Вы находитесь на странице: 1из 18

Available online at www.sciencedirect.

com

ScienceDirect
Journal of Cellular Immunotherapy 2 (2016) 3e20
http://www.elsevier.com/locate/jocit

Mesenchymal stem cells: Immunomodulatory capability and clinical


potential in immune diseases
Qinjun Zhao a, Hongying Ren b, Zhongchao Han a,b,*
a
National Engineering Research Center of Cell Products/AmCellgene Co. Ltd, Tianjin, China
b
State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Disease Hospital, Chinese Academy of Medical Sciences and Peking
Union of Medical College, Tianjin, China
Received 13 August 2014; revised 29 November 2014; accepted 15 December 2014
Available online 24 February 2015

Abstract

Mesenchymal stem cells (MSCs) represent a heterogenous population of adult, fibroblast-like multi-potent cells. MSCs have drawn much
attention during the last decade in the field of regenerative medicine, mainly due to their capacity to differentiate into specific cell types,
abundant production of soluble growth factors and cytokines, and hematopoiesis supporting properties. In addition, MSCs can migrate to the
sites of inflammation and hold potent of immunomodulatory and anti-inflammatory effects through cell and cell interactions between MSCs and
lymphocytes or production of soluble factors. Therefore, the application of MSCs in many disease situations is full of possibilities for future
clinical treatment. Phase III clinical trials have been run using MSCs for treatment of Graft versus Host Disease (GVHD), and MSCs product
approval has been achieved for pediatric GVHD treatment in Canada and New Zealand (Prochymal®; Osiris Therapeutics). In addition, hundreds
of clinical trials are being run using MSCs for treatment of several immune mediated diseases, including GVHD, aplastic anemia (AA), Crohn's
disease (CD), rheumatoid arthritis (RA), and multiple sclerosis (MS). In this review we mainly focus on immunomodulation potential of MSCs
and promising therapeutic application of MSCs in immune mediated diseases. Furthermore, we emphasize that biological effectiveness of MSCs
will be one of most important standards to determine the dose of MSCs infusion.
Copyright © 2015, Shanghai Hengrun Biomedical Technology Research Institute. Publishing Services By Elsevier B.V. This is an open access
article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

Keywords: MSCs; Immunomodulatory; Cell infusion; Biological effectiveness

1. Introduction tissues [3], fetal liver [4], cord blood and mobilized peripheral
blood [5], fetal lung [6], placenta [7], umbilical cord [8,9],
Friedenstein was the first person to describe the isolation of dental pulp [10], synovial membrane [11], periodontal liga-
clonogenic, proliferating fibroblastic MSCs from rat bone ment [12], endometrium [13], trabecular and compact bone
marrow (BM). And they showed that colony-forming unit fi- [14,15] (Fig. 1).
broblasts (CFU-Fs) derived stromal cells can serve as feeder Increased evidences have shown that under appropriate
layers for the culture of heamatopoietic stem cells (HSCs) and culture conditions, MSCs are capable of differentiating into
can differentiate into osteocytes, chondricytes and adipocytes mesodermal, endodermal and even ectodermal cells. MSCs are
[1,2]. Besides BM, MSCs can also be isolated from adipose capable of secreting growth factors and immunoprotective
cytokines which have been used in the field of cell and organ
transplantation. Most importantly, MSCs are safe, do not form
* Corresponding author. Institute of Hematology Chinese Academy of
teratoma, and can be used for tissue regeneration and repair
Medical Sciences and Peking Union of Medical College, 288 Nan Jing Road,
300020, Tianjin, China. Fax: þ86 22 27317273. [16,17]. And easy isolation, large quantity expansion and
E-mail address: hanzhongchao@hotmail.com (Z. Han). multipotential differentiation of MSCs make them ideal
Peer review under responsibility of Shanghai Hengrun Biomedical Tech- candidate for stem cell-based therapy and their application as
nology Research Institute.

http://dx.doi.org/10.1016/j.jocit.2014.12.001
2352-1775/Copyright © 2015, Shanghai Hengrun Biomedical Technology Research Institute. Publishing Services By Elsevier B.V. This is an open access article
under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
4 Q. Zhao et al. / Journal of Cellular Immunotherapy 2 (2016) 3e20

Fig. 1. The tissue source and multipotential differentiation of MSCs. MSCs are multipotent cells that can be isolated from various human tissues including BM,
adipose tissues, fetal liver, mobilized peripheral blood, cord blood, fetal lung, placenta, umbilical cord, dental pulp, synovial membrane, periodontal ligament,
endometrium, trabecular and compact bone. MSCs have the capacity to differentiate into mesenchymal tissues both in vitro and in vivo, including osteoblast,
condrocyte, adipocyte, tendon, skeletal muscle cells, cardiomyocyte and haematopoietic supporting stroma cells. In addition, MSCs can differentiate into tissues of
ectodermal, including skin, neurons. Furthermore, MSCs can differentiate into tissues of endodermal including lung, hepatocytes, renal, and pancreatic b-cells,
endothelial cells.

