Sunteți pe pagina 1din 10

TISSUE-SPECIFIC STEM CELLS

Immune Properties of Human Umbilical Cord Wharton’s


Jelly-Derived Cells
MARK L. WEISS,a CAMERON ANDERSON,a SATISH MEDICETTY,c KIRAN B. SESHAREDDY,a RITA J. WEISS,a
IRENE VANDERWERFF,a DERYL TROYER,a KEVIN R. MCINTOSHb
a
Department of Anatomy and Physiology, Kansas State University, Manhattan, Kansas, USA; bCognate BioServices,
Baltimore, Maryland, USA; cNeoStem, Inc., Boston, Massachusetts, USA

Key Words. Adherent cells • Immunogenicity • Immunosuppression • In vitro culture • Stromal cell

ABSTRACT
Cells isolated from Wharton’s jelly, referred to as umbilical ternal-fetal interface. Fourth, the expression of CD40, CD80,
cord matrix stromal (UCMS) cells, adhere to a tissue-culture and CD86 was determined by flow cytometry. Fifth, the
plastic substrate, express mesenchymal stromal cell (MSC) cytokine expression of UCMS cells was evaluated by focused
surface markers, self-renew, and are multipotent (differen- gene array. The results indicate that human UCMS cells
tiate into bone, fat, cartilage, etc.) in vitro. These properties inhibit splenocyte proliferation response to concanavalin A
support the notion that UCMS cells are a member of the stimulation, that they do not stimulate T-cell proliferation
MSC family. Here, the immune properties of UCMS cells in a one-way MLR, and that they inhibit the proliferation
are characterized in vitro. The overall hypothesis is that of stimulated T cells in a two-way MLR. Human UCMS
UCMS cells possess immune properties that would be per- cells do not inhibit nonstimulated splenocyte prolifera-
missive to allogeneic transplantation. For example, UCMS tion, suggesting specificity of the response. UCMS cells
cells will suppress of the proliferation of “stimulated” lym- express mRNA for pan-HLA-G. UCMS cells do not ex-
phocytes (immune suppression) and have reduced immuno- press the costimulatory surface antigens CD40, CD80,
genicity (e.g., would be poor stimulators of allogeneic lym- and CD86. UCMS cells express vascular endothelial
phocyte proliferation). Hypothesis testing was as follows: growth factor and interleukin-6, molecules previously im-
first, the effect on proliferation of coculture of mitotically plicated in the immune modulation observed in MSCs. In
inactivated human UCMS cells with concanavalin-A-stimu- addition, the array data indicate that UCMS cells make a
lated rat splenocytes was assessed in three different assays. cytokine and other factors that may support hematopoi-
Second, the effect of human UCMS cells on one-way and esis. Together, these results support previous observa-
two-way mixed lymphocyte reaction (MLR) assays was de- tions made following xenotransplantation; for example,
termined. Third, the expression of human leukocyte antigen there was no evidence of frank immune rejection of un-
(HLA)-G was examined in human UCMS cells using reverse differentiated UCMS cells. The results suggest that hu-
transcription-polymerase chain reaction, since HLA-G ex- man UCMS will be tolerated in allogeneic transplanta-
pression conveys immune regulatory properties at the ma- tion. STEM CELLS 2008;26:2865–2874
Disclosure of potential conflicts of interest is found at the end of this article.

stimuli [4 – 6]. MSCs suppress lymphocyte proliferation in vitro


INTRODUCTION and prolong skin graft survival [4]. Two different general mech-
anisms, one cell contact-dependent and one -independent, have
The Mesenchymal and Tissue Stem Cell Committee of the Inter- been advanced to explain this immunosuppression capability
national Society for Cell Therapy has suggested three defining [7]. The effect is most likely due to soluble factors secreted by
criteria for mesenchymal stromal cells (MSCs). MSCs (a) are a the MSCs [8], such as transforming growth factor ␤-1, hepato-
plastic-adherent cell population, (b) have specific surface markers cyte growth factor, interleukin-6, prostaglandin E2, indoleamine
(e.g., CD14-, CD34-, CD45-, and human leukocyte antigen [HLA] 2,3-dioxygenase-mediated tryptophan depletion, or nitric oxide
class II-negative and CD73-, CD29-, CD105-positive), and (c) (reviewed in [9]). MSCs downregulate the interleukin (IL)-2
have the capacity to self-renew and differentiate into various lin- receptor (CD25) and CD38 on phytohemagglutinin-activated
eages, including bone, cartilage, and adipose, in vitro [1]. lymphocytes [10]. Dendritic cells (DCs) cocultured with MSCs
MSCs possess the immune properties of immune suppres- develop into tolerogenic DCs [11].
sion and immune avoidance (reviewed in [2, 3]). MSCs exert In addition to MSCs derived from bone marrow, MSCs from
immunosuppression by inhibiting T-cell responses to polyclonal other tissues are reported to be immunosuppressive (e.g., MSCs

Author contributions: M.L.W.: conception and design, assembly of data, data analysis and interpretation, manuscript writing and final
approval of manuscript, financial and administrative support; C.A. and K.R.M.: design, assembly of data, data analysis and interpretation,
manuscript writing; S.M., K.B.S., R.J.W., and I.V.: collection and assembly of data; D.T.: administrative support, data interpretation.
Correspondence: Mark L. Weiss, Ph.D., Department of Anatomy and Physiology, Kansas State University, Manhattan, Kansas 66506, USA.
Telephone: 785-532-4520; Fax: 785-532-4557; e-mail: weiss@vet.ksu.edu Received December 10, 2007; accepted for publication July 28,
2008; first published online in STEM CELLS EXPRESS August 14, 2008. ©AlphaMed Press 1066-5099/2008/$30.00/0 doi: 10.1634/
stemcells.2007-1028

