in resting PBMCs and PBMCs activated for 24 h <strong>with</strong> 5 mg/ml of<br />

PHA <strong>with</strong> simultaneous HSA (Ctrl) or GdA treatment. Proteins were<br />

detected <strong>with</strong> anti-human Bax Ab and anti-b-actin HRP in whole <strong>cell</strong><br />

lysates. 50 mg protein was loaded per well. Data are representative of<br />

four independent experiments. (c) Quantification of Bax protein expression.<br />

Bax protein expression in PHA-activated (HSA-treated/Ctrl) and<br />

PHA-activated (GdA-treated) PBMCs was quantified from western<br />

blots using densi<strong>to</strong>metric analysis <strong>with</strong> multigauge software, normalized<br />

<strong>with</strong> b-actin expression and expressed relative <strong>to</strong> the level in resting<br />

PBMCs. Data represented as box plots showing median, interquartile<br />

range and range compiled from four independent experiments.<br />

role in memory CD8 + T-<strong>cell</strong> development (Intlekofer et al., 2005).<br />

So we looked at the effect of GdA on the induction of the two transcription<br />

fac<strong>to</strong>rs, T-Bet and Eomes upon activation. We found that<br />

Downloaded from<br />

http://humrep.oxfordjournals.org/ by guest on June 17, 2013


8 Soni and Karande<br />

<strong>with</strong> GdA treatment, T-bet was unaffected but Eomes was significantly<br />

down-regulated both at the RNA and at the protein level (Fig. 1).<br />

It is well established that the quality and magnitude of the CD8 +<br />

T-<strong>cell</strong> responses is governed by the strength of the stimula<strong>to</strong>ry<br />

signal, cy<strong>to</strong>kine milieu and availability of ‘CD4 help’. These fac<strong>to</strong>rs<br />

help shape the transcriptional program in the responding CD8 +<br />

T-<strong>cell</strong>s which in turn affect their phenotypic and functional attributes.<br />

We reasoned that GdA could potentially hamper Eomes expression<br />

by affecting the stimula<strong>to</strong>ry signals <strong>to</strong> the <strong>cell</strong>s during activation (attributable<br />

<strong>to</strong> its property of being a lectin; SundarRaj et al., 2009) orby<br />

altering the cy<strong>to</strong>kine milieu through its inhibi<strong>to</strong>ry effect on CD4 +<br />

T-<strong>cell</strong>s, whose survival and functions are known <strong>to</strong> be affected<br />

by GdA.<br />

First, we looked at the effect of GdA on CD28 and CTLA4 which<br />

are required for accelerating or braking T-<strong>cell</strong> proliferation upon activation,<br />

necessary for T-<strong>cell</strong> homeostasis. GdA did not affect the expression<br />

of CD28 on T-<strong>cell</strong>s; CTLA4 was also not up-regulated<br />

upon GdA treatment, suggesting that GdA (most likely) did not<br />

perturb the pro- or anti-stimula<strong>to</strong>ry signals generated upon T-<strong>cell</strong><br />

activation.<br />

Our knowledge about the involvement of <strong>IL</strong>-2 in the development<br />

of CD8 + T-<strong>cell</strong> effec<strong>to</strong>r and memory functions (Pipkin et al., 2010)<br />

and that GdA suppresses <strong>IL</strong>-2 production by stimulated T-<strong>cell</strong>s<br />

(Pockley and Bol<strong>to</strong>n, 1989) prompted us <strong>to</strong> check for CD25 expression<br />

as the read out for <strong>IL</strong>-2 <strong>signalling</strong> in both CD4 + and CD8 + T-<strong>cell</strong><br />

subsets upon GdA treatment. It is well known that responsiveness <strong>to</strong><br />

<strong>IL</strong>-2 is controlled by the <strong>IL</strong>-<strong>2R</strong> complex, which is composed of the<br />

high-affinity <strong>IL</strong>-<strong>2R</strong> a chain (CD25), the <strong>IL</strong>-<strong>2R</strong> b chain (CD122) and<br />

the common g chain (CD132), and that <strong>signalling</strong> through <strong>IL</strong>-<strong>2R</strong>a is<br />

essential for protective immunity (Williams et al., 2006). We observed<br />

that CD25 mRNA and protein expression were significantly reduced<br />

upon GdA treatment in both the T-<strong>cell</strong> subsets, suggesting weak<br />

