Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

REPRINTED BY PERMISSION OF THE AMERICAN ASOCIATION OF IMMUNOLOGISTS,

INC. FROM "UMBILICAL CORD-DERIVED MESENCHYMAL STROMAL CELLS MODULATE


MONOCYTE FUNCTION TO SUPPRESS T CELL PROLIFERATION” BY CUTLER AJ. ET AL.
THE JOURNAL OF IMMUNOLOGY VOL. 185 PP. 6617-23 COPYRIGHT 2010.

Umbilical Cord-Derived Mesenchymal


Stromal Cells Modulate Monocyte Function to
Suppress T Cell Proliferation

This information is current as Antony J. Cutler, Vasanti Limbani, John Girdlestone and
of December 15, 2010 Cristina V. Navarrete

J Immunol 2010;185;6617-6623; Prepublished online 27


October 2010;
doi:10.4049/jimmunol.1002239
http://www.jimmunol.org/content/185/11/6617

Downloaded from www.jimmunol.org on December 15, 2010


References This article cites 50 articles, 19 of which can be accessed free at:
http://www.jimmunol.org/content/185/11/6617.full.html#ref-list-1
Subscriptions Information about subscribing to The Journal of Immunology is
online at http://www.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://www.jimmunol.org/etoc/subscriptions.shtml/

The Journal of Immunology is published twice each month by


The American Association of Immunologists, Inc.,
9650 Rockville Pike, Bethesda, MD 20814-3994.
Copyright ©2010 by The American Association of
Immunologists, Inc. All rights reserved.
Print ISSN: 0022-1767 Online ISSN: 1550-6606.
The Journal of Immunology

Umbilical Cord-Derived Mesenchymal Stromal Cells


Modulate Monocyte Function to Suppress T Cell Proliferation

Antony J. Cutler,* Vasanti Limbani,* John Girdlestone,*,† and Cristina V. Navarrete*,†


Mesenchymal stromal cells (MSCs) may be derived from a variety of tissues, with human umbilical cord (UC) providing an abun-
dant and noninvasive source. Human UC-MSCs share similar in vitro immunosuppressive properties as MSCs obtained from bone
marrow and cord blood. However, the mechanisms and cellular interactions used by MSCs to control immune responses remain to
be fully elucidated. In this paper, we report that suppression of mitogen-induced T cell proliferation by human UC-, bone marrow-,
and cord blood-MSCs required monocytes. Removal of monocytes but not B cells from human adult PBMCs (PBMNCs) reduced the
immunosuppressive effects of MSCs on T cell proliferation. There was rapid modulation of a number of cell surface molecules on
monocytes when PBMCs or alloantigen-activated PBMNCs were cultured with UC-MSCs. Indomethacin treatment significantly
inhibited the ability of UC-MSCs to suppress T cell proliferation, indicating an important role for PGE2. Monocytes purified from
UC-MSC coculture had significantly reduced accessory cell and allostimulatory function when tested in subsequent T cell pro-
liferation assays, an effect mediated in part by UC-MSC PGE2 production and enhanced by PBMNC alloactivation. Therefore, we

Downloaded from www.jimmunol.org on December 15, 2010


identify monocytes as an essential intermediary through which UC-MSCs mediate their suppressive effects on T cell prolifer-
ation. The Journal of Immunology, 2010, 185: 6617–6623.

M
esenchymal stromal cells (MSCs) have a profound (moDCs) (reviewed in Ref. 2). MSCs secrete a plethora of growth
immunosuppressive capability in vitro (1) and are factors, cytokines, and immunomodulatory mediators. PGE2, NO,
currently being assessed as a novel cellular therapeutic HLA-G, IDO, IL-10, TGF-b, hepatocyte growth factor, IL-6, and
anti-inflammatory agent in numerous clinical trials. MSCs are insulin-like growth factor-1 have all been implicated in MSC in-
being considered as an adjunct to conventional therapy or a stand- duced immunosuppression (reviewed in Ref. 7).
alone therapy to treat a range of autoimmune diseases, acute and Acute GVHD is initiated by host-derived dendritic cells (DCs),
chronic kidney rejection posttransplant, and acute and chronic activated by the inflammatory environment induced by condition-
graft-versus-host disease (GVHD) and to aid tissue repair/wound ing regimes. Donor T cells recognize mismatched MHC molecules
healing (2, 3). Expanded MSCs have been shown to have no on the host tissues and cells including DCs, become activated,
toxicity or adverse events when infused (4), and the most prom- proliferate, and differentiate into mature effectors cells to drive
ising use of MSCs to date is in the treatment of patients with pathogenic responses (8). Following myeloablative conditioning or
steroid-resistant GVHD following hematopoietic stem cell trans- reduced intensity conditioning of the patient, host DCs are rapidly
plantation (5). Most clinical trials currently use MSCs derived replaced by donor-derived DCs in the blood (9) and the skin (10).
from bone marrow (BM); however, MSCs may also be derived Donor-derived DCs may then indirectly present host-derived Ag
from umbilical cord blood (UCB), adipose tissue, placenta, dental and exacerbate the graft-versus-host response (11). Because mono-
pulp, and umbilical cord (UC) (reviewed in Ref. 6). cytes are able to develop into DCs under inflammatory conditions
The mechanisms by which MSCs may ameliorate GVHD are (12), it is conceivable that suppression of GVHD by MSCs is related
unclear, but experimental evidence suggests that MSCs act on to their ability to influence the maturation and function of DCs
many of the leukocyte subsets involved directly or indirectly in the (13, 14).
generation of an antihost immune response posthematopoietic stem We and others have determined that UC-derived MSCs sup-
cell transplantation. MSCs reduce the proliferative capacity of naive press T cell proliferation (15, 16). In this paper, we extend these
and memory T cells, B cells and effector function of gdT cells, observations and explore the cellular interactions used by MSCs
NK cells, and neutrophils. MSCs also block and modulate the in for optimal suppression of T cell proliferation. We show that
vitro maturation and function of monocyte-derived dendritic cells monocytes are required for the optimal suppression of T cell
proliferation induced by TCR cross-linking by UCB-, BM-, and
UC-MSCs. Within 24 h of coculture and prior to their differen-
*Histocompatibility and Immunogenetics Research Group, National Health Service
Blood and Transplant; and †Division of Infection and Immunity, University College tiation into DCs, monocyte phenotype and function are modulated
London, London, United Kingdom by interaction with MSCs. Accessory function and the allosti-
Received for publication July 8, 2010. Accepted for publication September 30, 2010. mulatory capacity of monocytes are downregulated by MSCs via
This work was supported by Grant C043 from the National Institute for Health a mechanism that is in part driven by UC-MSC–derived PGE2.
Research UK. Thus, MSCs appear to act indirectly on T cells via monocytes,
Address correspondence and reprint requests to Dr. Cristina V. Navarrete, Histocom- which act as an essential intermediary cell allowing optimal sup-
patibility and Immunogenetics Research Group, National Health Service Blood and
Transplant, London NW9 5BG, U.K. E-mail address: Cristina.Navarrete@nhsbt.nhs.uk
pression of T cell proliferation by MSCs.
Abbreviations used in this paper: BM, bone marrow; CB, cord blood; DC, dendritic
cell; DC-SIGN, dendritic cell-specific intercellular adhesion molecule-3–grabbing non- Materials and Methods
integrin; GVHD, graft-versus-host disease; moDC, monocyte-derived dendritic cell; Cell lines and PBMCs
MSC, mesenchymal stromal cell; UC, umbilical cord; UCB, umbilical cord blood.
Human UC-derived and cord blood (CB)-derived MSCs, generated as pre-
Copyright ! 2010 by The American Association of Immunologists, Inc. 0022-1767/10/$16.00 viously described (16, 17), were maintained in culture in low glucose-DMEM