STEM CELLS 2008;26:2865–2874 www.StemCells.com


2866 Immune Properties of UCMS Cells

from dental pulp [12], adipose tissue [13, 14], and amniotic focused gene array. The results indicate that UCMS cells are
tissue [15]). The immune-suppressive property of MSCs has immunosuppressive, have reduced immunogenicity, express
been exploited for the successful treatment of severe human HLA-G, do not express costimulatory molecules, and express
graft-versus-host disease [16]. cytokines that may modulate immune function. Together, these
MSCs have immune avoidance mechanisms that reduce results suggest that human UCMS will be tolerated in allogeneic
immunogenicity. Reduced immunogenicity is suggested by the transplantation.
lack of acute rejection response by the host following xenoge-
neic transplantation into immune-competent animals [17, 18].
Mechanisms that may allow MSCs to escape from the immune MATERIALS AND METHODS
system in allogeneic hosts include modulation of host dendritic
and T-cell function and promotion of regulatory T-cell induc-
Approvals
tion, as well as limited expression of alloantigen [19]. Low
immunogenicity is also suggested to be due to the induction of The Kansas State University Institutional Review Board reviewed
divisional anergy in T cells [6], as well as secretion of the and approved this protocol. The Kansas State University Institu-
tional Animal Care and Use Committee reviewed and approved
soluble factors mentioned above by MSCs [19]. Coculture of
animal use.
bone marrow MSCs with T lymphocytes downregulated T-cell
alloresponsiveness via the TH2 path and resulted in the T cells Human Umbilical Cord Matrix Stromal Cells
assuming a T-regulatory phenotype [20]. Adipose-derived
MSCs demonstrate reduced immunogenicity [13, 14]. Thus, the The protocol for isolation and characterization of umbilical cord
immune properties described above are characteristics of MSCs matrix stromal cells has been previously described [22, 24]. Using
previously described protocols, a subset of the isolates was charac-
derived from several tissue sources. terized to confirm that they met the ISCT minimal marker set for
We identified a population of CD45-, CD34-, and HLA-DR- MSCs (e.g., they were shown to be plastic-adherent and to express
negative and CD73-, CD105-, CD90-, and CD29-positive cells surface markers CD105, CD90, and CD44) and that they did not
derived from human umbilical cord Wharton’s jelly, termed express surface markers CD34 and CD45, and they were verified to
umbilical cord matrix stromal (UCMS) cells [21–24]. Human differentiate along two or three mesenchymal lineages (e.g., chon-
UCMS cells can be isolated rapidly in large numbers from drogenic, osteogenic, and adipogenic lineages) [22, 28, 29, 36].
⬎90% of human cords. Thus, UCMS cells may be a robust
source of MSC-like cells for therapeutic use since they can be Splenocyte Proliferation Assay
frozen/thawed, clonally expanded, engineered to express exog- Rat splenocytes were obtained from euthanatized rats using stan-
enous proteins, and extensively expanded in culture. We spec- dard protocols. Briefly, the spleen was removed aseptically, washed
ulate that UCMS cells have therapeutic potential since they thoroughly, and diced, and spleen cells were released by trituration.
resemble MSCs in many respects, including surface phenotype, The splenocyte preparation was cleaned up by centrifugation and
plastic adherence, and multipotency; in the latter case, they can resuspending the pellet in RPMI 1640 (Invitrogen, Carlsbad, CA,
be induced in vitro to become bone, cartilage, adipose cells [22, http://www.invitrogen.com), and the erythrocytes were lysed. The
25–30], neural cells [22, 30, 31], skeletal muscle cells [25], and, splenocyte preparation was again centrifuged, and splenocytes
were washed two more times with RPMI. Cell count and viability
possibly, types of muscle cells [30, 32]. Following xenotrans-
were assessed by trypan blue exclusion prior to plating. Cells were
plantation of pig or human UCMS cells into immune-competent resuspended in splenocyte medium (i.e., RPMI containing 10% fetal
rats, little to no host immune cell infiltration was observed, bovine serum [FBS; HyClone, Logan, UT, http://www.hyclone.
suggesting low immunogenicity of UCMS cells [21, 23, 24]. com], 2 mM glutamine, 0.1 mM nonessential amino acids, 1 mM
Our hypothesis is that UCMS cells have immune properties pyruvate, 20 mM HEPES, and 5 ⫻ 10⫺5 M 2-mercaptoethanol).
that are permissive to allogeneic transplantation. MSCs derived Human umbilical cord matrix stromal (hUCMS) cells were plated in
from bone marrow [2, 4, 5, 16, 33, 34] and adipose tissue [13, a 12-well plate in low-serum medium (as described previously). At
14] are immune-suppressive and have low immunogenicity in 70% confluence, the medium was removed, and fresh medium
vitro. Our hypothesis would be supported if UCMS cells share containing mitomycin-C (20 ␮g/ml) was added for 2 hours at 37°C
these properties. to mitotically inactivate the hUCMS cells, followed by two washes
Furthermore, in some tissues, such as the chorionic plate of and the addition of low-serum medium. In some cases, the hUCMS
cells were inactivated by irradiation (7–9 Gy; Kansas State Univer-
the placenta and perhaps parts of the umbilical cord, additional sity CVM linear accelerator). Several hours later or the next day, the
immune avoidance mechanisms are found, because the early medium was removed from the inactivated hUCMS cells, and 1 ml
embryo must avoid immune-based destruction, especially in of splenocyte medium containing 5 ⫻ 104 concanavalin A (Con-
hemochorial species. One well-characterized mechanism is the A)-treated (10 ␮g/ml) splenocytes was added to three wells with
expression by the trophoblasts of a nonclassic HLA class I gene, hUCMS cells and three wells without hUCMS cells (each trial was
HLA-G. Six or seven splice variants of HLA-G have been done in triplicate, and each trial was performed with a different
identified: HLA-G1 through HLA-G4 are membrane-bound iso- splenocyte isolate).
forms; HLA-G5 and HLA-G6 are soluble forms. The soluble
forms of HLA-G have been shown to have immunoregulatory Assessing Splenocyte Proliferation
functions, including inhibition of T-cell activation [35]. Al- Splenocyte proliferation was assessed three different ways. First,
though HLA-G expression is downregulated and not found in live splenocytes were counted using a hemocytometer 3 days after
the fetus, its expression in the umbilical cord is unknown. The plating using trypan blue exclusion. Live cell splenocyte counts
hypothesis would be supported if UMCS cells express HLA-G. were done in triplicate and averaged for each well. Here, the effect
Here, the immune properties of human UCMS cells were of hUCMS cell coculture on splenocyte proliferation was evaluated
assessed in vitro using standard assays, including splenocyte by comparing the number of viable splenocytes in wells cocultured
with inactivated hUCMS cells with the number of viable spleno-
proliferation assays and one-way and two-way mixed lympho-
cytes in control wells (wells without an hUCMS cell layer; data
cyte assays, and their expression of HLA-G isoforms were were averaged from five independent trials).
assessed using reverse transcription (RT)-polymerase chain re- Second, in completely independent experiments, a colorimetric
action (PCR). The expression of costimulatory molecules CD40, assay using tetrazole reduction was used to assess splenocyte pro-
CD80, and CD86 by UCMS cells was characterized by flow liferation following Con-A stimulation using the manufacturer’s pro-
cytometry, and their expression of cytokines was examined by tocols (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium [MTT]
Weiss, Anderson, Medicetty et al. 2867

Figure 1. Immune suppression by coculture of human umbilical cord matrix stromal (hUCMS) cells with ConA-activated splenocytes. (A): Using
the trypan blue method to determine the number of live cells, the average suppression in splenocyte growth when cocultured with hUCMS cells was
40%. (B): Using the MTT assay, the average suppression after 4 days was 25%. Thus, both methods indicated immune suppression by coculture with
mitotically inactivated hUCMS cells. Data are presented as means ⫾ SE. Statistically significant differences are indicated. Abbreviations: ConA,
concanavalin A; MTT, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium; UCMS, umbilical cord matrix stromal.