gene carriers. Another intriguing feature of MSCs is that they 2. Definition of MSCs
are able to escape immune recognition and inhibit immune
responses. Therefore, MSCs will be a very promising tool for Pittenger et al. firstly defined MSCs by their ability to
immunomodulatory cell therapy in immune mediated diseases. proliferate in culture with an attached fibroblast-like
At the time of writing, there are currently 423 clinical trials morphology, by the presence of a consistent set of cell sur-
using MSCs registered at clinical-trials.gov. The clinical trials face protein markers, and by their extensive consistent dif-
have been run for tissue repair including cardiac ischemia, ferentiation to multiple lineages mesenchymal tissue under
limb ischemia, amyotrophic lateral sclerosis, diabetes, certain controlled in vitro conditions [18]. However, to date
ischemic stroke, osteoarthritis, liver cirrhosis, and liver failure because there is no specific or unique cell surface marker, so
and so on. More clinical trials have been run for immune the definition of MSCs was always controversy. Therefore,
mediated diseases including GVHD, CD, MS, respiratory currently MSCs have been defined by using a combination of
distress syndrome, AA, and RA (www.clinicaltrials.gov). In cell surface phenotypic protein markers, plastic adherent
fact, immune mediated diseases are actually the largest kind of fibroblast-like growth and functional properties. It is generally
diseases to be clinically studied now. In addition, the biolog- agreed that adult human MSCs express Stro-1 [19], CD105
ical effectiveness maybe one of important factors to determine (SH2) [20], CD73 (SH3/4) [21], CD44, CD71 (transferring
the cell dose for infusion. The biological effectiveness always receptor), CD90 (THY1), the ganglioside GD2 [22,23] and
different in different tissue derived MSCs or different MSCs CD271 (low-affinity nerve growth factor receptor), as well as
subpopulation. In other words, the treatment of various disease some cell adhesion molecules including intercellular adhesion
maybe will need different MSCs subpopulation and MSCs molecule-1,-2 (ICAM-1,-2), integrins (a1, a2, a3, a5, a6, aV,
dose. And, for the same disease, MSCs dose is also variable b1, b3, b4) [24], activated leukocyte-cell adhesion molecule
for different tissue derived MSCs. (ALCAM), lymphocyte function-associated antigen 3 (LFA-
Q. Zhao et al. / Journal of Cellular Immunotherapy 2 (2016) 3e20 5

3), vascular cell adhesion molecule-1 (VCAM-1), and CD72 transformation after extensive in vitro expansion [57]. Chen
[18,25]. They also express human leukocyte antigen (HLA) et al. reported that human umbilical cord MSCs (hUC-MSCs)
class I but not class II molecules on cell surface [26,27]. maintain their biological character and function after long-
Additionally, MSCs lack the expression of typical hemato- term in vitro culturing (P15). Since hUC-MSCs can be
poietic antigens CD11a, CD14, CD31, CD34 and CD45 safely expanded in vitro and are not susceptible to malignant
[9,18], Or co-stimulatory molecules CD40, CD80, and CD86 transformation in serum-free medium [58]. Therefore, basi-
[28,29]. The International Society for Cellular Therapy (ISCT) cally MSCs can maintain a stable immunophenotype and
has offered several minimal criteria to identify MSCs which chromosome structure and will not be malignant trans-
are listed as: 1) Plastic adherent fibroblast-like growth while formation after long term in vitro culture expansion.
maintaining these cells in standard conditions. 2) Expression At the same time, increased evidences show that MSCs is
of CD73, CD90 and CD105 markers in at least 95% of cell also safe in vivo normal animal or animal disease model cell
population and lack expression of CD34, CD45, CD14, infusion therapy. Wang et al. evaluate the overall toxicology of
CD11b, CD19 or CD79a and HLA-II markers as measured by hUC-MSCs in cynomolgus monkeys with repeated adminis-
flow cytometry. 3) Differentiation capability into adipogenic, trations and show that transplantation of hUC-MSCs did not
osteogenic and chondrogenic lineage cells in vitro [30]. Some affect the general health of cynomolgus monkeys [16]. Feng
markers, including GD2, nestin and CD271, have been used to et al. evaluated the safety of human MSCs transplanted in
investigate the function of certain tissue derived MSCs or cerebrum of Macaca fascicularis. Their results show that the
different MSCs subpopulation [22,23,31,32]. For example, transplantation of human MSCs in monkeys did not affect total
GD2þ umbilical cord derived MSCs are a subpopulation of IgM, IgG, CD3, CD4, or CD8 values. And transplantation of
MSCs with feature of primitive precursor cells and most hMSCs to the cerebrum represents a safe alternative for clin-
multipotential cells [23]. And CD106þ placenta derived ical application of neurological disorders [59]. Francois et al.
MSCs are the more mature and biological functional MSCs infused intravenously human MSCs to NOD/SCID mice with
with feature of stronger immunomodulatory capability [33] total body irradiation or local abdominal or leg irradiation and
and angiogenic potential (not published). investigated the long term side-effect. Their results demon-
Numbers studies also showed that the MSCs have the ca- strated that no tissue abnormality or abnormal human MSCs
pacity to differentiate into multiple mesenchymal tissues both proliferation was observed at 120 days after irradiation.
in vitro and in vivo, including osteoblast [25,34], condrocyte Meanwhile, These results showed that MSCs injection is safe
[35,36], adipocyte [37,38], tendon [39,40], skeletal muscle and efficient for long-term treatment of severe complications
cells [39], cardiomyocyte [41,42] and hematopoietic support- after radiotherapy for patients refractory to conventional
ing stroma cells [43]. In addition, MSCs also have capability treatments [60].
for differentiating into tissues of ectodermal, including skin Finally, human MSCs is also safe according to numerous
[44], neurons [45]. Furthermore, MSCs can differentiate into clinical trial reports. Shi et al. assessed the safety and initial
tissues of endodermal including lung [46,47], hepatocytes efficacy of UC-MSCs transfusions for acute-on-chronic liver
[27,48], renal [49,50], and pancreatic b-cells [51,52], endo- failure (ACLF) patients associated with hepatitis B virus
thelial cells [53,54] (Fig. 1). Actually, the tissue repair capa- (HBV) infection. Their results suggest that UC-MSCs trans-
bility of MSCs is mainly base on their multi-lineage fusions are safe in the clinic and may serve as a novel thera-
differentiation potential. peutic approach for HBV-associated ACLF patients [61].
Wang et al. assess the safety and efficacy of human UC-MSCs
3. The safety of MSCs in clinical application in the treatment of rheumatoid arthritis (RA). Their results
show that no serious adverse effects were observed during or
MSCs have been used in several approaches for regenera- after infusion. Furthermore, the treatment of UC-MSCs
tive cell therapy, as well as in the perspective of modulating induced a significant remission of disease according to the
immune response. Therefore, the biosafety features of MSCs 28-joint disease activity score [62]. Rodrigo et al. evaluates the
need to be carefully investigated to exclude the occurrence of safety and feasibility of intramyocardial MSCs injection in
functional or genetic alterations before their release for clin- nine patients, shortly after AMI during short-term and 5-year
ical use. Bernardo et al. show that human BM-derived MSCs follow-up. Their results suggest that intramyocardial injec-
can be cultured long-term in vitro, without losing their tion of MSCs in patients shortly after AMI is feasible and safe
morphologic, phenotypical, and functional characteristics. up to 5-year follow-up [63]. Gupta et al. conducted a pro-
Moreover, MSCs propagated in culture continuously for up to spective double blind randomized placebo controlled multi-
44 weeks maintained a normal karyotype [55]. Meza-zepeda center study to determine the safety of BM-MSCs in pa-
et al. also report that adipose derived MSCs do not bypass tients with critical limb ischemia. Their results show that BM-
senescence even after two months of post-senescence culture. MSCs are also safe when injected intramuscularly at a dose of
MSCs show no evidence of transformation in vitro on the basis 2 million cells/kg body weight [64]. In addition, Lee et al.
of re-entry into the cell cycle, mitotic index, acquisition of a performed a randomized pilot study to investigate the safety
rounded phenotype and loss of anchorage-dependence [56]. In and efficacy of MSCs in patients with AMI. Their results
addition, Poloni et al. show that human MSCs from BM, showed that there was no treatment-related toxicity during
chorionic villi and amniotic fluid are not susceptible to intracoronary administration of MSCs. Meanwhile, no
6 Q. Zhao et al. / Journal of Cellular Immunotherapy 2 (2016) 3e20