www.jimmunol.org/cgi/doi/10.4049/jimmunol.1002239
6618 UC-MSCs MODULATE MONOCYTES TO SUPPRESS T CELL PROLIFERATION

supplemented with 10% FBS, penicillin, streptomycin, and L-glutamine Inhibition and measurement of UC-MSC PGE2 production
(Invitrogen, Carlsbad, CA). Human BM-MSCs were purchased from Lonza,
Basel, Switzerland and maintained in culture in Mesencult medium with UC-MSCs were incubated with 5–10 mM indomethacin (Sigma-Aldrich) or
10% supplements (StemCell Technologies, Grenoble, France). Upon reach- an equivalent volume of carrier (DMSO) (Sigma-Aldrich) in low glucose-
ing confluence cells were released from the plate using trypsin-EDTA DMEM + 10% FBS and penicillin/streptomycin for 24 h. Indomethacin-
(Sigma-Aldrich, St. Louis, MO), counted, and replated. UC-MSCs were treated or control UC-MSCs were subsequently harvested, washed, and cul-
used from passages 3 to 15 with similar results obtained throughout. Human tured with nonactivated or alloantigen-activated PBMNCs at a ratio of 1:2.5
adult PBMCs (PBMNCs) of buffy coat samples chosen at random were in RPMI 1640 medium-10% AB serum for 24 h. PGE2 levels were measured
provided by National Health Service Blood and Transplant (Colindale, U.K). in tissue culture supernatant by ELISA following the manufacturer’s in-
PBMNCs were prepared by density centrifugation over Lymphoprep (Axis- structions (Assay Designs, Enzo Life Sciences, Farmingdale, NY).
Shield, Oslo, Norway), and HLA typing was performed by the Histocom-
patibility and Immunogenetics Department, National Health Service Blood
Statistical analysis
and Transplant, Colindale Center. UC-MSCs were obtained from third-party Analysis of statistics was carried out using SPSS (version 15) software by
donors and HLA mismatched with the PBMNCs used in these experiments. using unpaired or paired student’s t tests where appropriate. All data are
presented as mean 6 SD, and p , 0.05 was considered significant.
Flow cytometry
Cells in PBS, 0.05% sodium azide, 2.5% FBS and 1% Fc block (Miltenyi Results
Biotec, Bergisch Gladbach, Germany) were stained with CD14 specific Ab Monocytes are required for optimal suppression of T cell
(Immunotools, Friesoythe, Germany) to identify monocytes and counter-
stained using a selection of CD73-, CD123-, CD206-, DC-specific in- proliferation by MSCs
tercellular adhesion molecule-3–grabbing nonintegrin (DC-SIGN)–, and We have previously shown that UC-MSCs reduce the proliferation
HLA-DR–specific Abs (all BD Biosciences, Oxford, U.K.). Isotype con-
trols and unstained cells were used for all fluorochromes (BD Bio-
of T cells in vitro (16) and therefore sought to examine and define
sciences). Stained cells were then washed and resuspended in PBS, and the cellular interactions required for MSCs to exert their sup-

Downloaded from www.jimmunol.org on December 15, 2010


data were acquired using a FACSort flow cytometer and CellQuest soft- pressive effect. Addition of monocytes has previously been shown
ware (BD Biosciences). Data were analyzed using WinMDI software (The to restore the ability of BM-MSCs to suppress proliferation of
Scripps Research Institute, La Jolla, CA). purified CD4+ T cells in response to superantigen (18). We con-
Cell purification firmed that addition of monocytes to cocultures of CD4+ T cells
T + NK cells, monocytes, and B cells were removed from PBMNCs, using
anti-CD2, CD14, or CD19 microbeads (Miltenyi Biotec), respectively.
Untouched CD4+ T cells and monocytes were isolated using the CD4+
T cell isolation kit II or monocyte isolation kit II, respectively (Miltenyi
Biotec). All cells were purified using an autoMACS, according to the
manufacturer’s instructions (Miltenyi Biotec).