assay; Roche Diagnostics, Basel, Switzerland, http://www.roche- Flow Cytometry


applied-science.com). The plate was evaluated on an enzyme-linked
Three different hUCMS cell isolates (isolates 19, 12, and 21) were
immunosorbent assay reader at 550 – 690 nm. The effect of hUCMS
assessed via flow cytometry using standard methods described
cells on splenocyte proliferation was evaluated by comparing the
previously [24]. Briefly, approximately 1–2 ⫻ 106 cells were sus-
optical density (OD) in wells cocultured on inactivated hUCMS
pended in 2 ml of PBS containing 0.5% bovine serum albumin and
cells with the OD of splenocytes grown without an inactivated
2 mM EDTA. Aliquots of cells (100 ␮l) were incubated with
hUCMS cell layer (performed in triplicate and averaged from four
labeled antibody (or isotype control) for 30 minutes at 4°C then
independent trials).
washed twice by centrifugation. The cells were protected from light
Third, in independent trials, the effect of coculture with inacti-
and held at 4°C until analysis using a FACSCalibur. The antibodies
vated hUCMS cells on Con-A-stimulated splenocytes was assessed used here can be found in supplemental online Table 3.
followed dye-loading of splenocytes with carboxyfluorescein suc-
cinimidyl ester (CFSE). Briefly, splenocytes were incubated with 5
␮M CFSE for 10 minutes followed by two washes with phosphate-
Focused Array
buffered saline (PBS). The CFSE-labeled splenocytes were equally Nonradioactive GeArray cDNA expression array filters (OHS-022;
divided into four experimental groups: splenocytes alone (no Con-A SuperArray Bioscience Corporation, Frederick, MD, http://www.
stimulation), splenocytes cultured with mitotically inactivated superarray.com) were used to evaluate cytokine gene expression,
hUCMS cells in splenocyte medium (no Con-A), splenocytes stim- and procedures were as described by the manufacturer. The meth-
ulated with Con-A (10 ␮g/ml; no hUCMS cells), and splenocytes odological details are provided in the supplemental online data.
stimulated with Con-A cultured with inactivated hUCMS cells.
Twenty-four or 48 hours later the splenocytes were lifted, washed Statistical Analysis
with PBS, fixed with 4% paraformaldehyde, rinsed twice with PBS, Data collection was conducted in an experimenter-blind fashion,
and stored at 4°C until flow cytometry (FACSCalibur; BD Bio- when possible. Following data collection, the group status was
sciences, San Diego, http://www.bdbiosciences.com). The flow cy- decoded prior to statistical analysis. In general, the Student’s t test
tometry data were analyzed using ModFit (Verity Software House, was used to evaluate the differences in the means between groups,
Topsham, ME, http://www.vsh.com) to estimate the proliferation of and significance was set at p ⬍ .05. For the CFSE experiment,
the splenocytes. To compare the effects of the experimental vari- significance was set at p ⬍ .1. Data are presented as mean ⫾ 1 SEM
ables, the proliferation index, a statistic generated by ModFit that or mean ⫾ 1 SD, as noted.
relates to the number of population doublings the splenocytes had
undergone following CFSE loading, was used.
In each case, a paired t test was used to compare means of RESULTS
experimental groups to evaluate whether Con-A exposure increased
splenocyte proliferation (CFSE experiment; one-tailed test) and
whether culture with UCMS cells inhibited Con-A induced spleno- Characterization of hUCMS Cells
cyte proliferation (all three experiments; one-tailed test). A two- Human UCMS cells were rapidly extracted from the umbilical
tailed test was used to evaluate whether culture of UCMS cells with
cord and grown in culture. As shown in supplemental online
splenocytes affected splenocyte proliferation (compared with
splenocytes only; CFSE experiment). Table 1, the umbilical cord isolates yielded an average of
approximately 10,500 cells per centimeter of umbilical cord
length in the initial isolate; this fits within the 10,000 –15,000
Mixed Lymphocyte Proliferation Assays cells per centimeter of length that has historically been obtained
The mixed lymphyocyte proliferation assays were carried out using in our laboratory [37, 38]. Here, a randomly selected subset of
standard methods. The methodological details are found in the the isolates was fully characterized by plastic adherence, flow
supplemental online data. cytometry, and differentiation into mesenchymal lineages: os-
teogenic, chondrogenic, and adipogenic lineages (supplemental
RT-PCR online Table 1). Thus, hUCMS cells used here the minimal
RT-PCR was carried out using standard methods. The methodolog- definition of MSCs given by ISCT [1], as previously described
ical details are found in the supplemental online data. by our laboratory and others [22, 28, 29, 36].
www.StemCells.com
2868 Immune Properties of UCMS Cells

Figure 2. Immune suppression by coculture of human umbilical cord matrix stromal cells (hUCMS cells) with ConA-activated splenocytes assessed
by the CFSE method. Splenocyte proliferation was assessed after 4 days of coculture with hUCMS cells and analyzed by flow cytometry. The
proliferation index (PI; a statistic generated by ModFit) correlated to the number of cell divisions the splenocytes had undergone. (A): Data from one
of the three independent trials. Top left: Splenocytes stimulated with ConA proliferated extensively (PI, 3.63; no coculture). Top right: Coculture of
stimulated splenocytes with inactivated hUCMS cells decreased splenocyte proliferation (PI, 2.63). Bottom left: Coculture of unstimulated splenocytes
with hUCMS cells tended to increase, and did not decrease, unstimulated splenocyte proliferation (compare PI of this panel, 1.57, with the PI of
splenocytes alone [bottom right], 1.46). Bottom right: Unstimulated splenocytes alone (PI, 1.46). (B): The data were normalized to the PI of
splenocytes only (100%). Shown are averaged results from three independent trials. Splenocyte proliferation was significantly enhanced by ConA (p ⬍
.001). In contrast, culture of inactivated hUCMS cells with ConA-stimulated splenocytes significantly suppressed splenocyte proliferation (p ⬍ .08).
The immune suppression effect of inactivated hUCMS cells was specific to stimulated splenocytes, and coculture with inactivated hUCMS cells was
not in itself harmful to splenocytes because coculture of hUCMS cells did not affect proliferation of splenocytes (p ⬎ .1). Data are presented as
means ⫾ SE. Statistically significant differences are indicated with an asterisk (ⴱ). Abbreviations: CFSE, carboxyfluorescein succinimidyl ester;
ConA, concanavalin A; NS, not significant; UCMS, umbilical cord matrix stromal.

Splenocyte Proliferation Assay Results .01). The coculture of Con-A-stimulated splenocytes with mi-
Both the trypan blue (shown in Fig. 1A) and MTT assay (Fig. totically inactivated hUCMS cells produced a proliferation in-
1B) methods indicated significant suppression of Con-A-in- dex of 3.80 ⫾ 1.08. This represents a significant suppression of
duced splenocyte proliferation by coculture with mitotically activated splenocyte proliferation (p ⬍ .08). To evaluate
inactivated human UCMS cells. The average suppression in whether coculture of splenocytes with inactivated UCMS cells
splenocyte growth when cocultured with UCMS cells was 40% produced a nonspecific inhibition of splenocyte proliferation,
(Fig. 1A). Using the MTT assay, immune suppression ranged the proliferation index was assessed following coculture of
from 8% to 69% after 3 days and from 15% to 55% after 4 days. unstimulated (no Con-A stimulation) splenocytes with inacti-
The average suppression was 30% after 3 days and 25% after 4 vated hUCMS cells. In this case, the proliferation index was
days (Fig. 1B). Thus, both methods indicated significant sup- 2.39 ⫾ 1.38. Thus, splenocyte proliferation is mildly stimulated
pression of splenocyte proliferation by coculture with inacti- (and not significantly stimulated) and was not inhibited by
vated hUCMS cells. The MTT assay results indicated that coculture with human UCMS cells. This observation suggests
suppression was 25% after 4 days versus 40% suppression of that the suppressive effect of hUCMS cells on splenocytes is
proliferation obtained by manual counting after 4 days. specific to mitogen-induced splenocyte proliferation. These re-
The CFSE experiment was conducted to evaluate splenocyte sults provide independent confirmation of the data obtained
proliferation and to evaluate the specificity of the effect of from by manual counting and by MTT assays and extend those
hUCMS on activated splenocytes. The results are shown in findings to indicate that (a) hUCMS cells lack toxicity on
Figure 2 and are similar to those obtained using the other splenocytes when cocultured and (b) the immune-suppressive
methods; for example, they indicate a suppression of Con-A- effects of UCMS cells are specific to splenocytes stimulated by
induced splenocyte proliferation by coculture with mitotically Con-A.
inactivated hUCMS cells (Fig. 1). Specifically, splenocytes
grown in culture without the mitogen Con-A yielded a prolif- One-Way Mixed Lymphocyte Reaction
eration index of 2.17 ⫾ 0.77. Stimulation with Con-A signifi- (Immunogenicity Assay) Results
cantly increased the proliferation of splenocytes (proliferation The data shown in Figure 3A and 3B are derived from indepen-
index, 5.38 ⫾ 2.17). This indicates that the splenocytes prolif- dent experiments performed on different cell isolates. In both
erate in response to Con-A stimulation (positive control, p ⬍ cases, the experiments were performed the same way. As indi-
Weiss, Anderson, Medicetty et al. 2869