<strong>IL</strong>-2 <strong>signalling</strong> in the presence of GdA (Fig. 2). Our finding was<br />

strengthened by the observations made by other researchers, i.e.<br />

high <strong>IL</strong>-2 signals <strong>induce</strong> transcription fac<strong>to</strong>rs like STAT5 and Eomes<br />

which directly bind <strong>to</strong> perforin and granzyme gene locus and the<br />

amount of <strong>IL</strong>-2 <strong>signalling</strong> regulates the recruitment of polymerase II<br />

<strong>to</strong> the perforin (high <strong>IL</strong>-2) or the <strong>IL</strong>-7R (low <strong>IL</strong>-2) loci and that lack<br />

<strong>IL</strong>-<strong>2R</strong>a expression on CD8 + T-<strong>cell</strong>s causes aberrant killing of virally<br />

infected <strong>cell</strong>s (Pipkin et al., 2010). Additionally, a modest difference<br />

exists in effec<strong>to</strong>r molecules expression between CD25 hi and CD25 lo<br />

T-<strong>cell</strong>s (Kalia et al., 2010), <strong>with</strong> CD25 lo CD8 + T-<strong>cell</strong>s expressing<br />

fewer cy<strong>to</strong>lytic molecules.<br />

Thus, these findings indicate that the interaction of GdA <strong>with</strong> activated<br />

CD4 + and CD8 + T-<strong>cell</strong>s suppresses <strong>IL</strong>-2/<strong>IL</strong>-<strong>2R</strong> <strong>signalling</strong><br />

<strong>induce</strong>d upon T-<strong>cell</strong> activation, which seems <strong>to</strong> be the causative<br />

agent for the loss of proliferation of T-<strong>cell</strong>s upon GdA treatment. Additionally,<br />

aberrant <strong>IL</strong>-<strong>2R</strong>-mediated <strong>signalling</strong> also contributes <strong>to</strong><br />

reduced expression of cy<strong>to</strong>lytic genes (perforin and granzyme B) in<br />

CD8 + T-<strong>cell</strong>s, which are directly controlled by <strong>IL</strong>-2 as well (Janas<br />

et al., 2005).<br />

Consolidating data from our previous work (Soni and Karande,<br />

2010) and this study, we can say <strong>with</strong> much confidence that in<br />

CD8 + T-<strong>cell</strong>s, GdA <strong>induce</strong>s suboptimal <strong>IL</strong>-2 <strong>signalling</strong> that leads <strong>to</strong><br />

the inhibition of proliferation and suppression of perforin and granzyme<br />

synthesis directly and also indirectly by suboptimal induction<br />

of Eomes. T-bet levels were unaffected by GdA which explains the<br />

moderate effects of GdA seen on CD8 + T-<strong>cell</strong>s and the requirement<br />

of a very high concentration of GdA <strong>to</strong> affect CTL activity (Soni and<br />

Karande, 2010). It also explains why there was no significant effect<br />

of GdA treatment on IFN-g production in CD8 + T-<strong>cell</strong>s. We can envisage<br />

that a reduction in Eomes expression by GdA might also contribute<br />

<strong>to</strong> lowered number of CD8 + T-memory <strong>cell</strong>s, further<br />

contributing <strong>to</strong> fetal <strong>to</strong>lerance.<br />

Studies on <strong>to</strong>tal T-lymphocytes clearly demonstrate that in addition<br />

<strong>to</strong> promoting proliferation, <strong>IL</strong>-<strong>2R</strong>a expression also prevents apop<strong>to</strong>sis<br />

of T-<strong>cell</strong>s, by up-regulating Bcl-2 expression (Gomez et al., 1998). We<br />

observed a reduction in CD25 hi/bright surface expression on T-<strong>cell</strong>s<br />

upon GdA treatment (Fig. 2d). Previous work from our labora<strong>to</strong>ry<br />

attributed the apop<strong>to</strong>genic activity of GdA on primary T-<strong>cell</strong>s and<br />

T-<strong>cell</strong> lines via the intrinsic mi<strong>to</strong>chondrial pathway (Mukhopadhyay<br />

et al., 2001; SundarRaj et al., 2008), which could be competed out<br />

by the overexpression of Bcl-2 (SundarRaj et al., 2008). We questioned<br />

whether suppression of CD25 expression by GdA was<br />

causing a s<strong>to</strong>ichiometric imbalance between the pro- and anti-survival<br />

proteins <strong>with</strong>in T-<strong>cell</strong>s. Indeed, we found that GdA treatment did not<br />

allow an increase in the Bcl-2 protein level <strong>with</strong>in <strong>cell</strong>s which is otherwise<br />

observed in activated T-<strong>cell</strong>s (Fig. 3), although at the mRNA level,<br />

the decrease was not found <strong>to</strong> be statistically significant. Interestingly<br />