Monocyte conditioning and purification


Monocytes were purified from nonactivated or alloantigen-activated PBMNC-
conditioning cultures as follows. Nonactivated PBMNC cultures (5 3 104
PBMNC 6 2 3 104 UC-MSCs) and alloantigen-activated PBMNC cultures
(2.5 3 104 CD4+ T cell and 2.5 3 104 CD2-depleted allogeneic PBMNCs 6
2 3 104 UC-MSCs) were cultured in RPMI 1640 medium (Invitrogen) sup-
plemented with 10% human AB serum (Lonza, Basel, Switzerland) and
penicillin/streptomycin for 24 h. After 24 h, PBMNCs or alloantigen-activated
PBMNC MSC cocultures were harvested and initially centrifuged over a 28%
Percoll (Sigma-Aldrich) density gradient to remove MSCs. All cultures were
further centrifuged over a 35% Percoll density gradient to enrich for mono-
cytes. The resulting interface was harvested and washed and untouched
CD14+ cells isolated using the monocyte isolation kit II (Miltenyi Biotec).
All cell sorts were carried out using an autoMACS.
Cell proliferation
Alloantigen stimulation. This was perfomed using the MLR assay as fol-
lows: freshly isolated CD4+ T cells (2.5 3 106/ml in PBS) were incubated
with 1.25 mM CFSE (Sigma-Aldrich) at room temperature. After 5 min,
30.25 volume of FBS was added, and the cells were incubated for an FIGURE 1. MSCs require the presence of monocytes to suppress T cell
additional 10 min. CFSE-labeled cells were washed twice in PBS and proliferation. A, PBMNCs (black bar), CD19-depleted PBMNCs (gray
recounted prior to culture. CFSE-labeled CD4+ T cells (5 3 104) were hatched bar) or CD14-depleted PBMNCs (gray bar) from four independent
incubated with allogeneic highly purified monocytes from conditioning
blood donors were incubated with activation beads in the presence or
cultures (5 3 103) at a ratio of 10:1. After 7 d of culture, proliferating
and activated T cells were identified using CD3 Ab (Immunotools), CD25 absence of UC-MSCs. Proliferation assessed by [3H] uptake after 72 h of
(Serotec, Kidlington, U.K.), and CFSE dye dilution. Monocytes were culture. The degree of suppression of T cell proliferation induced by UC-
excluded from the analysis using CD14 Ab (Immunotools). Stained cells MSCs is expressed as a percentage relative to T cell proliferation in the
were then washed, resuspended, and acquired using the FACSort flow absence of UC-MSCs. Data are shown as mean values 6 SD. Statistical
cytometer as described previously. significance of differences between CD14-depleted PBMNCs versus
PHA and bead stimulations. PBMNCs or purified cell populations plated at PBMNCs and CD19-depleted PBMNCs were determined using paired
5 3 104 cells/well in RPMI 1640 medium + 10% AB serum (Lonza) in Student t test. ****p = 0.0001. B–D, PBMNCs (black bar) or CD14-de-
U-bottomed 96-well plates (Nunc, Naperville, IL) were stimulated with pleted PBMNCs (gray bar) (n = 4) were incubated with activation beads
anti-CD2, -CD3, -CD28, T cell activation beads (1.75 3 104; Miltenyi with or without MSCs from UCB (B), BM (C), or UC (D) (n = 2 MSC lines
Biotec), or PHA (1 mg/ml; Sigma-Aldrich) with or without MSCs (2 3 per source) and proliferation assessed by [3H] uptake after 72 h of culture.
104 cells/well). Some assays were carried out in the presence or absence
The degree of suppression of T cell proliferation induced by MSC
of indomethacin (5–10 mg/ml) (Sigma-Aldrich). Tritiated [3H]thymidine
(37 kBq/well; Amersham Biosciences, Little Chalfont, Buckinghamshire, expressed as a percentage relative to T cell proliferation in the absence of
U.K.) was added after 2 d of culture, and the cells were harvested 16–18 h MSCs. The data are representative of two independent experiments and
later. Thymidine incorporation was measured in a beta counter (PerkinElmer, shown as mean values 6 SD. Statistical significance of differences were
Wellesley, MA). determined using paired Student t test.
The Journal of Immunology 6619

FIGURE 2. UC-MSCs induce pheno-


typic and physical changes in the mono-
cyte population. Flow cytometric analysis
of monocyte phenotype in culture. Non-
activated PBMNC (A, B) or alloantigen
activated (CD4+ T cells: CD2-depleted
APCs in a 1:1 ratio) (C) were cultured
with (n, n = 4) or without (N, n = 6) UC-
MSCs for 24 h or 7 d. The characteristics
and surface phenotype of cells gated on
CD14 expression were analyzed. Statis-
tical significance of differences in geo-
metric mean channel fluorescence was

Downloaded from www.jimmunol.org on December 15, 2010


determined using paired Student t test.
*p , 0.05; **p , 0.01; and ***p ,
0.001. The data are representative of four
independent experiments and shown as
mean values 6 SD.

and MSCs reconstituted the optimal suppressive action of MSCs culture, but notably HLA-DR expression was equivalent after 24 h
on T cell proliferation (data not shown) and extended these in all culture conditions (Fig. 2B, 2C). CD14 expression initially
observations to demonstrate that depletion of monocytes from increased after 24 h but then diminished by day 7 to levels that
PBMNCs completely abolished the capacity of UC-MSCs to were lower or equivalent to control cultures of nonactivated or
suppress T cell proliferation induced by direct activation of T cells alloantigen-activated culture, respectively (Fig. 2B, 2C). CD123
by anti-CD2, -CD3, and -CD28 stimulation. Depletion of B cells expression on monocytes was maintained by day 7 where non-
had no effect on the ability of MSCs to suppress T cell pro- activated PBMNCs were cultured with UC-MSCs but not in me-
liferation (Fig. 1A). Importantly, MSCs derived from UCB (Fig. dium alone (Fig. 2B), whereas no significant difference was
1B) and BM (Fig. 1C) in addition to those derived from UC (Fig. observed on monocytes from alloantigen-activated PBMNC/UC-
1D) all required monocytes to suppress proliferation of T cells MSC coculture (Fig. 2C). Importantly, similar alterations in mono-
in vitro. cyte phenotype were seen with MSCs derived from multiple UC
tissue donors, BM, and CB (data not shown). The majority of the
Coculture of PBMNCs and UC-MSCs induces rapid
changes observed with UC-MSC coculture could be replicated by
phenotypic changes in the monocyte population
using UC-MSC–conditioned medium or culturing UC-MSCs and
BM-MSCs alter the phenotype of moDCs differentiated in vitro PBMNCs across a Transwell (data not shown). However, DC-SIGN
with GM-CSF and IL-4 (13, 14, 19) and that of mature macro- and CD123 expression were unaffected, indicating that additional
phages (20). We therefore followed the phenotype of monocytes signals other than soluble factors produced by MSCs may be re-
cocultured for 24 h and 7 d in vitro with third party UC-MSCs quired for induction of all of the phenotypic changes observed in
in nonactivated (PBMNC 6 UC-MSC) or alloantigen-activated monocytes.
(CD4+ T cell and CD2-depleted allogeneic PBMNC 6 UC-MSC)
PBMNC cell culture conditions. UC-MSCs were found to rap- MSC coculture reduces the accessory and allostimulatory
idly induce physical and phenotypic changes in the monocyte function of monocytes
population within 8 h (data not shown). UC-MSC coculture Monocytes are a major component of the cellular influx into an
increased the size and granularity of monocytes only in non- inflammatory environment (21). They are also able to differentiate
activated PBMNC cocultures (Fig. 2A). UC-MSC coculture in- into inflammatory DCs or macrophages with both pro- and anti-
creased surface expression of CD73, CD206, and DC-SIGN to inflammatory properties dependent on environmental stimuli (22–
a significant degree on CD14+ cells from both culture conditions 24). To investigate whether monocyte function as well as pheno-
(Fig. 2B, 2C). HLA-DR expression was decreased on monocytes type was modulated following UC-MSC exposure, we analyzed
by UC-MSCs compared with those in medium alone by day 7 in the capacity of monocytes to support T cell proliferation in response
6620 UC-MSCs MODULATE MONOCYTES TO SUPPRESS T CELL PROLIFERATION