Figure 3. Immunogenicity of human um-


bilical cord matrix stromal (hUCMS) cells
by one-way mixed lymphocyte reaction as-
say. Four different hUCMS cell isolates at
early (P5–P6) or later (P9) passage were
tested for their ability to stimulate prolifera-
tion of allogeneic T cells derived from two
different donors, responder 008 and re-
sponder 009. The proliferation of T cells was
determined in the presence of autologous
irradiated PBMCs (negative control, back-
ground), in the presence of allogeneic irra-
diated PBMCs (positive control), or in the
presence of UCMS cells. Results are shown
for hUCMS donors 4-3 and 4-5 in (A) and
for hUCMS donors 4-6 and 4-7 in (B). The
stimulator cells were tested at densities of
5,000, 10,000, or 20,000 cells per well. Only
the highest cell dose is shown here since the
lower doses of positive control allogeneic
PBMCs induced poor T-cell proliferation.
Data are presented as mean counts per
minute ⫾ SD. An asterisk (ⴱ) indicates sig-
nificant T-cell stimulation above back-
ground responses by criteria described in
Materials and Methods. Abbreviations: P,
passage; PBMC, peripheral blood mononu-
clear cell; UCMS, umbilical cord matrix
stromal.

cated in Figure 3A and 3B, the purified T-cell response to 19%– 49%). This dose of cells corresponds to a 1:40 ratio of
hUCMS cell isolates derived from four donors was not signif- hUCMS to responder cells.
icantly higher than the response to autologous peripheral blood The immune suppression data from both sets of experiments
mononuclear cells (PBMCs), indicating that hUCMS cells did is summarized in Figure 5, comparing passage number of
not elicit a proliferation response in vitro. Importantly, the UCMS cells. In two of three isolates (4-3, 4-5) suppression
response of all four isolates did not change with passage: neither decreased with increased passage by approximately 19%. Im-
early passage (P5 or P6) nor later passage (P9) cells elicited an mune suppression by the third hUCMS cell isolate (4-7) was
appreciable T-cell proliferation response. In contrast, vigorous unchanged by passage. Overall suppression by early passage
T-cell responses were detected to allogeneic PBMCs (the pos- (P5–P6) hUCMS cells was 37% ⫾ 14%, and suppression by late
itive control). passage (P9) cells was 33% ⫾ 15%. The immune suppression
observed by allogeneic MLR (approximately 35%) was similar
Two-Way Mixed Lymphocyte Reaction (Immune to that observed by the xenogeneic splenocyte proliferation
Suppression) Results assays (Figs. 1 and 2, above).
The data shown in Figure 4A and 4B were derived from inde- RT-PCR Results
pendent experiments performed on different cell isolates. In Pan-HLA-G primers indicated that HLA-G mRNA was ex-
both cases, the experiments were performed the same way. As pressed by hUCMS cells at both passage 4 and passage 8 (data
shown in Figure 4A and 4B, all four hUCMS cell isolates not shown). As shown in Figure 6, hUCMS cells differed from
suppressed the mixed lymphocyte reaction (MLR) response to the term placenta in terms of the expression of mRNA for
various degrees, generally in a dose-dependent manner. In the HLA-G6 isoforms. Human UCMS cells weakly expressed
first set of experiments (Fig. 4A), control splenic fibroblasts HLA-G6 and did not express HLA-G5 (Fig. 6). In contrast, term
were not suppressive at the highest dose of 20,000 cells per well. placenta expressed both HLA-G5 and HLA-G6. Data from the
At the same dose, hUCMS cells suppressed the MLR by an same PCR experiment depicting results from the JAR p745
average of 35% ⫾ 14% (range, 18%–53%). This dose of cells negative control cells, and the jeg-3 HLA-G positive controls
corresponds to a 1:10 ratio of hUCMS to responder cells, are shown as well.
assuming that half of the PBMCs are T cells. In the second set
of experiments (Fig. 4B), splenic fibroblasts were suppressive at Flow Cytometry Results
10,000 cells per well (34% suppression) but not at 5,000 cells As shown in Table 1, three different isolates of hUCMS cells
per well (8% suppression). At 5,000 cells per well, hUCMS cells tested at passage 4 did not express costimulatory molecules
suppressed the MLR by an average of 36% ⫾ 15% (range, (e.g., CD40, CD80, or CD86).
www.StemCells.com
2870 Immune Properties of UCMS Cells

Figure 4. Suppression of two-way MLR


by human umbilical cord matrix stromal
(hUCMS) cells. Four different hUCMS cell
isolates were tested at early (P5 or P6) or
later (P9) passage for their ability to sup-
press T-cell proliferation in a two-way MLR.
hUCMS cells or splenic fibroblasts were
added to MLR cultures at the numbers indi-
cated, ranging from 5,000 to 20,000 cells per
well. T-cell proliferation, expressed as cpm,
was determined 7 days later by pulsing the
cells with [3H]thymidine during the final 16
hours of culture. Data are represented as
mean counts per minute ⫾ SD. Significant
suppression (p ⬍ .05) of the MLR by
hUCMS cells is denoted by an asterisk for
the blocking of responses at the highest dose
of splenic fibroblasts that did not mediate
significant suppression. This dose was
20,000 cells per well in (A) and 5,000 cells
per well in (B). Abbreviations: MLR, mixed
lymphocyte reaction; P, passage; UCMS,
umbilical cord matrix stromal.