though, pro-apop<strong>to</strong>tic protein Bax was up-regulated at both the<br />

mRNA and the protein level (Fig. 4). At least four proteins Bcl-2,<br />

14-3-3, humanin and Ku-70 have been shown <strong>to</strong> inhibit apop<strong>to</strong>sis<br />

by binding <strong>to</strong> Bax and sequestering it from the mi<strong>to</strong>chondria (Amsel<br />

et al., 2008). With the data available on the activity of GdA and<br />

from the present study, we can surmise that the perturbation in the<br />

ratio of Bcl-2 and Bax leads <strong>to</strong> the availability of free Bax <strong>to</strong> oligomerize<br />

and initiate mi<strong>to</strong>chondrial membrane permeabilization leading <strong>to</strong><br />

apop<strong>to</strong>sis. This phenomenon seems <strong>to</strong> operate in CD4 + T-<strong>cell</strong>s,<br />

where the suboptimal <strong>IL</strong>-<strong>2R</strong> expression may lead <strong>to</strong> a reduced <strong>IL</strong>-2<br />

level which is insufficient <strong>to</strong> support the optimal up-regulation of<br />

Bcl-2 upon T-<strong>cell</strong> stimulation, necessary for survival. Unable <strong>to</strong> cope<br />

<strong>with</strong> the mi<strong>to</strong>chondrial stress, CD4 + T-<strong>cell</strong>s undergo apop<strong>to</strong>sis. Intriguingly,<br />

although the CD8 + T-lymphocytes have reduced CD25 expression<br />

upon GdA treatment, they are resistant <strong>to</strong> GdA-<strong>induce</strong>d<br />

apop<strong>to</strong>sis (Soni and Karande, 2010). Similarly, the Th2 <strong>cell</strong>s are also<br />

resistant <strong>to</strong> GdA-<strong>induce</strong>d apop<strong>to</strong>sis (Lee et al., 2011), suggesting<br />

that the process of apop<strong>to</strong>sis induction may not be as simplistic and<br />

there may be many more players involved that may vary in different<br />

T-<strong>cell</strong> subsets.<br />

The different mechanisms put forth by various researchers <strong>to</strong><br />

explain GdA activity on different <strong>cell</strong> types can be explained by the<br />

fact that GdA functions as a galac<strong>to</strong>se-specific lectin (SundarRaj<br />

et al., 2009), and it can potentially bind <strong>to</strong> varied glycosylated proteins<br />

on the T-<strong>cell</strong> surface (Rachmilewitz et al., 2003; Yaniv et al., 2003;<br />

SundarRaj et al., 2009). Among the many potential candidates as the<br />

recep<strong>to</strong>r for GdA on T-<strong>cell</strong>s, CD45 (Rachmilewitz et al., 2003) and<br />

CD7 are the most extensively discussed. We observed that<br />

GdA-<strong>induce</strong>d apop<strong>to</strong>sis in T-<strong>cell</strong> lines in a CD7-dependent manner<br />

(SundarRaj et al., 2009). GdA treatment itself did not affect the expression<br />

of CD7 on both the T-<strong>cell</strong> subsets (data not shown). CD7<br />

is known <strong>to</strong> augment T-<strong>cell</strong> proliferation/function by up-regulating<br />

CD25 expression and <strong>IL</strong>-2 production (Wallace et al., 2000; Stillwell<br />

and Bierer, 2001). Very interestingly, CD7-deficient mice show<br />

decreased CTL effec<strong>to</strong>r functions (Lee et al., 1998). Until now, a<br />

Downloaded from<br />

http://humrep.oxfordjournals.org/ by guest on June 17, 2013


<strong>Glycodelin</strong>-A dampens CD25 expression in T-<strong>cell</strong>s 9<br />

definitive ligand for CD7 is not known and only lectins have been proposed<br />

<strong>to</strong> be potential CD7 ligands (Leta et al., 1996). Considering the<br />

fact that glycodelin is also a lectin, it will be interesting <strong>to</strong> probe<br />

whether glycodelin might exert its inhibi<strong>to</strong>ry effect on T-<strong>cell</strong>s by interacting<br />

<strong>with</strong> CD7 and inhibiting its normal co-stimula<strong>to</strong>ry function.<br />