to PHA stimulation, because highly purified CD4+ T cells only re- was constitutively produced by UC-MSCs and increased levels
spond to PHA in the presence of accessory cells such as monocytes were detected where nonactivated or alloantigen-activated PBMNCs
(25). Monocytes (.85% CD14+ with ,0.5% UC-MSC contami- were cultured with UC-MSCs (Fig. 4A). As MSC-derived PGE2 may
nation) purified from nonactivated PBMNCs following 24 h of modulate moDC maturation (26) and suppress T cell proliferation
coculture with UC-MSCs had profoundly reduced accessory cell (27), we therefore assessed whether UC-MSCs mediated their im-
function in subsequent PHA stimulation assays when compared munosuppressive effects via PGE2. In agreement with a previous
with those cultured in medium alone (Fig. 3A). Monocytes purified report (15, 15), we found that UC-MSCs were unable to inhibit PHA
from highly pure CD14+ and UC-MSC coculture also had reduced induced proliferation of PBMNC in the presence of indomethacin
accessory function compared with those cultured in medium alone (a nonspecific cyclooxygenase-1/2 inhibitor) (Fig. 4B).
(Fig. 3B), indicating that other mononuclear cell populations are not
UC-MSCs reduce monocyte allostimulatory capacity—an
required for the MSC effect on monocytes. UC-MSC–modulated
effect partially mediated by UC-MSC PGE2 production and
monocytes were further assessed for their capacity to induce pro-
enhanced by activation
liferation of allogeneic CD4+ T cells. CD14+ cells purified from
nonactivated PBMNC and UC-MSC coculture had a reduced ability PGE2 has complex and often divergent actions on different cells
to induce proliferation of allogeneic T cells (Fig. 3C). In addition, of the immune system (28). We therefore addressed whether UC-
monocytes purified from highly pure CD14+ and UC-MSC co- MSC–derived PGE2 had a role in modulating monocyte allosti-
culture exhibited reduced allostimulatory capacity in two of three mulatory function. UC-MSCs were pretreated with indomethacin
experiments (data not shown). The data suggest that UC-MSCs may for 24 h prior to coculture with nonactivated PBMNCs or alloantigen-
interact directly with monocytes to rapidly impair their accessory activated PBMNCs. Treatment abolished constitutive PGE2 produc-
functions without any requirement for a third-party leukocyte subset. tion by UC-MSCs but did not affect PGE2 production in the sub-
sequent culture (data not shown). Indomethacin treatment of UC-

Downloaded from www.jimmunol.org on December 15, 2010


Inhibition of PGE2 production releases T cells from MSCs significantly reduced their ability to modulate monocyte allo-
UC-MSC–induced suppression stimulatory function compared with those purified from UC-MSCs
PGE2 has previously been identified as one of many MSC-derived treated with carrier (DMSO) alone. Indeed, monocytes purified from
soluble factors proposed to modulate immune function (2). PGE2 nonactivated PBMNC or alloantigen-activated PBMNC coculture
with indomethacin-pretreated UC-MSCs were able to stimulate al-
logeneic T cells to approximately a 2- and 4-fold greater extent, re-
spectively, than monocytes purified from nonactivated PBMNCs or
alloantigen-activated PBMNCs cultured with control UC-MSCs (Fig.
5A, 5B).
Inflammation has been proposed to “license” MSCs to suppress
immune function (29). We therefore compared the allostimulatory

FIGURE 3. Monocytes from PBMNC/UC-MSC coculture have reduced


accessory cell and allostimulatory function. A, CD4+ T cells were cultured
in medium alone (gray bar), with PHA (1 mg/ml) (hatched bar), with PHA
and CD14+ cells purified from a 24-h UC-MSC/nonactivated PBMNC
coculture (black bar), or with PHA and CD14+ cells purified from non-
activated PBMNC culture alone (white bar). Proliferation was measured by
[3H] uptake after 72 h of culture. The data shown are representative of five
independent experiments and shown as mean values 6 SD. B, CD4+ T cells
were cultured in medium alone (gray bar), with PHA (hatched bar), with
PHA and CD14+ cells purified from CD14+ cell/UC-MSC coculture after FIGURE 4. PGE2 mediates UC-MSC suppression of T cell proliferation.
24 h (black bar), or with PHA and CD14+ cells purified from CD14+ cells A, PGE2 was measured in the supernatant of PBMNCs (5 3 104) (spotted
in medium alone (white bar). Proliferation was measured by [3H] uptake bar, n = 2), MLR (2.5 3 104 CD4+ T cells + 2.5 3 104 CD2-depleted
after 72 h of culture. The data shown are representative of two independent APCs) (white bar, n = 3), PBMNC + UC-MSC (2 3 104) (black spotted
experiments and shown as mean values 6 SD. C, CD14+ cells were purified bar, n = 2), MLR + UC-MSC (black bar, n = 2), or UC-MSC alone (gray
from nonactivated PBMNCs cultured in medium alone (white bar, n = 2) or bar) cultures after 24 h. B, A total of 5 3 104 PBMNCs were cultured with
with UC-MSC (black bar, n = 2) for 24 h and cultured with allogeneic PHA (1 mg/ml) (white bar), with PHA + 2 3 104 UC-MSC + DMSO
CFSE-labeled CD4+ T cells. CD4+ T cell proliferation was assessed at 7 (black bar) or with PHA + 2 3 104 UC-MSC + 5 mM indomethacin
d by CFSE dye dilution and upregulation of CD25. The data are presented (hatched bar). Proliferation was measured by [3H] uptake after 72 h of
as proliferation induced by CD14+ cells relative to controls where pro- culture. The data shown are representative of five independent experiments
liferation is a reference at 100% and is shown as mean values 6 SD. and shown as mean values 6 SD.
The Journal of Immunology 6621

population to modulate their function and suppress T cell prolife-


ration.