DISCUSSION
Here, the first description of the immune properties of Whar-
ton’s jelly-derived cells (here called hUCMS cells) was pro-
vided. Five observations were reported. First, human UCMS
cells suppressed the proliferation of stimulated immune cells.
Con-A-stimulated rat splenocytes (xenograft model) or acti-
vated human peripheral blood mononuclear cells (allogeneic
transplant model) were cocultured with human UCMS cells.
Second, human UCMS cells did not stimulate proliferation of
Figure 5. Suppression summary. The data from Figure 4 are sum- allogeneic or xenogeneic immune cells. Human UCMS cells
marized and expressed as percent age of suppression of the control were cocultured with two different isolates of purified T cells,
mixed lymphocyte reaction response. The data used for this figure
were derived from doses of cells added in which fibroblasts were not and no proliferation of the T cells was found. When human
suppressive: 20,000 (isolates 4-3 and 4-5) and 5,000 (isolates 4-6 and UCMS cells were cocultured with rat splenocytes, no prolifer-
4-7) cells per well. Abbreviations: ND, not done; P, passage; UCMS, ation was observed. Third, RT-PCR indicated that hUCMS cells
umbilical cord matrix stromal. produced an immunosuppressive isoform of human leukocyte
antigen (HLA), HLA-G6. Fourth, flow cytometry revealed that
the expression of immune response-related surface antigens
Focused Array Results CD40, CD80, and CD86 was absent on hUCMS cells. Fifth, the
As shown in supplemental online Fig. 1, cytokines implicated in presence of mRNA for cytokine genes by hUCMS cells was
hematopoietic stem cell proliferation, cell proliferation factors, evaluated, and it indicated that hUCMS cells make several
cytokines, such as IL-6 and VEGF. Together, these results
or rescue functions or with specific cytokine activity were
indicate that human UCMS cells have immunosuppressive prop-
expressed by hUCMS cells. Specifically, mRNA for interleu- erties in vitro and that human UCMS cells have low immuno-
kins 1A, 1B, 6, 8, 11, and 14 was expressed by hUCMS cells. In genicity. These findings indicate that UCMS cells that have
addition, mRNA was present for BMP1, CSF3, FAM3C, been expanded for four to nine passages in vitro appear to have
GDF15, PDGFB, and tumor necrosis factors 4, 11b, 12, and the same immune properties as MSCs derived from other tissue
vascular endothelial growth factor (VEGF) was expressed by sources. Furthermore, these results suggest that UCMS cells
hUCMS cells (supplemental online Fig. 1). would be tolerated in an allogeneic transplant.
Weiss, Anderson, Medicetty et al. 2871

multipotent and can be differentiated into bone, cartilage, fat,


and neural cells [28 –30, 40].
It is important to note that in vitro data must be confirmed
with in vivo data. Although in vitro data appear to be fairly
consistent among laboratories (showing suppression and low
immunogenicity of MSCs), the animal data have been contra-
dictory. Recently, it has been shown that porcine MSCs, which
appeared to have low immunogenicity in vitro, elicited an im-
mune response after intracardiac transplantation into allogeneic
recipients, although subcutaneous transplants required a second
injection to effect a similar reaction [41]. However, multiple
injections of large numbers of allogeneic baboon MSCs (1 ⫻
106 cells per kilogram of body weight) via multiple routes
resulted in significant engraftment and the induction of host
T-cell hyporesponsiveness, although all recipient baboons pro-
duced alloantibodies [42]. In contrast, it was reported that mul-
tiple injections of allogeneic swine UCMS cells could elicit an
immune response [43]. This change is likely due to changes in
major histocompatibility complex (MHC) class II expression,
since human or swine UCMS cells could be induced to express
MHC class II following a 48-hour exposure to interferon-␥ in
culture [43]. Similarly, allogeneic swine UCMS cells activated
by interferon-␥ exposure were immunogenic upon their first
injection into swine, and nonactivated UCMS cells were not.
These results do not discount the idea of using hUCMS cells in
an allogeneic setting and suggest that care may be needed when
multiple injections of UCMS cells are given.
Work from several laboratories indicates that UCMS cells
have therapeutic potential for neurodegenerative disease [24,
Figure 6. Reverse transcription polymerase chain reaction (RT-PCR)
27], stroke [44], photoreceptor degeneration [40], and breast
for HLA-G isoforms. RT-PCR for HLA-G5 and HLA-G6 isoforms
revealed that human umbilical cord matrix stromal cells express mRNA cancer [45]. Finally, UCMS cells may be useful for tissue
for HLA-G6. Positive controls were human term placenta and jeg- engineering [25, 29, 46 – 49]. These factors together suggest that
HLA-G cells. Negative controls were JARp745 cells. Abbreviations: hUCMS cells may be an important source of cells for cell
GAPDH, glyceraldehyde-3-phosphate dehydrogenase; HLA, human therapy in the future. The ability to use UCMS cells in alloge-
lymphocyte antigen; P, passage; UCM, umbilical cord matrix. neic transplantation would enable their therapeutic use. For
example, this would simplify the “stock” of cryogenically stored
Table 1. Flow cytometry results
UCMS cells required for off-the-shelf therapeutic use.
FACS hUCMS 21, hUCMS 12, hUCMS 9, Do MSCs Have Specific Immune Properties?
antibody passage 4 passage 4 passage 4
In the Introduction, the minimal marker set of MSCs was given
CD40 2% ND ND
as plastic adherence, a particular surface marker set, and mul-
CD80 ND ND ND
CD86 ND ND ND tipotency. MSCs may have additional properties. For example,
it is well documented that (a) MSCs act as support cells for
Abbreviations: FACS, fluorescence-activated cell sorting; hematopoietic stem cells [50, 51], (b) they migrate to sites of
hUCMS, human umbilical cord matrix stromal; ND, not detected.
pathology and infiltrate tumors [52, 53], and (c) they may have
particular immune properties of low immunogenicity and im-
Properties of UCMS Cells mune suppression [2, 4, 5, 34]. A similar set of immune prop-
erties is reported here with UCMS cells. Furthermore, the cur-
The umbilical cord matrix, also known as Wharton’s jelly, is the rent work suggests that the UCMS cells act via to suppress
loose connective tissue that surrounds and cushions the umbil- proliferation of activated splenocytes, specifically in allogeneic
ical vessels. The Wharton’s jelly contains hyaluronic acid, col- and xenogeneic formats. The suppression of the proliferation
lagen, and primitive multipotent cells [21, 24, 26, 28, 29, 36, 39, was 25%– 40% (in Fig. 1) to 38% (Fig. 2), showing good
40]. Human UCMS cells used here met the definition estab- agreement among three methods used to evaluate xenogeneic
lished for MSCs by the ISCT based upon their plastic adherence, suppression. The suppression in the allogeneic human MLR
morphology, surface phenotype, and multipotency (supplemen- averaged 38% (range, 17%–54%; Fig. 5). UCMS cells did not
tal online Table 1). The differences between adult-derived inhibit proliferation of cultured splenocytes, and there is no
MSCs and UCMS cells are that (a) UCMS cells have the evidence that UCMS cell coculture caused splenocyte death.
potential for faster expansion ex vivo, and (b) UCMS cells have Interestingly, a recent publication reports that VEGF and IL-6
telomerase activity [22, 24, 28, 40], and the umbilical cord is secreted by MSCs result in immunosuppression [54]. VEGF is
relatively enriched for CFU-F compared with bone marrow [36]. thought to cause this effect by blocking the emigration or
Several laboratories have reported that 10,000 –15,000 nucle- differentiation of bone marrow lymphoid precursors [55]. IL-6
ated cells can be isolated per centimeter of human umbilical has been postulated to inhibit the differentiation of monocytes to
cord length [24, 29, 34]; another laboratory has reported 106 dendritic cells [56]. The focused array data indicated that
nucleated cells per centimeter [36]. Thus, Wharton’s jelly is hUCMS cells express IL-6 and VEGF, as was reported previ-
enriched for MSC-like cells. Furthermore, UCMS cells are ously [24]. The role of these factors will require confirmation to
www.StemCells.com
2872 Immune Properties of UCMS Cells