significant adverse cardiovascular events occurred during cultures in the presence of MSCs [73,74]. Yan et al. reported
follow-up [65]. Futhermore,Pak et al. evaluate the safety of that MSC-exposed Treg cells are capable of more immuno-
adipose tissue derived MSCs (AT-MSCs) that was used for suppressive than Tregs without coculturing with MSCs. And
patients suffering from orthopedic conditions. Ninety one their results showed that programmed cell death 1 receptor/
patients suffering from orthopedic conditions were treated B7-H1 interactions and IL-10 might be responsible for the
with autologous AT-MSCs. AT-MSCs were injected with PRP enhanced suppressive capability of MSC-exposed regulatory T
into various joints (n ¼ 100). No neoplastic complications cells [75]. However, depletion of regulatory T cells had no
were detected at any AT-MSCs implantation sites. Based on effect on the suppression of T cell proliferation by MSCs, and
longitudinal cohort, the autologous and uncultured ADSCs/ MSCs physically hinder T cells from the contact with antigen
PRP therapy could be considered to be safe when used as presenting cells (APCs) in a noncognate fashion [76]. Human
percutaneous local injections [66]. Till now, scientists evaluate adipocyte-MSCs undergo different modes of activation, when
the safety of MSCs through three different level including they were treated with mouse splenic T cell culture superna-
in vitro culture expansion, animal disease model and clinical tant, compared to when they were stimulated with human
trials and get a conclusion that MSCs is safe cell. In addition, PBMC supernatant. However, they still exerted an anti-
MSCs preparation and production must be manipulated in proliferative effect on mouse splenic T cells in vitro, primar-
GMP or GTP laboratory and only qualified MSCs can be used ily through COX-2 expression [77]. Human adipose-AT-MSCs
for clinical application for safety and achieving ideal clinical obtained from kidney donors induced a 2$1-fold increase in
effect. the percentage of CD25(þ) CD127() FoxP3(þ) cells within
the CD4(þ) T cell population from allostimulated CD25()
4. Interaction between MSCs and immune cells cells. And AT-MSCs induced Treg cells inhibited effector cell
proliferation as effectively as natural regulatory T cells. The
MSCs have also been shown to possess broad immunoreg- vast majority of cells within the induced Treg fraction had a
ulatory capabilities and are capable of influencing both adap- methylated FOXP3 gene Treg-specific demethylated region
tive and innate immune responses. MSCs inhibit immune cells indicating that they were not of natural regulatory T cells
proliferation and maturation and suppress immune reactions origin [78].
both in vitro and in vivo in a non-MHC restricted manner [67]. Although the exact mechanism underlying the immuno-
Therefore, MSCs are considered to be hypoimmunogenic, suppressive effects of MSCs is still not clear, most evidences
displaying low expression levels of HLA class I, no expression supported that soluble factors are involved. These factors
of HLA class Ⅱ, and no expression of costimulatory molecules, include prostaglandin E2 (PGE2) [73,79e81], indoleamine
including CD40, CD80, and CD86 [28,68]. Basically, MSCs 2,3-dioxygenase (IDO) [82,83], hepatocyte growth factor
could exert widespread immunomodulatory effects on cells of (HGF) [84,85] and transforming growth factor (TGF)-b 1
both the innate and adaptive immune system. Ex-vivo [69,86]. Additionally, it is well-established that IFN-g plays an
expanded MSCs have also been showed to suppress the activ- important role in the enhancement of MSCs suppressive ac-
ity of a broad range of immune cells, including T cells, natural tivity [82]. Furthermore, increased evidences support that
killer T (NKT) cells, dendritic cells (DCs), B cells, neutrophils, MSCs inhibit the proliferation and/or functions of CD4þ Th1
monocytes, macrophages and so on. and Th17 cells, CD8þ T cells, and natural killer cells pre-
dominantly via the secretion of soluble factors including TGF-
4.1. MSCs inhibit T cells proliferation and suppress b1 and HGF [87e90]. In addition, MSCs play a key role in
allogeneic T-cell response cytotoxic CD8þ T cells against intracellular pathogens.
Schurch et al. reported that IFN-g can promote the release of
The main features of the T cell response is cell proliferation hematopoietic cytokines, including IL-6 from MSCs, which in
and cytokines secretion. Inhibit T cell proliferation is the most turn reduced the expression of the transcription factors Runx-1
significant effect for MSCs. In vitro, MSCs are capable of and Cebpa in early hematopoietic progenitor cells and
suppressing T lymphocyte proliferation induced by mitogens increased myeloid differentiation and triggered the temporary
[67,69,70], alloantigens [67,68,71,72], as well as activation of activation of emergency myelopoiesis and promote clearance
T cells by CD3 and CD28 antibodies [68]. Suppression of T of the infection [91] (Fig. 2).
cell proliferation by MSCs has no immunological restriction,
similar suppressive effects being observed with cells that were 4.2. MSCs block natural killer T (NKT) cells
autologous or allogeneic to the responder cells [28,67,72]. proliferation and cytotoxicity
MSCs also modulate immune responses through the induction
of regulatory T cells which are important in maintaining im- NKT cells are part of the innate immune system, and bridge
mune homeostasis and self-tolerance. MSCs have been re- the adaptive immune system with the innate immune system.
ported to induce formation of regulatory T cells that were Once activated, these cells can perform functions ascribed to
responsible for inhibition of allogeneic lymphocyte prolifera- both Th and Tc cells, involving the enhance of cell mediated
tion [70]. In addition, an increase in the population of regu- immune response [92]. In addition, NKT cells play a key role
latory T (Treg) cells has been demonstrated in mitogen- in the elimination of virus infected cells and tumor cells.
stimulated peripheral blood mononuclear cell (PBMCs) MSCs alter the phenotype of NKT cells and suppress
Q. Zhao et al. / Journal of Cellular Immunotherapy 2 (2016) 3e20 7