Al<strong>to</strong>gether, from the present study and available literature, it can be<br />

proposed that the binding of GdA <strong>to</strong> CD4 + and CD8 + T-<strong>cell</strong>s blocks<br />

the co-stimula<strong>to</strong>ry signals required for optimal CD25 expression and<br />

hence <strong>IL</strong>-2 production. Resultant suboptimal <strong>IL</strong>-2 <strong>signalling</strong> has<br />

varied effects; (i) <strong>cell</strong> cycle arrest as seen in both the major T-<strong>cell</strong><br />

subsets; (ii) reduced induction of Eomes that causes retarded synthesis<br />

of cy<strong>to</strong>lytic genes, causing suppression of CTL activity in CD8 + T-<strong>cell</strong>s;<br />

(iii) insufficient induction of anti-apop<strong>to</strong>tic genes upon T-<strong>cell</strong> activation,<br />

which increases the ratio of pro- <strong>to</strong> anti-apop<strong>to</strong>tic proteins,<br />

affecting mi<strong>to</strong>chondrial integrity, finally leading <strong>to</strong> <strong>cell</strong> death in some<br />

CD4 + T-<strong>cell</strong> subsets. The mechanism of GdA activity suggested<br />

here seems <strong>to</strong> be operational in vivo and the expression of CD25<br />

has also been found <strong>to</strong> be selectively reduced on decidual CD4 +<br />

and CD8 + T-lymphocytes in normal pregnancies (Chao et al.,<br />

2002). Nevertheless, it is important that similar studies be carried<br />

out using decidual T-lymphocytes <strong>to</strong> further confirm the effects of<br />

GdA seen on peripheral T-<strong>cell</strong>s, as the localized abundance of progesterone<br />

in the uterus during pregnancy may alter the activity of GdA on<br />

decidual T-<strong>cell</strong>s. It will also be pertinent <strong>to</strong> mention here that peripheral<br />

T-<strong>cell</strong>s from men and women (at different times in their menstrual<br />

cycle), used as the model system for this study, are exposed <strong>to</strong> varying<br />

concentrations of serum progesterone and oestrogen, which may have<br />

a bearing on the effect of glycodelin on the T-<strong>cell</strong>s and can be an interesting<br />

fac<strong>to</strong>r <strong>to</strong> study in depth.<br />

In conclusion, the results put forth in this study demonstrate a previously<br />

unappreciated/underplayed effect of GdA on <strong>IL</strong>-2/<strong>IL</strong>-<strong>2R</strong> <strong>signalling</strong>.<br />

<strong>IL</strong>-2 growth fac<strong>to</strong>r deprivation can explain effects of GdA on<br />

T-<strong>cell</strong>s, including the inhibition of proliferation, apop<strong>to</strong>sis and loss of<br />

effec<strong>to</strong>r functions which contribute <strong>to</strong> the establishment, progression<br />

and maintenance of primate pregnancy but the variations observed in<br />

the responsiveness of different <strong>cell</strong> types <strong>to</strong> GdA indicate that the<br />

understanding of the mechanistic details of GdA activity on hema<strong>to</strong>poietic<br />

<strong>cell</strong>s is still far from complete.<br />

Supplementary data<br />

Supplementary data are available at http://humrep.oxfordjournals.<br />

org/.<br />

Authors’ roles<br />

C.S. has participated in the study design, execution of experiments,<br />

data analysis, writing the manuscript and discussions. A.A.K. supervised<br />

the study and was involved in the design of experiments and critical<br />

analysis of the data, manuscript corrections and discussions.<br />

Funding<br />

This work is supported by grants from the Department of Biotechnology<br />

(DBT), India and the Indian Institute of Science, (IISc) Bangalore,<br />

India.<br />

Conflict of interest<br />

None declared.<br />

References<br />

Akbar AN, Borthwick NJ, Wickremasinghe RG, Panayoitidis P, Pilling D,<br />

Bofill M, Krajewski S, Reed JC, Salmon M. Interleukin-2 recep<strong>to</strong>r<br />

common gamma-chain signaling cy<strong>to</strong>kines regulate activated T <strong>cell</strong><br />

apop<strong>to</strong>sis in response <strong>to</strong> growth fac<strong>to</strong>r <strong>with</strong>drawal: selective induction<br />

of anti-apop<strong>to</strong>tic (bcl-2, bcl-xL) but not pro-apop<strong>to</strong>tic (bax, bcl-xS)<br />

gene expression. Eur J Immunol 1996;26:294–299.<br />

Alok A, Karande AA. The role of glycodelin as an immune-modulating<br />

agent at the fe<strong>to</strong>-maternal interface. J Reprod Immunol 2009;<br />

83:124–127.<br />

Alok A, Mukhopadhyay D, Karande AA. <strong>Glycodelin</strong> A, an<br />

immunomodula<strong>to</strong>ry protein in the endometrium, inhibits proliferation<br />

and <strong>induce</strong>s apop<strong>to</strong>sis in monocytic <strong>cell</strong>s. Int J Biochem Cell Biol 2009;<br />