determine whether they are playing a role in the immune mod- Here, the one-way and two-way MLR assays were examined
ulation observed here. as a function of passage (time in cell culture). Thus, additional
MSCs have been reported in most publications to be MHC information was obtained. Specifically, the immunogenicity of
class II-negative (e.g., [1, 57]), but in at least two publications human UCMS cells was low and did not change from passage 5
they have been reported as weakly positive [34, 58]. To date, through passage 9 in four isolates examined. In contrast, human
cultured UCMS cells have been reported to be MHC class UCMS cells were found to be immunosuppressive at passages
II-negative [28, 29, 36, 40]. As discussed above, exposure to 5– 6 and passage 9, but the degree of immune suppression
interferon-␥ stimulates increased MHC class I and induces observed in two of three isolates may have decreased slightly
MHC class II expression by human and swine UCMS cells [43]. during passage. Since chondrogenic differentiation alters the
Importantly, UCMS cells express IL-6 and VEGF genes (sup- immunosuppressive properties of MSCs [65], it is possible that
plemental online Table 1; [24]), and since, as stated above, these differentiation of UCMS cells may explain why two of the three
two proteins have recently been shown to be pivotal in the isolates lost immunosuppressive properties at passage 9. This is
immunosuppressive capability of MSCs [54], their expression not likely, though, because UCMS cells were not exposed to
by the UCMS cells may be a mechanism for the immunosup- medium with specific differentiation factors, and there were no
pressive properties demonstrated here. In support of the cyto- data to suggest that the UCMS cells were differentiating (e.g.,
kine gene expression profile provided here, the previous gene no lipid in cells, no slowing down of proliferation, no evidence
expression analysis of UCMS cells confirms the focus array data of matrix deposition, and no changes in cellular morphology to
provided here [24], and Friedman et al. have reported that indicate differentiation of UCMS cells). Further work is needed
UCMS cells release hematopoietic and immune regulatory cy- to determine whether prolonged culture or, specifically, differ-
tokines into the medium [59]. In addition, MSCs do not express entiation plays a role in UCMS cell immunophysiological prop-
costimulatory molecules (e.g., CD80, CD86, or CD40) that erties. In this regard, recent work by Cho et al. indicates that
contribute to immunogenicity [34, 60]. Human UCMS cells do culture of umbilical cord-derived cells with inflammatory cyto-
not express these molecules either (Table 1). Together, these kine interferon-␥ may increase immunogenicity by expression
findings may explain the lack of an apparent host immune of MHC molecules [43]. The mechanism for this change in
response observed following transplantation of either porcine immune properties is unknown but may be due to changes in
[21, 23] or human [24] UCMS cells into the rat brain, and they cytokine profile or nitric oxide release (Dr. L. Wang, Auburn
support the notion that UCMS cells share properties of MSCs. University, personal communication).

HLA-G Xenotransplantation Studies with hUCMS Cells


The reduced immunogenicity of MSCs has been likened to UCMS cells, unlike ESCs, do not induce tumors or death after
maternal acceptance of the fetal allograft [19]. The human MHC 1 ⫻ 106 to 6 ⫻ 106 undifferentiated hUCMS cells were trans-
class I molecule HLA-G has long been known as a molecule planted either i.v. or s.c. into Beige/SCID mice [24, 45]. In
selectively expressed by cytotrophoblastic cells. By inhibiting addition, UCMS cells are not acutely rejected when transplanted
the cytolytic function of decidual natural killer cells, HLA-G as xenografts in immune-competent rats. For example, pig
protects the fetal tissue from the mother’s immune system. UCMS cells undergo a moderated expansion following trans-
HLA-G molecules have 15 alleles [61– 63]; we tested for plantation into rat brain without obvious untoward behavioral
pan-HLA-G (data not shown) and for the soluble HLA isoforms effects or host immune response [21, 23]. Pig UCMS cells were
HLA-G5 and HLA-G6. Since UCMS cells are fetal cells derived recovered from rat brain more than 6 weeks after transplantation
from postnatal tissue anatomically continuous with the placenta, without immune suppression. Similarly, human UCMS trans-
we examined HLA-G expression in UCMS cells. The expres- plantation into rats did not trigger obvious host immune re-
sion of pan-HLA-G (data not shown) by both early (P4) and sponse around the transplantation site, although cells were not
later (P8) passage human UCMS cells and expression of the recovered more than 1 week after transplantation [24]. This
HLA-G6 isoform suggest that UCMS cells share some of the degree of tolerance may be due to either immune property tested
gene expression profile expressed by placental cells. Therefore, here: that is, either immunosuppressivity or low immunogenic-
the importance of the presence of HLA-G in the UCMS cells in ity. In either case, the lack of infiltration by host immune cells
immune suppression and immunogenicity is currently unknown. bodes well for engraftment of UCMS cells. To date, there are no
A more complete examination of the role of HLA-G6 in the reports of engraftment, expansion, or differentiation of UCMS
immune physiology of UCMS cells is slated for the future. cells. Therefore, UCMS cells have not met the definition of true
An important physiological variable for cells under consid- stem cells. It is possible that syngeneic grafting, perhaps to-
eration for cellular therapy is how well those cells are tolerated gether with opening of the niche via irradiation, will be needed
by the host’s immune system. For example, will complete to permit UCMS cells to engraft.
haplotype matching be required for engraftment, or will mis-
matched (e.g., allogeneic) transplantation be permitted? This is
important information for two independent reasons. First, since SUMMARY
graft rejection and graft-versus-host disease seen in bone mar-
row transplants are serious, life-threatening complications, it is Human UCMS cells from passages 4 –9 have low immunoge-
important for engraftment, reduction of risk, and therapeutic nicity and suppress the proliferation of activated immune cells.
efficacy to understand the immunogenicity and immune sup- The mRNA for HLA-G6, an immunosuppressive HLA-class I
pression function of therapeutic cells. Second, the tissue match- marker synthesized by cytotrophoblasts at the maternal-fetal
ing requirements will define commercial variables, such as how interface, was found in UCMS cells. In contrast, HLA-G5
many samples the stem cell bank must contain to serve a diverse expression was not found. This differentiates umbilical cord matrix
population. This mathematical exercise has been conducted by stromal cells from cytotrophoblastic cells, which express both
embryonic stem cell scientists, since embryonic stem cells will HLA-G5 and HLA-G6, and from bone marrow-derived MSCs,
require tissue matching [64]. If allogeneic transplantation is safe which do not express HLA-G. Human UCMS cells do not express
and effective, a smaller bank of human UCMS cells would be the costimulatory surface antigens CD40, CD80, and CD86. Fo-
required, compared with embryonic stem cells. cused gene array data revealed that UCMS cells express IL-6 and
Weiss, Anderson, Medicetty et al. 2873

VEGF, molecules thought to play a role in MSC immune regula- dra Rao, Sherry Flemming, and Frank Blecha for assistance with
tion. These data, taken together with previous observations follow- the research. We thank Drs. F. Blecha and S. Flemming for
ing xenotransplantation of UCMS cells, suggest that hUCMS cells critically reviewing a draft of the manuscript. Dr. Susan Bennett
would be tolerated in allogenic transplantation. and the OB/Gyn Staff at Mercy Medical Center are thanked for
assistance in obtaining specimens. The anonymous donors are
thanked for their contribution. S.M. is currently affiliated with
ACKNOWLEDGMENTS Athersys Inc., Cleveland, OH.