Fig. 2. The MSCs exert their immune suppressive potential by cell to cell contact and by secretion of immune regulatory molecules. MSCs display broad
immunomodulatory properties, MSCs can inhibit the proliferation and function of T cells, Natural killer T (NKT) cells, T regulatory cells, B cells and dendritic
cells (DCs). However, MSCs secrete IL-6 and preserve neutrophils viability by inhibiting apoptosis. Several soluble factors have been shown to play a major role in
the immunosuppressive effects of MSCs, including prostaglandin E2 (PGE2), transforming growth factor (TGF)-b1, indoleamine 2,3-dioxygenase (IDO), nitric
oxide, hepatocyte growth factor (HGF), interleukin (IL)-6 and IL-10.

proliferation, cytokine secretion, and cytotoxicity against that MSCs can inhibit several functions of B cells, Corcione
HLA-class I-expressing targets. Some of these effects require et al. showed that B-cell proliferation was inhibited by MSCs
cell-to-cell contact, whereas others are mediated by soluble through an arrest in the G0/G1 phase of the cell cycle. A major
factors, including TGF-b1 and PGE2, suggesting the existence mechanism of B-cell suppression was soluble factors of MSCs
of diverse mechanisms for MSC-mediated NKT-cell suppres- production [96]. Furthermore, MSCs inhibited B-cell differ-
sion [84]. Spaggiari et al. also showed that MSCs can inhibit entiation because IgM, IgG, and IgA production was signifi-
the IL-2-induced proliferation of unactivated NKT cells [93]. cantly impaired. And CXCR4, CXCR5, and CCR7 in B-cell
In addition, the results reported by Selmani et al. showed that expression, as well as chemotaxis to CXCL12, the CXCR4
MSCs inhibited iNKT (Va24(þ)Vb11(þ)) and gd T (Vd2(þ)) ligand, and CXCL13, the CXCR5 ligand, were significantly
cell expansion from PBMC in both cell-to-cell contact and down-regulated by MSCs, suggesting that these cells affect
transwell systems. MSCs, through HLA-G5, affect innate chemotactic properties of B cells. In addition, MSCs also
immunity by inhibiting both NKT cell-mediated cytolysis and affect migrate to inflammatory regions under the guidance of
IFN-g secretion [94]. Furthermore, Prigione et al. reported that cell adhesion molecules and receptors for inflammatory che-
MSCs inhibited IFN-g production by activated Va24(þ) mokines [97]. One recent study by Lee et al. showed that the
Vb11(þ) and impaired CD3-mediated proliferation of acti- conditioned medium of MSCs infected with a mycoplasma
vated Va24(þ)Vb11(þ) and Vd2(þ) T cells, without affecting strain, Mycoplasma arginini, has marked inhibitory effects on
their cytotoxic potential [95]. Therefore, MSCs achieve inhi- Ig production by lipopolysaccharide/interleukin-4-induced B
bition of the NKT cells proliferation and immune regulatory cells compared with mycoplasma-free MSC-CM [98]. Yan
function by involving of multiple cytokines and signal et al. found that BAFF in MSCs was expressed at a higher
pathway (Fig. 2). level after TLR4-priming, indicating that TLR4 and a down-
stream pathway play a role in BAFF secretion and thus exert
4.3. MSCs affect B cells proliferation and maturation an important function in B lymphocyte-related immune
regulation [99]. Another recent study highlighted galectin-9
B cells are a type of lymphocyte in the humoral immunity (Gal-9) strongly upregulated upon activation of the cells by
of the adaptive immune system specialized in antigen pre- IFN-g. And their results showed that Gal-9 is a major mediator
sentation and antibody production. Recent studies also showed of the anti-proliferative and functional effects of MSCs not
8 Q. Zhao et al. / Journal of Cellular Immunotherapy 2 (2016) 3e20