41:1138–1147.<br />

Amsel AD, Rathaus M, Kronman N, Cohen HY. Regulation of the<br />

proapop<strong>to</strong>tic fac<strong>to</strong>r Bax by Ku70-dependent deubiquitylation. Proc<br />

Natl Acad Sci USA 2008;105:5117–5122.<br />

Araki Y, Fann M, Wers<strong>to</strong> R, Weng NP. His<strong>to</strong>ne acetylation facilitates rapid<br />

and robust memory CD8 T <strong>cell</strong> response through differential expression<br />

of effec<strong>to</strong>r molecules (eomesodermin and its targets: perforin and<br />

granzyme B). J Immunol 2008;180:8102–8108.<br />

Belz GT, Masson F. Interleukin-2 tickles T <strong>cell</strong> memory. Immunity 2010;<br />

32:7–9.<br />

Bol<strong>to</strong>n AE, Pockley AG, Clough KJ, Mowles EA, S<strong>to</strong>ker RJ,<br />

Westwood OM, Chapman MG. Identification of placental protein 14<br />

as an immunosuppressive fac<strong>to</strong>r in human reproduction. Lancet 1987;<br />

1:593–595.<br />

Boyum A. Separation of white blood <strong>cell</strong>s. Nature 1964;204:793–794.<br />

Chao KH, Wu MY, Yang JH, Chen SU, Yang YS, Ho HN. Expression of the<br />

interleukin-2 recep<strong>to</strong>r alpha (CD25) is selectively decreased on decidual<br />

CD4+ and CD8+ T lymphocytes in normal pregnancies. Mol Hum<br />

Reprod 2002;8:667–673.<br />

Dal<strong>to</strong>n CF, Laird SM, Estdale SE, Saravelos HG, Li TC. Endometrial protein<br />

PP14 and CA-125 in recurrent miscarriage patients; correlation <strong>with</strong><br />

pregnancy outcome. Hum Reprod 1998;13:3197–3202.<br />

Deng G, Podack ER. Suppression of apop<strong>to</strong>sis in a cy<strong>to</strong><strong>to</strong>xic T-<strong>cell</strong> line by<br />

interleukin 2-mediated gene transcription and deregulated expression of<br />

the pro<strong>to</strong>oncogene bcl-2. Proc Natl Acad Sci USA 1993;90:2189–2193.<br />

Gomez J, Martinez AC, Gonzalez A, Garcia A, Rebollo A. The Bcl-2 gene<br />

is differentially regulated by <strong>IL</strong>-2 and <strong>IL</strong>-4: role of the transcription fac<strong>to</strong>r<br />

NF-AT. Oncogene 1998;17:1235–1243.<br />

Hengartner MO. The biochemistry of apop<strong>to</strong>sis. Nature 2000;<br />

407:770–776.<br />

Intlekofer AM, Takemo<strong>to</strong> N, Wherry EJ, Longworth SA, Northrup JT,<br />

Palanivel VR, Mullen AC, Gasink CR, Kaech SM, Miller JD et al.<br />

Effec<strong>to</strong>r and memory CD8+ T <strong>cell</strong> fate coupled by T-bet and<br />

eomesodermin. Nat Immunol 2005;6:1236–1244.<br />

Janas ML, Groves P, Kienzle N, Kelso A. <strong>IL</strong>-2 regulates perforin and<br />

granzyme gene expression in CD8+ T <strong>cell</strong>s independently of its<br />

effects on survival and proliferation. J Immunol 2005;175:8003–8010.<br />

Jayachandran R, Shaila MS, Karande AA. Analysis of the role of<br />

oligosaccharides in the apop<strong>to</strong>tic activity of glycodelin A. J Biol Chem<br />

2004;279:8585–8591.<br />

Jayachandran R, Radcliffe CM, Royle L, Harvey DJ, Dwek RA, Rudd PM,<br />

Karande AA. Oligosaccharides modulate the apop<strong>to</strong>tic activity of<br />

glycodelin. Glycobiology 2006;16:1052–1063.<br />

Downloaded from<br />

http://humrep.oxfordjournals.org/ by guest on June 17, 2013


10 Soni and Karande<br />

Joshi SG. A progestagen-associated protein of the human endometrium:<br />

basic studies and potential clinical applications. J Steroid Biochem 1983;<br />

19:751–757.<br />

Kalia V, Sarkar S, Subramaniam S, Haining WN, Smith KA, Ahmed R.<br />

Prolonged interleukin-<strong>2R</strong>alpha expression on virus-specific CD8+ T<br />