Discussion
MSCs have been shown to modulate the actions of a diverse range
of immune cells and a number of soluble factors derived from, or
induced by MSCs have been proposed to mediate the anti-
inflammatory or suppressive effects of MSCs (2). Given the in-
creasing use of MSCs as a cellular therapy product in many
clinical trials it is of critical importance to elucidate the cellular
interactions required for MSCs to suppress immune responses and
to understand whether MSCs act directly on effector cells or via
a third party. In this study, we demonstrate that MSCs from di-
verse sources are dependent on monocytes to suppress T cell
proliferation induced by TCR cross-linking and that UC-MSCs act
via an indomethacin-sensitive agent PGE2. MSCs derived from
UC tissue rapidly alter monocyte phenotype, affect accessory cell
FIGURE 5. UC-MSC–derived PGE2 in part diminishes the allostimu- function, and fundamentally reduce their ability to activate and
latory capacity of monocytes. CD14+ cells were purified from (A) non- induce the proliferation of allogeneic T cells. The modulatory
activated PBMNCs cultured in medium alone (white bar, n = 8), effect of UC-MSCs on monocytes was enhanced in an activated
nonactivated PBMNC + UC-MSC coculture (black bar, n = 8) or non-

Downloaded from www.jimmunol.org on December 15, 2010


environment and was in part dependent on constitutive UC-MSC
activated PBMNC + indomethacin-pretreated UC-MSC coculture (hatched PGE2 production. Importantly, the modulatory effect of UC-MSC–
black bar, n = 3), (B) alloantigen-activated PBMNCs in medium alone
derived PGE2 was effective only during initial contact between
(white bar, n = 5), alloantigen-activated PBMNC + UC-MSC coculture
UC-MSCs and monocytes contained within nonactivated PBMNCs
(black bar, n = 5), or alloantigen-activated PBMNC + indomethacin-pre-
treated UC-MSC coculture (hatched black bar, n = 4), or (C) nonactivated or alloantigen-activated PBMNCs.
PBMNC (n = 8), nonactivated PBMNC + UC-MSC (n = 2), or PBMNC + Our data clearly implicate monocytes as a key intermediary in
225 nM PGE2 (n = 8) after 24 h of culture. Purified CD14+ cells were UC-MSC–induced suppression of T cell proliferation. This may
subsequently cultured with allogeneic CFSE-labeled CD4+ T cells at be a key interaction in vivo as peripheral blood and splenic
a ratio of 1:10, and the ability of purified monocytes to stimulate the monocytes are rapidly recruited to inflammatory sites (30, 31) and
proliferation of allogeneic CD4+ T cells was assessed by CFSE dye di- are able to differentiate into both inflammatory DCs and macro-
lution and upregulation of CD25 after 7 d. The data are presented as phages (21, 22). Once differentiated, both of these mononuclear
proliferation relative to medium controls where proliferation is a reference phagocytic cell populations play pivotal roles in promoting im-
at 100% and is shown as mean values 6 SD. Statistical significance was
mune responses and resolving inflammation depending on the
determined using the unpaired Student t test where *p , 0.05.
environmental cues (22, 32). BM-MSCs have been shown to alter
or block the maturation of monocytes into DCs (13, 14, 19, 26).
capacity of monocytes purified from a nonactivated PBMNC or However, in contrast to our studies, monocytes and BM-MSCs in
alloantigen-activated PBMNC coculture with UC-MSCs. The abil- earlier reports were cultured in the presence of GM-CSF and IL-4
ity of monocytes to stimulate an alloresponse from CD4+ T cells over 5–6 d in vitro to induce DC differentiation. Our data suggest
was more profoundly affected when purified from alloantigen- that monocyte function is directly and stably modulated by
activated PBMNC and UC-MSC coculture compared with those UC-MSCs within 24 h of coculture (Figs. 3, 5) and that MSCs
purified from nonactivated PBMNC and UC-MSC culture. Mono- from disparate sources all act through monocytes to reduce T cell
cytes stimulated allogeneic T cells on average 5-fold versus a 2-fold proliferation (Fig. 1B–D). Furthermore, the allostimulatory ca-
lesser extent, respectively (compare Fig. 5A, 5B). Importantly, pacity of highly purified CD14+ monocytes was greatly reduced
HLA-DR expression was equivalent on monocytes purified from following culture with UC-MSCs (data not shown). The data re-
all cultures at 24 h (Fig. 2B, 2C). Furthermore, pretreatment of inforce the notion that MSCs from a variety of sources primarily
UC-MSCs with indomethacin only allowed recovery of monocyte and directly act through monocytes to reduce T cell proliferation.
allostimulatory capacity to ∼50% of control values when purified In agreement, BM-MSCs have previously been shown to interact
from cocultures (Fig. 5B). Whereas monocytes purified from non- with monocytes to modulate immune responses through soluble
activated PBMNC/indomethacin-treated UC-MSC cultures recovered mediators (33) and to reduce monocyte accessory function in
allostimulatory capacity to ∼80% of controls (Fig. 5A). These data superantigen-stimulated cultures (18). It is of note that BM-MSCs
raise the possibility that inflammation may induce additional factors may in addition reduce mature DC function (14) and alter mac-
in MSCs and/or PBMNCs other than PGE2 to suppress T cell pro- rophage phenotype and function (20).
liferation. The phenotypic changes observed in the monocyte population
The effects of indomethacin indicated a role for PGE2 pro- suggest that UC-MSCs induce macrophages rather than a DC
duction by UC-MSCs in modulating the allostimulatory ability of subset. We found no evidence for induction of a CD14lo/neg CD1a,
monocytes. We therefore assessed whether PGE2 acted alone or CD1c, or CD83+ DC population or modulation of CD80 or CD86
required other factors to mediate this effect. PBMNCs were cul- on the CD14 positive population (data not shown). Although
tured with PGE2 at levels observed in nonactivated PBMNC and originally described as a DC-specific marker, DC-SIGN may be
UC-MSC coculture. Monocytes purified following 24 h of culture expressed by macrophages (34). CD123 (IL-3Ra) expression is
were then used as stimulator cells for allogeneic CD4+ T cells. associated with a plasmacytoid DC phenotype and was maintained
Monocytes from PGE2 cultures exhibited an increased capacity to following culture with UC-MSCs; however, plasmacytoid DCs do
stimulate allogeneic T cells compared with those from control not coexpress CD14 (35). Indeed, CD14 expression was signifi-
cultures (Fig. 5C). Therefore, our data suggest that additional fac- cantly increased initially in the presence of UC-MSCs but then
tors in concert with UC-MSC–derived PGE2 act on the monocyte declined over time dependent on the activation status of the cultures
6622 UC-MSCs MODULATE MONOCYTES TO SUPPRESS T CELL PROLIFERATION