only on T cells but also on B cells. Moreover, Gal-9 and results in a significant down-regulation of CCR7 and CD49d,
activated MSCs contribute to the suppression of antigen trig- two molecules involved in DCs homing to lymphoid organs.
gered immunoglobulin release [100]. By contrast, Rosado This event leads to inhibition of migration to the draining
et al. reported that BM-MSCs are able to promote in vitro lymph nodes and subsequent impairment of priming of
proliferation and differentiation of transitional and naive B antigen-specific naive T cells [109] (Fig. 2).
cells isolated from both healthy donors (HDs) and pediatric
patients with Systemic Lupus Erythematosus (SLE) upon 4.5. MSCs preserve neutrophils viability and function
stimulation with CpG, soluble CD40L, anti-Ig antibodies and
IL-2 [101]. Meanwhile, the results by our group show that Neutrophils form an essential part of the innate immune
treatment with UC-MSCs resulted in an increase of prolifer- system. They are differentiated from HSC in the BM. Neutro-
ation, differentiation of B cells into plasma cells and produc- phil progenitors have a quite high proliferation rate and mature
tion of antibodies in vitro. And our results also showed that neutrophils have a very short lifespan. Neutrophils are one type
PGE2 partially mediated the immunosuppressive activity of of phagocytes and are found in the peripheral blood, tissues and
MSC [102]. These conflicting results hinted that it is important BM. Neutrophils possess the capability of chemotaxis,
to distinguish the direct action of MSCs on B cells from in- phagocytosis and bactericide. During the beginning phase of
direct effects mediated by other cell types contained in the inflammation, neutrophils are one of the first-responders of
different culture conditions. inflammatory cells to migrate towards the site of inflammation.
MSCs inhibit in vitro apoptosis of resting and IL-8-activated
4.4. MSCs modulate dendritic cells (DCs) generation neutrophils and reduce N-formyl-l-methionin-l-leucyl-L-
and maturation phenylalanine (f-MLP)-induced respiratory burst while not
affecting phagocytosis, expression of adhesion molecules, or
DCs are the main APCs in the mammalian immune system. the migration capability of neutrophils in response to classic
Their main function is to present antigen material on the cell stimuli. And further research showed that MSCs rescued neu-
surface to the T cells. DCs affect significantly the balance trophils from apoptosis by constitutive release of IL-6 [110].
between helper and regulatory T cell and establish tolerance to Cassatella et al. reported that MSCs, upon TLR activation, may
self antigen [103]. The presence of MSCs blocked the differ- sustain and amplify the functions of neutropils. Their results
entiation of peripheral and umbilical cord blood derived showed that TLR3-activated MSCs are powerful in preserving
CD14 þ CD1a  precursors into dermal/interstitial DCs neutropils viability and function, and a concerted action of
[104]. Coculture of MSCs with DCs resulted in reduced endogenously produced IL-6, IFN-b, and GM-CSF determines
expression of CCR7 by DCs following stimulation. Likewise, most of the modulatory effects exerted on PMN by TLR3-
DCs matured in the presence of MSCs, showed significantly activated MSC [111]. The results reported by Maqbool et al.
less migration to CCL19 [105]. Zhang et al. showed that also indicated MSCs rescue neutrophils from nutrient- or
MSCs inhibit the up-regulation of CD1a, CD40, CD80, CD86, serum-deprived cell death [112] (Fig. 2). Therefore, neutrophils
and HLA-DR during DCs differentiation and prevent an in- may be one of most important immune cells mediated the
crease of CD40, CD86, and CD83 expression during DCs forward immunomodulatory of MSCs.
maturation [80]. In addition, Jiang et al. also showed that
MSCs coculture could strongly inhibit the initial differentia- 4.6. MSCs induces macrophage M1/M2 phenotype
tion of monocytes to DCs. In addition, mature DCs treated transformation
with MSCs were significantly reduced in the expression of
CD83, suggesting their skew to immature status. Meanwhile, Macrophages are differentiated from monocytes residing in
decreased expression of presentation molecules (HLA-DR and tissues. Macrophages play a key role in innate immune system.
CD1a) and costimulatory molecules (CD80 and CD86) and They are highly specialized in removal of dying or dead cells and
down-regulated IL-12 secretion were also observed [106]. cellular debris. And macrophage also is important immune
Moreover, the differentiation of BM progenitors into DCs effector cell involving cell mediated immune response.
cultured with conditioned supernatants from MSCs was partly Increasing evidences have demonstrated that MSCs-mediated
inhibited through the secretion of IL-6 [107]. Furthermore, regulation of macrophages is critical for inflammation response
MSCs secreted PGE2 plays a major role in MSC-mediated and tissue injury repair. Nemeth et al. reported that injection of
inhibition of DCs maturation, and inhibitory effect on DCs BM-MSCs can beneficially modulate the response of the host
maturation by acting early, during the progression from immune system to sepsis and significantly improve animal sur-
monocytes to iDCs induced by GM-CSF and IL-4, although vival [113]. And their results suggest that the injected MSCs
they do not interfere with LPSeinduced iDC differentiation interact with circulating and tissue monocytes and macrophages
into mDCs [108]. Additionally, MSCs inhibit in vitro DCs and reprogram them. Treated monocytes and macrophages pro-
effector properties, including antigen processing and presen- duce large amounts of IL-10, and the treatment decreases the
tation to T cells through the inhibition of the activation of amounts of circulating TNF-a and IL-6. Cao et al. transplanted
mitogen-activated protein kinases (MAPKs) occurring upon mouse MSCs into the pancreas of the streptozotocin-treated
TLR4 stimulation. In addition, in vitro exposure of DCs to hyperglycemic isogeneic mice, resulting in a decrease in blood
MSCs as well as in vivo intravenous administration of MSCs glucose due to a recovery in b cell mass. Their analysis suggest
Q. Zhao et al. / Journal of Cellular Immunotherapy 2 (2016) 3e20 9