<strong>cell</strong>s favors terminal-effec<strong>to</strong>r differentiation in vivo. Immunity 2010;<br />

32:91–103.<br />

Lee DM, Staats HF, Sundy JS, Patel DD, Sempowski GD, Scearce RM,<br />

Jones DM, Haynes BF. Immunologic characterization of CD7-deficient<br />

mice. J Immunol 1998;160:5749–5756.<br />

Lee CL, Pang PC, Yeung WS, Tissot B, Panico M, Lao TT, Chu IK, Lee KF,<br />

Chung MK, Lam KK et al. Effects of differential glycosylation of<br />

glycodelins on lymphocyte survival. J Biol Chem 2009;<br />

284:15084–15096.<br />

Lee CL, Chiu PC, Lam KK, Chan RW, Chu IK, Koistinen R, Koistinen H,<br />

Seppala M, Lee KF, Yeung WS. <strong>Glycodelin</strong>-A modulates cy<strong>to</strong>kine<br />

production of peripheral blood natural killer <strong>cell</strong>s. Fertil Steril 2010;<br />

94:769–771.<br />

Lee CL, Chiu PC, Lam KK, Siu SO, Chu IK, Koistinen R, Koistinen H,<br />

Seppala M, Lee KF, Yeung WS. Differential actions of glycodelin-A on<br />

Th-1 and Th-2 <strong>cell</strong>s: a paracrine mechanism that could produce the<br />

Th-2 dominant environment during pregnancy. Hum Reprod 2011;<br />

26:517–526.<br />

Leta E, Hou Z, Lederman L, Jung LK. Interaction between the extra<strong>cell</strong>ular<br />

domain of CD7 and concanavalin A: a clue <strong>to</strong> the identity of the ligand<br />

for CD7. Cell Immunol 1996;173:15–21.<br />

Mackenna A, Li TC, Dal<strong>to</strong>n C, Bol<strong>to</strong>n A, Cooke I. Placental protein 14<br />

levels in uterine flushing and plasma of women <strong>with</strong> unexplained<br />

infertility. Fertil Steril 1993;59:577–582.<br />

Mishan-Eisenberg G, Borovsky Z, Weber MC, Gazit R, Tykocinski ML,<br />

Rachmilewitz J. Differential regulation of Th1/Th2 cy<strong>to</strong>kine responses<br />

by placental protein 14. J Immunol 2004;173:5524–5530.<br />

Miyazaki T, Liu ZJ, Kawahara A, Minami Y, Yamada K, Tsujimo<strong>to</strong> Y,<br />

Barsoumian EL, Permutter RM, Taniguchi T. Three distinct <strong>IL</strong>-2<br />

signaling pathways mediated by bcl-2, c-myc, and lck cooperate in<br />

hema<strong>to</strong>poietic <strong>cell</strong> proliferation. Cell 1995;81:223–231.<br />

Morrison TB, Weis JJ, Wittwer CT. Quantification of low-copy transcripts<br />

by continuous SYBR Green I moni<strong>to</strong>ring during amplification.<br />

Biotechniques 1998;24:954–958, 960, 962.<br />

Mukhopadhyay D, Sundereshan S, Rao C, Karande AA. Placental protein<br />

14 <strong>induce</strong>s apop<strong>to</strong>sis in T <strong>cell</strong>s but not in monocytes. J Biol Chem 2001;<br />

276:28268–28273.<br />

Mukhopadhyay D, SundarRaj S, Alok A, Karande AA. <strong>Glycodelin</strong> A, not<br />

glycodelin S, is apop<strong>to</strong>tically active. Relevance of sialic acid<br />

modification. J Biol Chem 2004;279:8577–8584.<br />

Niederkorn JY. See no evil, hear no evil, do no evil: the lessons of immune<br />

privilege. Nat Immunol 2006;7:354–359.<br />

Okamo<strong>to</strong> N, Uchida A, Takakura K, Kariya Y, Kanzaki H, Riittinen L,<br />

Koistinen R, Seppala M, Mori T. Suppression by human placental<br />

protein 14 of natural killer <strong>cell</strong> activity. Am J Reprod Immunol 1991;<br />

26:137–142.<br />

Oltvai ZN, Milliman CL, Korsmeyer SJ. Bcl-2 heterodimerizes in vivo <strong>with</strong><br />

a conserved homolog, Bax, that accelerates programmed <strong>cell</strong> death. Cell<br />