(Fig. 2). In agreement, monocytes or CD14+ precursors cultured with (49, 50). The allostimulatory capacity of monocytes purified from
BM-MSC in DC differentiation conditions maintained expression of a MLR was significantly reduced compared with those purified
CD14 (13, 14) and were suggested to exhibit a macrophage-like from PBMNCs following coculture with UC-MSC (Fig. 5A, 5B).
morphology (13). IL-6 may play a key role in driving monocyte Our data support the idea that MSC suppression is enhanced in an
differentiation into macrophages (36). IL-6 is produced by UC-MSCs allogeneic environment. As discussed, IFN-g, PGE2, and IL-10 all
(data not shown) and in concert with M-CSF may (13, 14) or may not may play a role in MSC-induced suppression. However, in our
(26) mediate BM-MSC modulation of moDC differentiation. How- studies, induction of PGE2 and IL-10 secretion were equivalent in
ever, in our hands, neutralization of IL-6 and M-CSF had no effect on either nonactivated PBMNC or alloantigen-activated PBMNC
the ability of UC-MSCs to modulate monocyte phenotype (data not coculture with UC-MSCs, and neutralizing IL-10 in these cultures
shown). Finally, expression of CD73, a component of the immuno- did not affect the ability of UC-MSCs to modulate monocyte
suppressive adenosine pathway, is upregulated in the presence of UC- allostimulatory function (data not shown). Secretion of IFN-g,
MSC (Fig. 2B, 2C) and PGE2 (data not shown) and is induced on TNF-a, IL-6, and IL-1b were all induced following PBMNC
mature activated macrophages (37). culture with UC-MSCs but were not enhanced by stimulation (data
Macrophage and monocyte populations are heterogeneous with not shown). Therefore, as-yet unidentified factors are induced dur-
respect to the activatory and tissue environment guiding their ing UC-MSC and MLR coculture, which are able to further reduce
phenotype and function (23). UC-MSC upregulated expression of monocyte allostimulatory capacity and to enhance the suppressive
CD206 on CD14+ cells (Fig. 2B, 2C) a molecule frequently asso- capacity of UC-MSCs.
ciated with an alternatively activated macrophage phenotype (38). In conclusion, our data demonstrate that UC-MSCs require
CD206 was also induced on mature macrophages by BM-MSCs monocytes to suppress mitogen-stimulated T cell proliferation.
(20). Alternatively activated macrophage defined by their induction Monocyte phenotype and accessory and allostimulatory functions

Downloaded from www.jimmunol.org on December 15, 2010


by IL-4/13 signaling may induce tissue remodeling, regulate im- were stably and profoundly modulated following interaction with
mune responses (24) and inhibit T cell proliferation in vitro (39). UC-MSCs. UC-MSC PGE2 production, an indomethacin-sensitive
Indeed, DC-SIGN expression another molecule induced by UC- agent, appeared to play a key role in the suppression of T cell
MSCs (Fig. 2) may also be induced by IL-4 (40). However, IL-4 proliferation and in part the modulation of monocyte function.
was not detected following UC-MSC and PBMC cocultures (data Importantly, the influence of UC-MSC–derived PGE2 on mono-
not shown), and therefore from our data, we would not classify the cyte function was limited to the initial phase of contact. Finally,
induced macrophage population as alternatively activated. PGE2 treatment of PBMNCs alone could not reproduce a similar
PGE2 can exert opposing effects on the immune system (28). modulation of monocyte function. Therefore UC-MSC–derived PGE2
Depending on the context, PGE2 may either be pro- (41) or anti- acts at the initial contact between MSC and monocyte and in concert
inflammatory (42). BM-MSC–derived PGE2 has been proposed to with other factors to modulate monocyte function and suppress T cell
block DC differentiation (26) and induce monocyte IL-10 pro- proliferation.
duction (43). We and others (15) have shown that PGE2 pro-
duction may play a key role in UC-MSC–induced suppression of Acknowledgments
T cell proliferation (Fig. 4). PGE2 is constitutively produced by We thank the National Health Service Blood and Transplant for the provi-
BM-MSCs (27) and monocytes (44). Therefore to study the role sion of adult buffy coats and the Histocompatibility and Immunogenetics
of UC-MSC–derived PGE2 in modulating monocyte function, Department, Colindale, for assistance.
UC-MSCs were pretreated with indomethacin, thus reversibly
blocking constitutive UC-MSC PGE2 production (data not shown), Disclosures
prior to culture with immune cells. However, comparable amounts The authors have no financial conflicts of interest.
of PGE2 were detected in subsequent cultures with nonactivated,
alloantigen-activated PBMNCs or purified CD14+ cells whether
UC-MSCs were pretreated with indomethacin or not (data not References
1. Di Nicola, M., C. Carlo-Stella, M. Magni, M. Milanesi, P. D. Longoni,
shown). The data indicating, as previously described (27), a strong P. Matteucci, S. Grisanti, and A. M. Gianni. 2002. Human bone marrow
induction of de novo cyclooxygenase activity when MSCs are stromal cells suppress T-lymphocyte proliferation induced by cellular or non-
cultured with leukocytes. Despite this observation, monocytes pu- specific mitogenic stimuli. Blood 99: 3838–3843.
2. Le Blanc, K., and O. Ringdén. 2007. Immunomodulation by mesenchymal
rified from coculture with indomethacin-treated UC-MSC–stimu- stem cells and clinical experience. J. Intern. Med. 262: 509–525.
lated allogeneic CD4+ T cells to proliferate to a significant degree 3. Stappenbeck, T. S., and H. Miyoshi. 2009. The role of stromal stem cells in
above those from control UC-MSC cultures (Fig. 5). PGE2 treat- tissue regeneration and wound repair. Science 324: 1666–1669.
4. Lazarus, H. M., S. E. Haynesworth, S. L. Gerson, N. S. Rosenthal, and
ment of PBMNCs reproduced some but not all of the phenotypic
A. I. Caplan. 1995. Ex vivo expansion and subsequent infusion of human bone
changes induced by UC-MSCs on monocytes (data not shown). marrow-derived stromal progenitor cells (mesenchymal progenitor cells):
However, treatment of PBMNCs with PGE2 in isolation in some implications for therapeutic use. Bone Marrow Transplant. 16: 557–564.
instances enhanced the allostimulatory capacity of monocytes (Fig. 5. Le Blanc, K., F. Frassoni, L. Ball, F. Locatelli, H. Roelofs, I. Lewis, E. Lanino,
B. Sundberg, M. E. Bernardo, M. Remberger, et al. 2008. Mesenchymal
5C). Therefore, the absence of PGE2 production by UC-MSCs at the stem cells for treatment of steroid-resistant, severe, acute graft-versus-host dis-
initial stages of priming may therefore allow other mediators to “re- ease: a phase II study. Lancet 371: 1579–1586.
wire” monocytes to be either nonresponsive to PGE2 or alter their 6. Kode, J. A., S. Mukherjee, M. V. Joglekar, and A. A. Hardikar. 2009. Mesen-
chymal stem cells: immunobiology and role in immunomodulation and tissue
response to PGE2. regeneration. Cytotherapy 11: 377–391.
BM-MSCs have shown promise in the treatment of GVHD but 7. Bernardo, M. E., F. Locatelli, and W. E. Fibbe. 2009. Mesenchymal
only in patients with active disease (5, 45). MSCs share with stromal cells. Ann.N.Y. Acad.Sci. 1176: 101–117.
8. Ferrara, J. L., J. E. Levine, P. Reddy, and E. Holler. 2009. Graft-versus-host
monocytes the ability to home to sites of inflammation (46) and disease. Lancet 373: 1550–1561.
may enhance recruitment of macrophages to sites of injury (47). 9. Auffermann-Gretzinger, S., I. S. Lossos, T. A. Vayntrub, W. Leong,
However, depending upon the stimulus, the suppressive action of F. C. Grumet, K. G. Blume, K. E. Stockerl-Goldstein, R. Levy, and J. A. Shizuru.
MSCs may be enhanced or diminished by inflammation. Trig- 2002. Rapid establishment of dendritic cell chimerism in allogeneic hemato-
poietic cell transplant recipients. Blood 99: 1442–1448.
gering through TLRs inhibited MSC-suppressive function (48), 10. Haniffa, M., F. Ginhoux, X. N. Wang, V. Bigley, M. Abel, I. Dimmick,
whereas IFN-g enhanced the suppressive ability of BM-MSCs S. Bullock, M. Grisotto, T. Booth, P. Taub, et al. 2009. Differential rates of
The Journal of Immunology 6623