that the grafted MSCs recruited M2 macrophages in a stromal increase in alveolar macrophages and inhibited hallmark fea-
cell-derived factor 1 (SDF-1)-dependent manner, which activates tures of asthma, including airway hyperresponsiveness, eosin-
Wnt/b-catenin signaling in pancreatic bcells to promote b cell ophilic accumulation, and Th2 cytokine production.
replication [114]. Another recent study demonstrated that mouse Importantly, selective depletion of this macrophage compart-
MSCs skewed macrophages towards an M2 phenotype in a cell ment reversed the therapeutic benefit of MSCs treatment on
contact-independent manner. MSCs exerted this effect through airway hyperresponsiveness [121]. Lee et al. established one
the inhibition of NF-kB p65 and the activation of STAT3 humanized mouse model by injecting isolated hUCB-derived
pathways [115]. In addition, preferential shift of the macrophage CD34 þ cells intravenously into immunocompromised NOD/
phenotype from M1 to M2 may be related to the immune- SCID IL2g null mice. After repeated intravenous injection of
modulating characteristics of MSCs [116,117] (Fig. 3). There- hPBMSCs or MRC5 cells into these mice, immunological al-
fore, the discovery of monocytes and M1 macrophage trans- terations including T cell proliferation and increased IFN-g,
forming into M2 macrophage in presence of MSCs may be quite TNF-a, and human IgG levels, were observed. In contrast,
significant for inflammatory biology development and inflam- hUCB-MSCs injection did not elicit these responses [122].
matory disease treatment in future. Meanwhile, Zanotti et al. reported that the use of encapsulated
cells unequivocally demonstrates that MSCs do not require
4.7. MSCs inhibit immune cells in vivo homing to specific organs or cellecell contacts to control
inflammation and their immunosuppressive action is based on
The immunomodulatory capacity of MSCs has also been the release of soluble factors that may act systemically [123]. In
evaluated in vivo. First, in vivo administration of MSCs pro- addition, anti-inflammatory is another important capability of
longed the survival of skin graft [72]. In addition, results MSCs and was confirmed in vivo too. Nemeth et al. showed that
demonstrated that MSCs do ameliorate mice experimental an intravenous injection of MSCs can beneficially modulate the
autoimmune encephalomyelitis (EAE) [90]. Allogeneic MSCs response of the host immune system to sepsis and improve
improve the survival of corneal allografts without engraftment survival. And their results suggested that the injected MSCs
and primarily by secreting TSG-6 that acts by aborting early interact with monocytes and macrophages in circulating and
inflammatory responses [118]. AT-MSCs therapy efficiently tissue and reprogram them. Treated monocytes and macro-
ameliorates autoimmune diabetes pathogenesis in diabetic phages produce large amounts of IL-10, and the treatment de-
NOD/SCID mice by attenuating the Th1 immune response creases the amounts of circulating TNF-a and IL-6 [113]. This
concomitant with the expansion/proliferation of Treg cells, reduces harm caused by unbridled immune responses to the host
thereby contributing to the maintenance of functional b-cells tissues. Another recent study showed that systemic infusion of
[119]. In established chronic colitis, therapeutic injection of MSCs significantly ameliorated the clinical and histopatholog-
activated macrophages by coculture with AT-MSCs alleviated ical severity of colitis, abrogating weight loss, diarrhea and
its progression and avoided disease recurrence. And AT-MSCs inflammation, and increasing survival. And their results
protected from severe sepsis by reducing the infiltration of in- demonstrated that the therapeutic effect was associated with
flammatory cells into various organs and by downregulating the downregulation of the Th1-driven inflammatory responses.
production of several inflammatory mediators, where AT-MSCs MSCs decreased a wide panel of inflammatory cytokines and
mediated macrophages derived IL-10 played a critical role chemokines and increased IL10, acting on macrophages [124].
[120]. In addition, administration of MSCs isolated from human Furthermore, systemic infusion of AT-MSCs on the activation
BM, umbilical cord, or adipose tissue provoked a pronounced state of macrophages inhibited colitis in mice, reducing mor-
tality and weight loss while lowering the colonic and systemic
levels of inflammatory cytokines [120]. However, ambiguous
results were reported that intravenously infused C57BL/6 green
fluorescent protein (GFP) MSCs home to the lungs in C57BL/6
recipient mice and induce an inflammatory response. This
response was characterized by increased mRNA expression of
monocyte chemoattractant protein-1 (MCP1), IL1-b, and TNF-
a and an increase in macrophages in lung tissue after MSCs
infusion [125]. These results of pre-clinical studies showed that
MSCs possess broad immunomodulatory and anti-
inflammatory activity in disease animal model in vivo and hin-
ted us MSCs may be a promising candidate for potential clinical
application in treating immune-based disorders in future.