1993;74:609–619.<br />

Pearce EL, Mullen AC, Martins GA, Krawczyk CM, Hutchins AS,<br />

Zediak VP, Banica M, DiCioccio CB, Gross DA, Mao CA et al.<br />

Control of effec<strong>to</strong>r CD8+ T <strong>cell</strong> function by the transcription fac<strong>to</strong>r<br />

eomesodermin. Science 2003;302:1041–1043.<br />

Pearson H. Reproductive immunology: imunity’s pregnant pause. Nature<br />

2002;420:265–266.<br />

Pipkin ME, Sacks JA, Cruz-Guilloty F, Lichtenheld MG, Bevan MJ, Rao A.<br />

Interleukin-2 and inflammation <strong>induce</strong> distinct transcriptional programs<br />

that promote the differentiation of effec<strong>to</strong>r cy<strong>to</strong>lytic T <strong>cell</strong>s. Immunity<br />

2010;32:79–90.<br />

Pockley AG, Bol<strong>to</strong>n AE. Placental protein 14 (PP14) inhibits the synthesis<br />

of interleukin-2 and the release of soluble interleukin-2 recep<strong>to</strong>rs from<br />

phy<strong>to</strong>haemagglutinin-stimulated lymphocytes. Clin Exp Immunol 1989;<br />

77:252–256.<br />

Poornima BL, Karande AA. Differential sialylation regulates the apop<strong>to</strong>tic<br />

activity of glycodelin A. FEBS Lett 2007;581:4366–4370.<br />

Rachmilewitz J, Riely GJ, Tykocinski ML. Placental protein 14 functions as a<br />

direct T-<strong>cell</strong> inhibi<strong>to</strong>r. Cell Immunol 1999;191:26–33.<br />

Rachmilewitz J, Riely GJ, Huang JH, Chen A, Tykocinski ML. A rheostatic<br />

mechanism for T-<strong>cell</strong> inhibition based on elevation of activation<br />

thresholds. Blood 2001;98:3727–3732.<br />

Rachmilewitz J, Borovsky Z, Riely GJ, Miller R, Tykocinski ML. Negative<br />

regulation of T <strong>cell</strong> activation by placental protein 14 is mediated by<br />

the tyrosine phosphatase recep<strong>to</strong>r CD45. J Biol Chem 2003;<br />

278:14059–14065.<br />

Scholz C, Toth B, Brunnhuber R, Rampf E, Weissenbacher T, San<strong>to</strong>so L,<br />

Friese K, Jeschke U. <strong>Glycodelin</strong> A <strong>induce</strong>s a <strong>to</strong>lerogenic phenotype in<br />

monocyte-derived dendritic <strong>cell</strong>s in vitro. Am J Reprod Immunol 2008;<br />

60:501–512.<br />

Seppala M, Taylor RN, Koistinen H, Koistinen R, Milgrom E.<br />

<strong>Glycodelin</strong>: a major lipocalin protein of the reproductive axis <strong>with</strong><br />

diverse actions in <strong>cell</strong> recognition and differentiation. Endocr Rev<br />

2002;23:401–430.<br />

Soni C, Karande AA. <strong>Glycodelin</strong> A suppresses the cy<strong>to</strong>lytic activity of<br />

CD8+ T lymphocytes. Mol Immunol 2010;47:2458–2466.<br />

Starkey PM, Sargent <strong>IL</strong>, Redman CW. Cell populations in human early<br />

pregnancy decidua: characterization and isolation of large granular<br />

lymphocytes by flow cy<strong>to</strong>metry. Immunology 1988;65:129–134.<br />

Stillwell R, Bierer BE. T <strong>cell</strong> signal transduction and the role of CD7 in<br />

costimulation. Immunol Res 2001;24:31–52.<br />

Sullivan BM, Juedes A, Szabo SJ, von Herrath M, Glimcher LH.<br />

Antigen-driven effec<strong>to</strong>r CD8 T <strong>cell</strong> function regulated by T-bet. Proc<br />

Natl Acad Sci USA 2003;100:15818–15823.<br />

SundarRaj S, Mukhopadhyay D, Karande AA. <strong>Glycodelin</strong> A triggers<br />

mi<strong>to</strong>chondrial stress and apop<strong>to</strong>sis in T <strong>cell</strong>s by a mechanism distinct<br />

and independent of TCR signaling. Mol Immunol 2008;45:2391–2400.<br />

SundarRaj S, Soni C, Karande AA. <strong>Glycodelin</strong> A triggers T <strong>cell</strong> apop<strong>to</strong>sis<br />

through a novel calcium-independent galac<strong>to</strong>se-binding lectin activity.<br />