replacement of human dermal dendritic cells and macrophages during hemato- 32. Porcheray, F., S. Viaud, A. C. Rimaniol, C. Léone, B. Samah, N. Dereuddre-
poietic stem cell transplantation. J. Exp. Med. 206: 371–385. Bosquet, D. Dormont, and G. Gras. 2005. Macrophage activation switching: an
11. Matte, C. C., J. Liu, J. Cormier, B. E. Anderson, I. Athanasiadis, D. Jain, asset for the resolution of inflammation. Clin. Exp. Immunol. 142: 481–489.
J. McNiff, and W. D. Shlomchik. 2004. Donor APCs are required for maximal 33. Groh, M. E., B. Maitra, E. Szekely, and O. N. Koç. 2005. Human mesenchymal
GVHD but not for GVL. Nat. Med. 10: 987–992. stem cells require monocyte-mediated activation to suppress alloreactive T cells.
12. Geissmann, F., S. Jung, and D. R. Littman. 2003. Blood monocytes consist of Exp. Hematol. 33: 928–934.
two principal subsets with distinct migratory properties. Immunity 19: 71–82. 34. Granelli-Piperno, A., A. Pritsker, M. Pack, I. Shimeliovich, J. F. Arrighi,
13. Nauta, A. J., A. B. Kruisselbrink, E. Lurvink, R. Willemze, and W. E. Fibbe. C. G. Park, C. Trumpfheller, V. Piguet, T. M. Moran, and R. M. Steinman. 2005.
2006. Mesenchymal stem cells inhibit generation and function of both CD34+ Dendritic cell-specific intercellular adhesion molecule 3-grabbing nonintegrin/
-derived and monocyte-derived dendritic cells. J. Immunol. 177: 2080–2087. CD209 is abundant on macrophages in the normal human lymph node and is not
14. Jiang, X. X., Y. Zhang, B. Liu, S. X. Zhang, Y. Wu, X. D. Yu, and N. Mao. 2005. required for dendritic cell stimulation of the mixed leukocyte reaction. J.
Human mesenchymal stem cells inhibit differentiation and function of Immunol. 175: 4265–4273.
monocyte-derived dendritic cells. Blood 105: 4120–4126. 35. Olweus, J., A. BitMansour, R. Warnke, P. A. Thompson, J. Carballido,
15. Chen, K., D. Wang, W. T. Du, Z. B. Han, H. Ren, Y. Chi, S. G. Yang, D. Zhu, L. J. Picker, and F. Lund-Johansen. 1997. Dendritic cell ontogeny: a human
F. Bayard, and Z. C. Han. 2010. Human umbilical cord mesenchymal stem cells dendritic cell lineage of myeloid origin. Proc. Natl. Acad. Sci. USA 94: 12551–
hUC-MSCs exert immunosuppressive activities through a PGE2-dependent 12556.
mechanism. Clin. Immunol. 135: 448–458. 36. Chomarat, P., J. Banchereau, J. Davoust, and A. K. Palucka. 2000. IL-6 switches
16. Girdlestone, J., V. A. Limbani, A. J. Cutler, and C. V. Navarrete. 2009. Efficient the differentiation of monocytes from dendritic cells to macrophages. Nat.
expansion of mesenchymal stromal cells from umbilical cord under low serum Immunol. 1: 510–514.
conditions. Cytotherapy 11: 738–748. 37. Clifford, E. E., K. A. Martin, P. Dalal, R. Thomas, and G. R. Dubyak. 1997.
17. Manca, M. F., I. Zwart, J. Beo, R. Palasingham, L. S. Jen, R. Navarrete, Stage-specific expression of P2Y receptors, ecto-apyrase, and ecto-59-nucleo-
J. Girdlestone, and C. V. Navarrete. 2008. Characterization of mesenchymal tidase in myeloid leukocytes. Am. J. Physiol. 273: C973–C987.
stromal cells derived from full-term umbilical cord blood. Cytotherapy 10: 54– 38. Stein, M., S. Keshav, N. Harris, and S. Gordon. 1992. Interleukin 4 potently
68. enhances murine macrophage mannose receptor activity: a marker of alternative
18. Beyth, S., Z. Borovsky, D. Mevorach, M. Liebergall, Z. Gazit, H. Aslan, immunologic macrophage activation. J. Exp. Med. 176: 287–292.
E. Galun, and J. Rachmilewitz. 2005. Human mesenchymal stem cells alter 39. Schebesch, C., V. Kodelja, C. Müller, N. Hakij, S. Bisson, C. E. Orfanos, and
antigen-presenting cell maturation and induce T-cell unresponsiveness. Blood S. Goerdt. 1997. Alternatively activated macrophages actively inhibit pro-
105: 2214–2219. liferation of peripheral blood lymphocytes and CD4+ T cells in vitro. Immu-