4.8. Factors affecting the immunomodulatory activity of


Fig. 3. MSCs promote the survival of monocytes and induce differentiation MSCs
toward macrophage M2 anti-inflammatory phenotype that express CD206 and
CD163. In addition, activated MSCs secrete prostaglandin E2 (PGE2) that
drives resident macrophages with an M1 proinflammatory phenotype toward to Evidences showed that inflammation environment can
an M2 anti-inflammatory phenotype. change the immunomodulation activity of MSCs. Inflamed
10 Q. Zhao et al. / Journal of Cellular Immunotherapy 2 (2016) 3e20

periodontal ligament stem cells showed significantly dimin- induction of lytic activity against MSCs [84]. Another study by
ished inhibition of T-cell proliferation. In cocultures, stimu- Spaggiari et al. indicated that the activated NK cells by IL-2
lated PBMSCs showed significantly less induction of explosion, but not freshly isolated NK cells, displayed strong
CD4þCD25 þ FOXP3þ regulatory T cells and IL-10 secre- cytolytic activity. Meanwhile, their investigation showed that the
tion in the presence of inflamed compared with healthy peri- expression of NKp30, NKG2D and DNAM-1 in NK cells
odontal ligament stem cells. Furthermore, suppression of Th17 involved in the lysis of MSCs [93]. In addition, Eugela et al.
differentiation and IL-17 production by inflamed periodontal investigated the effect of Tregs against MSCs. Their results
ligament stem cells was significantly lesser than by healthy showed that kidney perirenal AT-MSCs and originating Tregs
cells [126]. Waterman et al. have suggested that MSCs may from healthy donors do not impair each other's suppressive effect
polarize into two distinctly acting phenotypes following spe- on allo-reactivity [133]. Actually, There are still lots of work to
cific TLR stimulation, resulting in different immune modula- do to clear the effect on MSCs by other immune cells now.
tory effects and distinct secretomes. The TLR4-primed MSCs
population exhibits a proinflammatory profile (MSC1) and the 5. Therapeutic application of MSCs for immune diseases
TLR3-primed MSCs population delivers anti-inflammatory
signals (MSC2) [127]. Furthermore, Micro RNA expression Currently, there are many promising clinical trials using
level also is another important factor affect the immunomo- MSCs in cell-based therapies of numerous diseases. MSCs
dulation capability. Reported results showed that miR-181a suppress T-cells, B cells and DCs function and represent a
[128] and miR-155 [129] reduces the immunosuppressive promising strategy for cell therapy of immune-mediated dis-
capacity of inflammatory cytokine-activated MSCs. In addi- eases. The immunomodulatory activities of MSCs provide a
tion, Evidence had also shown that different cell subsets show rational basis for their application in the treatment of immune-
different immunomodulation activity. Yang et al. reported mediated diseases. Our analysis below will include the most
CD106þ chorionic villi(CV)-MSCs possess high immunomo- frequently studied immune mediated diseases: GVHD, AA,
dulation activity, whereas CD106- CV-MSCs possess high MS, RA, CD and SLE.
colony formation capacity. In addition, the immunosuppres-
sive activity of MSCs is highly related to their ability for the 5.1. MSCs and GVHD
secretion of immunoregulatory cytokines and PGE2. In
CD106þ MSCs, a list of genes was up regulated, including GVHD is a difficult and potentially lethal complication of
COX-2, IL-1a, IL-1b, IL-6 and IL-8 [33]. Recently, chemical hematopoietic stem cell transplantation (HSCT). This can
that can correct the MSCs biological function was also re- occur in up to 30e50% of patients for HLA matched sibling
ported. The research by Ma et al. showed that Immunomod- transplant and even more frequently in HLA-mismatched un-
ulatory agent thalidomide corrects impaired MSCs function in related donor transplants (60e80%) [134]. GVHD is normally
inducing tolerogenic DCs in patients with immune thrombo- treated with corticosteroid and other immunosuppressive
cytopenia. The thalidomide modulated MSCs from ITP pa- therapy [135,136]. However, response of GVHD to steroid
tients induced mature DCs to become tolerogenic DCs. And therapy is between 20% and 40%, mortality of GVHD with
the induction of tolerogenicity in DCs by MSCs was depen- refractory to steroid therapy approaches 80% [137]. MSCs can
dent on the expression of TIEG1 in DCs [130]. Furthermore, be expanded in culture and possess complex and diverse
recent experiments clearly documented the inhibitory effect of immunomodulatory activity. Moreover, human MSCs carry
gamma irradiation on proliferation of MSCs, but does not low levels of class 1 and no class 2 HLA antigens, making
impair the immunosuppressive capacity of BM-MSCs in vitro them immunoprivileged and able to be used without HLA
and thus might increase the safety of MSC-based cell products matching. Increased evidences have shown that MSCs may be
in clinical applications [131]. Zhang et al. reported that MSCs promising cell product for GVHD treatment (Table 1). First
from late passage displayed a stronger immunosuppressive clinical trial displayed that BM-MSCs have a potent immu-
activities. And their results also showed that increased nosuppressive effect in vivo, and induced complete response
interleukin-6 production may be a possible underlying mech- (CR) of aGVHD that is refractory to conventional immuno-
anism for enhanced immunomodulatory ability of MSCs suppressive therapy [138]. A later phase II clinical study from
[132]. These results hinted that some growth factors, cytokines the same group involved 55 steroid-resistant patients with
and chemicals may be used for correcting impaired MSCs severe aGVHD. Treatment with HLA-identical and hap-
function to benefit patients with MSCs deficiency in future. loidentical sibling donor BM or third-party mismatched BM-
MSCs induced a 70% initial response rate that was not
4.9. The effect of immune cells against MSCs related to age or HLA match. None of the patients had side
effects either during or immediately after the MSCs infusion
The interaction between MSC and immune cells is usually [139]. Ringden et al. injected MSCs to eight patients with
bidirectional. Sotiropoulou tested the ability of different cyto- steroid-refractory grades IIIeIV GVHD. GVHD disappeared
kines to induce NK cytotoxicity against MSCs, their results completely in six of eight patients. Two died soon after MSCs
show that NK cells cultured for 4 days in IL-15esupplemented treatment with no obvious response. Five patients are still alive
medium could effectively lyse MSCs. In addition, IL-2 and the 3 years post MSCs transplantation. Their survival rate was
combinations of IL-12/IL-15 and IL-12/IL-18 also resulted in significantly better than that of 16 patients with steroid-
Q. Zhao et al. / Journal of Cellular Immunotherapy 2 (2016) 3e20 11

resistant biopsy-proven gastrointestinal GVHD, not treated

MSC, mesenchymal stem cell; BM, bone marrow; aGVHD: acute graft versus host disease; cGVHD: chronic graft versus host disease; CR: complete response; PR: partial response; NR: no response; FP: follow up
(Herrmann et al., 2012)
(Herrmann et al., 2012)
(Le Blanc et al., 2004)