Mol Immunol 2009;46:3411–3419.<br />

Szabo SJ, Kim ST, Costa GL, Zhang X, Fathman CG, Glimcher LH. A novel<br />

transcription fac<strong>to</strong>r, T-bet, directs Th1 lineage commitment. Cell 2000;<br />

100:655–669.<br />

Szabo SJ, Sullivan BM, Stemmann C, Sa<strong>to</strong>skar AR, Sleckman BP,<br />

Glimcher LH. Distinct effects of T-bet in TH1 lineage commitment<br />

and IFN-gamma production in CD4 and CD8 T <strong>cell</strong>s. Science 2002;<br />

295:338–342.<br />

Taylor RN, Savouret JF, Vaisse C, Vigne JL, Ryan I, Hornung D, Seppala M,<br />

Milgrom E. Promeges<strong>to</strong>ne (R5020) and mifepris<strong>to</strong>ne (RU486) both<br />

function as progestational agonists of human glycodelin gene<br />

expression in isolated human epithelial <strong>cell</strong>s. J Clin Endocrinol Metab<br />

1998;83:4006–4012.<br />

Tee MK, Vigne JL, Yu J, Taylor RN. Natural and recombinant human<br />

glycodelin activate a proapop<strong>to</strong>tic gene cascade in monocyte <strong>cell</strong>s.<br />

J Leukoc Biol 2008;83:843–852.<br />

Terness P, Kallikourdis M, Betz AG, Rabinovich GA, Sai<strong>to</strong> S, Clark DA.<br />

Tolerance signaling molecules and pregnancy: IDO, galectins, and the<br />

renaissance of regula<strong>to</strong>ry T <strong>cell</strong>s. Am J Reprod Immunol 2007;<br />

58:238–254.<br />

Trowsdale J, Betz AG. Mother’s little helpers: mechanisms of<br />

maternal-fetal <strong>to</strong>lerance. Nat Immunol 2006;7:241–246.<br />

Downloaded from<br />

http://humrep.oxfordjournals.org/ by guest on June 17, 2013


<strong>Glycodelin</strong>-A dampens CD25 expression in T-<strong>cell</strong>s 11<br />

Tulppala M, Julkunen M, Tiitinen A, Stenman UH, Seppala M. Habitual<br />

abortion is accompanied by low serum levels of placental protein 14<br />

in the luteal phase of the fertile cycle. Fertil Steril 1995;63:792–795.<br />

van Kampen CA, Versteeg-van der Voort Maarschalk MF,<br />

Langerak-Langerak J, van Beelen E, Roelen DL, Claas FH. Pregnancy<br />

can <strong>induce</strong> long-persisting primed CTLs specific for inherited paternal<br />

HLA antigens. Hum Immunol 2001;62:201–207.<br />

Van Parijs L, Biuckians A, Ibragimov A, Alt FW, Willerford DM, Abbas AK.<br />

Functional responses and apop<strong>to</strong>sis of CD25 (<strong>IL</strong>-<strong>2R</strong> alpha)-deficient T<br />

<strong>cell</strong>s expressing a transgenic antigen recep<strong>to</strong>r. J Immunol 1997;<br />

158:3738–3745.<br />

Veenstra van Nieuwenhoven AL, Heineman MJ, Faas MM. The<br />

immunology of successful pregnancy. Hum Reprod Update 2003;<br />

9:347–357.<br />

Wallace DL, Matear PM, Davies DC, Hicks R, Lebosse C, Eyeson J,<br />

Beverley PC, Vyakarnam A. CD7 expression distinguishes subsets<br />

of CD4(+) T <strong>cell</strong>s <strong>with</strong> distinct functional properties and<br />

ability <strong>to</strong> support replication of HIV-1. Eur J Immunol 2000;<br />

30:577–585.<br />

Williams MA, Tyznik AJ, Bevan MJ. Interleukin-2 signals during priming are<br />

required for secondary expansion of CD8+ memory T <strong>cell</strong>s. Nature<br />

2006;441:890–893.<br />

Williams PJ, Searle RF, Robson SC, Innes BA, Bulmer JN. Decidual<br />

leucocyte populations in early <strong>to</strong> late gestation normal human<br />

pregnancy. J Reprod Immunol 2009;82:24–31.<br />

Yaniv E, Borovsky Z, Mishan-Eisenberg G, Rachmilewitz J. Placental protein<br />

14 regulates selective B <strong>cell</strong> responses. Cell Immunol 2003;<br />

222:156–163.<br />

Downloaded from<br />

http://humrep.oxfordjournals.org/ by guest on June 17, 2013

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!