We acknowledge the support of K-INBRE, Kansas State Uni- DISCLOSURE OF POTENTIAL CONFLICTS
versity (KSU) Developing Scholars program, KSU Department OF INTEREST
of Anatomy and Physiology, KSU College of Veterinary Med-
icine Dean’s office, KSU Provost’s office, KSU Terry C. John- M.L.W. is a paid consultant for the Regenerative Medicine
son Center for Basic Cancer Research, NIH (NS34160), and Institute (Las Vegas, NV) and Toucan Capital Fund II (Be-
support from the State of Kansas to the Midwest Institute for thesda, MD); K.R.M. is employed by Cognate Bioservices;
Comparative Stem Cell Biology. The Northeast Kansas Parkin- S.M. is employed by Athersys; and M.L.W. has acted as a
son Association is thanked for its generous support. We thank consultant and performed contract work for Toucan Capital
Robert Deans, Tony Ting, Kathy Mitchell, Alan Smith, Mahen- Corporation.

20 Batten P, Sarathchandra P, Antoniw JW et al. Human mesenchymal stem


REFERENCES cells induce T cell anergy and downregulate T cell allo-responses via the
TH2 pathway: Relevance to tissue engineering human heart valves.
Tissue Eng 2006;12:2263–2273.
1 Dominici M, Le Blanc K, Mueller I et al. Minimal criteria for defining 21 Medicetty S, Bledsoe AR, Fahrenholtz CB et al. Transplantation of pig
multipotent mesenchymal stromal cells. The International Society For stem cells into rat brain: Proliferation during the first 8 weeks. Exp
Cellular Therapy Position Statement. Cytotherapy 2006;8:315–317. Neurol 2004;190:32– 41.
2 Le Blanc K. Immunomodulatory effects of fetal and adult mesenchymal 22 Mitchell KE, Weiss ML, Mitchell BM et al. Matrix cells from Wharton’s
stem cells. Cytotherapy 2003;5:485– 489. jelly form neurons and glia. STEM CELLS 2003;21:50 – 60.
3 Nauta AJ, Fibbe WE. Immunomodulatory properties of mesenchymal 23 Weiss ML, Mitchell KE, Hix JE et al. Transplantation of porcine
stromal cells. Blood 2007;110:3499 –3506. umbilical cord matrix cells into the rat brain. Exp Neurol 2003;182:
4 Bartholomew A, Sturgeon C, Siatskas M et al. Mesenchymal stem cells 288 –299.
suppress lymphocyte proliferation in vitro and prolong skin graft survival 24 Weiss ML, Medicetty S, Bledsoe AR et al. Human umbilical cord matrix
in vivo. Exp Hematol 2002;30:42– 48. stem cells: Preliminary characterization and effect of transplantation in a
5 Di Nicola M, Carlo-Stella C, Magni M et al. Human bone marrow rodent model of Parkinson’s disease. STEM CELLS 2006;24:781–792.
stromal cells suppress T-lymphocyte proliferation induced by cellular or
25 Conconi MT, Burra P, Di LR et al. CD105(⫹) cells from Wharton’s jelly
nonspecific mitogenic stimuli. Blood 2002;99:3838 –3843.
show in vitro and in vivo myogenic differentiative potential. Int J Mol
6 Glennie S, Soeiro I, Dyson PJ et al. Bone marrow mesenchymal stem
Med 2006;18:1089 –1096.
cells induce division arrest anergy of activated T cells. Blood 2005;105:
26 Fu YS, Shih YT, Cheng YC et al. Transformation of human umbilical
2821–2827.
mesenchymal cells into neurons in vitro. J Biomed Sci 2004;11:652–660.
7 English K, Barry FP, Field-Corbett CP et al. IFN-gamma and TNF-alpha
27 Fu YS, Cheng YC, Lin MY et al. Conversion of human umbilical cord
differentially regulate immunomodulation by murine mesenchymal stem
cells. Immunol Lett 2007;110:91–100. mesenchymal stem cells in Wharton’s jelly to dopaminergic neurons in
8 Xu G, Zhang L, Ren G et al. Immunosuppressive properties of cloned vitro: Potential therapeutic application for Parkinsonism. STEM CELLS
bone marrow mesenchymal stem cells. Cell Res 2007;17:240 –248. 2006;24:115–124.
9 Le Blanc K, Pittenger M. Mesenchymal stem cells: Progress toward 28 Karahuseyinoglu S, Cinar O, Kilic E et al. Biology of stem cells in
promise. Cytotherapy 2005;7:36 – 45. human umbilical cord stroma: In situ and in vitro surveys. STEM CELLS
10 Le Blanc K, Rasmusson I, Gotherstrom C et al. Mesenchymal stem cells 2007;25:319 –331.
inhibit the expression of CD25 (interleukin-2 receptor) and CD38 on 29 Sarugaser R, Lickorish D, Baksh D et al. Human umbilical cord perivas-
phytohaemagglutinin-activated lymphocytes. Scand J Immunol 2004;60: cular (HUCPV) cells: A source of mesenchymal progenitors. STEM
307–315. CELLS 2005;23:220 –229.
11 Li YP, Latger-Canard V, Marchal L et al. The regulatory role of dendritic 30 Wang HS, Hung SC, Peng ST et al. Mesenchymal stem cells in the
cells in the immune tolerance. Biomed Mater Eng 2006;16:S163–S170. Wharton’s jelly of the human umbilical cord. STEM CELLS 2004;22:
12 Pierdomenico L, Bonsi L, Calvitti M et al. Multipotent mesenchymal 1330 –1337.
stem cells with immunosuppressive activity can be easily isolated from 31 Ma L, Feng XY, Cui BL et al. Human umbilical cord Wharton’s
dental pulp. Transplantation 2005;80:836 – 842. Jelly-derived mesenchymal stem cells differentiation into nerve-like
13 Puissant B, Barreau C, Bourin P et al. Immunomodulatory effect of cells. Chin Med J (Engl) 2005;118:1987–1993.
human adipose tissue-derived adult stem cells: Comparison with bone 32 Wu KH, Zhou B, Yu CT et al. Therapeutic potential of human umbilical
marrow mesenchymal stem cells. Br J Haematol 2005;129:118 –129. cord derived stem cells in a rat myocardial infarction model. Ann Thorac
14 McIntosh K, Zvonic S, Garrett S et al. The immunogenicity of human Surg 2007;83:1491–1498.
adipose-derived cells: Temporal changes in vitro. STEM CELLS 2006; 33 Le Blanc K, Tammik C, Rosendahl K et al. HLA expression and
24:1246 –1253. immunologic properties of differentiated and undifferentiated mesenchy-
15 Wolbank S, Peterbauer A, Fahrner M et al. Dose-dependent immuno- mal stem cells. Exp Hematol 2003;31:890 – 896.
modulatory effect of human stem cells from amniotic membrane: A 34 Klyushnenkova E, Mosca JD, Zernetkina V et al. T cell responses to
comparison with human mesenchymal stem cells from adipose tissue. allogeneic human mesenchymal stem cells: Immunogenicity, tolerance,
Tissue Eng 2007;13:1173–1183. and suppression. J Biomed Sci 2005;12:47–57.
16 Le Blanc K, Rasmusson I, Sundberg B et al. Treatment of severe acute 35 Riteau B, Moreau P, Menier C et al. Characterization of HLA-G1, -G2,
graft-versus-host disease with third party haploidentical mesenchymal -G3, and -G4 isoforms transfected in a human melanoma cell line.
stem cells. Lancet 2004;363:1439 –1441. Transplant Proc 2001;33:2360 –2364.
17 Grinnemo KH, Mansson A, Dellgren G et al. Xenoreactivity and en- 36 Lu LL, Liu YJ, Yang SG et al. Isolation and characterization of human
graftment of human mesenchymal stem cells transplanted into infarcted umbilical cord mesenchymal stem cells with hematopoiesis-supportive
rat myocardium. J Thorac Cardiovasc Surg 2004;127:1293–1300. function and other potentials. Haematologica 2006;91:1017–1026.
18 Liechty KW, MacKenzie TC, Shaaban AF et al. Human mesenchymal 37 Seshareddy K, Troyer D, Weiss ML. Method to isolate mesenchymal-
stem cells engraft and demonstrate site-specific differentiation after in like cells from Wharton’s jelly of umbilical cord. Methods Cell Biol
utero transplantation in sheep. Nat Med 2000;6:1282–1286. 2008;86:101–119.
19 Barry FP, Murphy JM, English K et al. Immunogenicity of adult mes- 38 Weiss ML, Troyer DL. Stem cells in the umbilical cord. Stem Cell Rev
enchymal stem cells: Lessons from the fetal allograft. Stem Cells Dev 2006;2:155–162.
2005;14:252–265. 39 Carlin R, Davis D, Weiss M et al. Expression of early transcription