Downloaded from www.jimmunol.org on December 15, 2010


19. Zhang, B., R. Liu, D. Shi, X. Liu, Y. Chen, X. Dou, X. Zhu, C. Lu, W. Liang, nology 92: 478–486.
L. Liao, et al. 2009. Mesenchymal stem cells induce mature dendritic cells into 40. Relloso, M., A. Puig-Kröger, O. M. Pello, J. L. Rodrı́guez-Fernández, G. de la
a novel Jagged-2–dependent regulatory dendritic cell population. Blood 113: 46– Rosa, N. Longo, J. Navarro, M. A. Muñoz-Fernández, P. Sánchez-Mateos, and
57. A. L. Corbı́. 2002. DC-SIGN (CD209) expression is IL-4 dependent and is
20. Kim, J., and P. Hematti. 2009. Mesenchymal stem cell-educated macrophages: negatively regulated by IFN, TGF-b, and anti-inflammatory agents. J. Immunol.
a novel type of alternatively activated macrophages. Exp. Hematol. 37: 1445– 168: 2634–2643.
1453. 41. Sakata, D., C. Yao, and S. Narumiya. 2010. Prostaglandin E2, an immunoacti-
21. Van Furth, R., M. C. Diesselhoff-den Dulk, and H. Mattie. 1973. Quantitative vator. J. Pharmacol. Sci. 112: 1–5.
study on the production and kinetics of mononuclear phagocytes during an acute 42. Scher, J. U., and M. H. Pillinger. 2009. The anti-inflammatory effects of pros-
inflammatory reaction. J. Exp. Med. 138: 1314–1330. taglandins. J. Investig. Med. 57: 703–708.
22. Geissmann, F., C. Auffray, R. Palframan, C. Wirrig, A. Ciocca, L. Campisi, 43. Németh, K., A. Leelahavanichkul, P. S. Yuen, B. Mayer, A. Parmelee, K. Doi,
E. Narni-Mancinelli, and G. Lauvau. 2008. Blood monocytes: distinct subsets, P. G. Robey, K. Leelahavanichkul, B. H. Koller, J. M. Brown, et al. 2009. Bone
how they relate to dendritic cells, and their possible roles in the regulation of marrow stromal cells attenuate sepsis via prostaglandin E2-dependent reprog-
T-cell responses. Immunol. Cell Biol. 86: 398–408. ramming of host macrophages to increase their interleukin-10 production. Nat.
23. Gordon, S., and P. R. Taylor. 2005. Monocyte and macrophage heterogeneity. Med. 15: 42–49.
Nat. Rev. Immunol. 5: 953–964. 44. Goodwin, J. S., A. D. Bankhurst, and R. P. Messner. 1977. Suppression of human
24. Martinez, F. O., L. Helming, and S. Gordon. 2009. Alternative activation of T-cell mitogenesis by prostaglandin: existence of a prostaglandin-produc-
macrophages: an immunologic functional perspective. Annu. Rev. Immunol. 27: ing suppressor cell. J. Exp. Med. 146: 1719–1734.
451–483. 45. Gonzalo-Daganzo, R., C. Regidor, T. Martı́n-Donaire, M. A. Rico, G. Bautista,
25. Kern, J. A., J. C. Reed, R. P. Daniele, and P. C. Nowell. 1986. The role of the I. Krsnik, R. Forés, E. Ojeda, I. Sanjuán, J. A. Garcı́a-Marco, et al. 2009. Results
accessory cell in mitogen-stimulated human T cell gene expression. J. Immunol. of a pilot study on the use of third-party donor mesenchymal stromal cells in
137: 764–769. cord blood transplantation in adults. Cytotherapy 11: 278–288.
26. Spaggiari, G. M., H. Abdelrazik, F. Becchetti, and L. Moretta. 2009. MSCs 46. Ortiz, L. A., F. Gambelli, C. McBride, D. Gaupp, M. Baddoo, N. Kaminski, and
inhibit monocyte-derived DC maturation and function by selectively interfering D. G. Phinney. 2003. Mesenchymal stem cell engraftment in lung is enhanced in
with the generation of immature DCs: central role of MSC-derived prostaglandin response to bleomycin exposure and ameliorates its fibrotic effects. Proc. Natl.
E2. Blood 113: 6576–6583. Acad. Sci. USA 100: 8407–8411.
27. Aggarwal, S., and M. F. Pittenger. 2005. Human mesenchymal stem cells 47. Chen, L., E. E. Tredget, P. Y. Wu, and Y. Wu. 2008. Paracrine factors of mes-
modulate allogeneic immune cell responses. Blood 105: 1815–1822. enchymal stem cells recruit macrophages and endothelial lineage cells and en-
28. Tilley, S. L., T. M. Coffman, and B. H. Koller. 2001. Mixed messages: modu- hance wound healing. PLoS ONE 3: 1886.
lation of inflammation and immune responses by prostaglandins and throm- 48. Liotta, F., R. Angeli, L. Cosmi, L. Filı̀, C. Manuelli, F. Frosali, B. Mazzinghi,
boxanes. J. Clin. Invest. 108: 15–23. L. Maggi, A. Pasini, V. Lisi, et al. 2008. Toll-like receptors 3 and 4 are expressed
29. Dazzi, F., and F. M. Marelli-Berg. 2008. Mesenchymal stem cells for graft- by human bone marrow-derived mesenchymal stem cells and can inhibit their
versus-host disease: close encounters with T cells. Eur. J. Immunol. 38: 1479– T-cell modulatory activity by impairing Notch signaling. Stem Cells 26: 279–289.
1482. 49. Krampera, M., L. Cosmi, R. Angeli, A. Pasini, F. Liotta, A. Andreini,
30. Randolph, G. J., K. Inaba, D. F. Robbiani, R. M. Steinman, and W. A. Muller. V. Santarlasci, B. Mazzinghi, G. Pizzolo, F. Vinante, et al. 2006. Role for in-
1999. Differentiation of phagocytic monocytes into lymph node dendritic cells terferon-g in the immunomodulatory activity of human bone marrow mesen-
in vivo. Immunity 11: 753–761. chymal stem cells. Stem Cells 24: 386–398.
31. Swirski, F. K., M. Nahrendorf, M. Etzrodt, M. Wildgruber, V. Cortez-Retamozo, 50. Polchert, D., J. Sobinsky, G. Douglas, M. Kidd, A. Moadsiri, E. Reina,
P. Panizzi, J. L. Figueiredo, R. H. Kohler, A. Chudnovskiy, P. Waterman, et al. K. Genrich, S. Mehrotra, S. Setty, B. Smith, and A. Bartholomew. 2008. IFN-g
2009. Identification of splenic reservoir monocytes and their deployment to in- activation of mesenchymal stem cells for treatment and prevention of graft
flammatory sites. Science 325: 612–616. versus host disease. Eur. J. Immunol. 38: 1745–1755.

You might also like