www.StemCells.com
2874 Immune Properties of UCMS Cells

factors Oct-4, Sox-2 and Nanog by porcine umbilical cord (PUC) matrix 52 Barry FP. Biology and clinical applications of mesenchymal stem cells.
cells. Reprod Biol Endocrinol 2006;4:8 Birth Defects Res C Embryo Today 2003;69:250 –256.
40 Lund RD, Wang S, Lu B et al. Cells isolated from umbilical cord tissue 53 Devine SM, Bartholomew AM, Mahmud N et al. Mesenchymal stem
rescue photoreceptors and visual functions in a rodent model of retinal cells are capable of homing to the bone marrow of non-human primates
disease. STEM CELLS 2007;25:602– 611. following systemic infusion. Exp Hematol 2001;29:244 –255.
41 Poncelet AJ, Vercruysse J, Saliez A et al. Although pig allogeneic 54 Djouad F, Charbonnier LM, Bouffi C et al. Mesenchymal stem cells
mesenchymal stem cells are not immunogenic in vitro, intracardiac inhibit the differentiation of dendritic cells through an interleukin-6-
injection elicits an immune response in vivo. Transplantation 2007;83: dependent mechanism. STEM CELLS 2007;25:2025–2032.
783–790. 55 Ohm JE, Gabrilovich DI, Sempowski GD et al. VEGF inhibits T-cell
42 Beggs KJ, Lyubimov A, Borneman JN et al. Immunologic consequences development and may contribute to tumor-induced immune suppression.
of multiple, high-dose administration of allogeneic mesenchymal stem Blood 2003;101:4878 – 4886.
cells to baboons. Cell Transplant 2006;15:711–721. 56 Hegde S, Pahne J, Smola-Hess S. Novel immunosuppressive properties
43 Cho PS, Messina DJ, Hirsh EL et al. Immunogenicity of umbilical cord of interleukin-6 in dendritic cells: Inhibition of NF-kappaB binding
tissue derived cells. Blood 2008;111:430 – 438. activity and CCR7 expression. FASEB J 2004;18:1439 –1441.
44 Jomura S, Uy M, Mitchell K et al. Potential treatment of cerebral global 57 Deans RJ, Moseley AB. Mesenchymal stem cells: Biology and potential
ischemia with Oct-4⫹ umbilical cord matrix cells. STEM CELLS 2007; clinical uses. Exp Hematol 2000;28:875– 884.
25:98 –106. 58 Potian JA, Aviv H, Ponzio NM et al. Veto-like activity of mesenchymal
45 Rachakatla RS, Marini F, Weiss ML et al. Development of human stem cells: Functional discrimination between cellular responses to al-
umbilical cord matrix stem cell-based gene therapy for experimental lung loantigens and recall antigens. J Immunol 2003;171:3426 –3434.
tumors. Cancer Gene Ther 2007;14:828 – 835. 59 Friedman R, Betancur M, Boissel L et al. Umbilical cord mesenchymal
46 Bailey MM, Wang L, Bode CJ et al. A comparison of human umbilical stem cells: Adjuvants for human cell transplantation. Biol Blood Marrow
cord matrix stem cells and temporomandibular joint condylar chondro-
Transplant 2007;13:1477–1486.
cytes for tissue engineering temporomandibular joint condylar cartilage.
60 Tse WT, Pendleton JD, Beyer WM et al. Suppression of allogeneic T-cell
Tissue Eng 2007;13:2003–2010.
47 Baksh D, Yao R, Tuan RS. Comparison of proliferative and multilineage proliferation by human marrow stromal cells: Implications in transplan-
differentiation potential of human mesenchymal stem cells derived from tation. Transplantation 2003;75:389 –397.
umbilical cord and bone marrow. STEM CELLS 2007;25:1384 –1392. 61 Carosella ED, Moreau P, Le MJ et al. HLA-G molecules: From maternal-
48 Hoerstrup SP, Kadner A, Breymann C et al. Living, autologous pulmo- fetal tolerance to tissue acceptance. Adv Immunol 2003;81:199 –252.
nary artery conduits tissue engineered from human umbilical cord cells. 62 Hunt JS, Petroff MG, McIntire RH et al. HLA-G and immune tolerance
Ann Thorac Surg 2002;74:46 –52. in pregnancy. FASEB J 2005;19:681– 693.
49 Schmidt D, Mol A, Odermatt B et al. Engineering of biologically active 63 Hviid TV HLA-G in human reproduction: Aspects of genetics, function
living heart valve leaflets. Tissue Eng 2006;12:3223–3232. and pregnancy complications. Hum Reprod Update 2006;12:209 –232.
50 Kadereit S, Deeds LS, Haynesworth SE et al. Expansion of LTC-ICs and 64 Drukker M, Urbach A, Schuldiner M et al. Characterization of the
maintenance of p21 and BCL-2 expression in cord blood CD34(⫹)/ expression of MHC proteins in human embryonic stem cells. Proc Natl
CD38(-) early progenitors cultured over human MSCs as a feeder layer. Acad Sci U S A 2002;99:9864 –9869.
STEM CELLS 2002;20:573–582. 65 Chen X, McClurg A, Zhou GQ et al. Chondrogenic differentiation alters
51 Reese JS, Koc ON, Gerson SL. Human mesenchymal stem cells provide the immunosuppressive property of bone marrow-derived mesenchymal
stromal support for efficient CD34⫹ transduction. J Hematother Stem stem cells, and the effect is partially due to the upregulated expression of
Cell Res 1999;8:515–523. B7 molecules. STEM CELLS 2007;25:364 –370.

See www.StemCells.com for supplemental material available online.

S-ar putea să vă placă și