(Le Blanc et al., 2008)


(Ringden et al., 2006)

(Ringden et al., 2013)


with MSCs during the same period (P ¼ 0.03) [140]. Herr-

(Muroi et al., 2013)


(Ball et al., 2013)
mann et al. also undertook a phase I trial in patients suffer
from steroid-refractory aGVHD and cGVHD utilizing BM-
References

MSCs. The response rate overall for aGVHD was complete


in seven, partial in four and no response in one patient. Two
patients with cGVHD achieved CR with two partial responses
(PR) and three with no response (NR). The survival for those
Improved
Improved
Improved

Improved
Improved
Improved
Improved
Improved
Outcome

with aGVHD who achieved a complete response compared


with those who did not was significant (p ¼ 0.03) [141].
Further research showed that MSCs infusions are safe and
effective for children with steroid-refractory aGVHD, espe-
21 Months
12 months

30 months
30 months

96 weeks
2.9 years

cially when applied early in the disease course [142]. In


3 years
2 years

addition, Muroi et al. conducted a multicenter phase I/II study


FP

using BM-MSCs manufactured from healthy unrelated vol-


unteers to treat steroid-refractory aGVHD. By week 4, 13 of
NR(%)

14 patients (92.9%) had responded to MSCs therapy with a CR


(n ¼ 8) or PR (n ¼ 5). At 24 weeks, 11 patients (10 with CR
29.1

42.9
13.5
8.3

7.1
25

25
e

and 1 with PR) were alive. At 96 weeks, 8 patients were alive


in CR. Their results showed that third party-derived BM-
PR(%)

MSCs may be safe and effective for patients with steroid-


16.4

33.3
28.5
21.6

35.7

refractory GVHD [143]. Meanwhile, Ringden et al. tested


50
e
e

placenta derived MSCs (PDMSCs) for treatment of steroid-


CR(%)

refractory aGVHD. Nine patients who had undergone HSCT


and who had developed steroid-refractory grade IIIeIV
54.5

58.3
28.5

57.1
100
75

65
25

aGVHD were treated by PDMSCs infusion. Their results


showed that there was an overall response rate of 75% [144].
Side effect

The results by Ringden et al. showed that MSCs derived from


1 seizurs
(patient)

other tissue also can be tried to treat GVHD.


There have been several studies using MSCs as prophylaxis
e
e
e

e
e
e

to prevent GVHD and promote HSCs engraftment and ob-


Infusions

tained useful and inspiring results (Table 2). In phase I studies,


2e19
2e11
1e13

8e16
1e5

Lazarus et al. estimated the feasibility of transplanting autol-


15
1
1

ogous or allogeneic MSCs to improve engraftment of HSCs, as


well as to reduce GVHD [145,146]. Kuzmina et al. accom-
(1.3e2.7)  106/kg
(1.3e2.7)  106/kg

(0.9e2.8)  106/kg
(0.7e9)  106/kg
(0.4e9)  106/kg

(0.9e3)  106/kg

plished a Phase II clinical trial and investigated that the pro-


phylaxis of MSCs for aGVHD. Their results showed that the
2  106/kg

2  106/kg

patients with BM-MSCs infusion have significant lower rate of


grade II-IV aGVHD (5.3%) compared with patients without
Dose

BM-MSCs infusion (38.9%) of patients (P ¼ 0.002) [147].


Several researches have shown that MSCs from amnion,
Sibling, haploidentical
Donor source of MSC

placenta, and umbilical cord can be potentially used for


Allogeneic placenta
and third-party BM
Haploidentical BM

substituting BM-MSC in several therapeutic applications,


Clinical trials of using MSCs treatment for GVHD.

third-party BM

third-party BM
third-party BM
third-party BM

including the treatment of GVHD [9,79,148]. Therefore, our


group tested to cotransplant the culture-expanded UC-MSCs
in 50 people with refractory/relapsed hematologic malignancy
BM

undergoing haplo-HSCT with myeloablative conditioning.


Grade II-IV aGVHD was observed in 12 of 50 (24.0%) pa-
GVHD grade

tients. cGVHD was observed in 17 of 45 (37.7%) patients and


was extensive in 3 patients. The probability that patients
IIIeIV

IIIeIV
ⅢeⅣ
ⅡeIV

ⅡeIV

ⅡeIII

would attain progression-free survival at 2 years was 66.0%.


period; -: not observed.
IV

Our research indicated that MSCs cotransplantation with


HSCs is effective in improving donor engraftment and
No. of patients

reducing severe GVHD [149]. Meanwhile, our group observed


12 aGVHD

that Patients with severe AA (n ¼ 17) received haploidentical


7 cGVHD
Table 1

HSCT plus MSCs infusion. The 3-month and 6-month survival


55

37

14

rates for all patients were 88.2% and 76.5%, respectively;


1
8

9
12 Q. Zhao et al. / Journal of Cellular Immunotherapy 2 (2016) 3e20

mean survival time was 56.5 months. Therefore, our results

MSC, mesenchymal stem cell; AA: aplastic anemia; BM, bone marrow; aGVHD: acute graft versus host disease; cGVHD: chronic graft versus host disease; PBS: Phosphate Buffer solution; FP: follow up period;
(Kuzmina et al., 2012)
(Lazarus et al., 2005)
showed that Combined transplantation of haploidentical HSCs

(Wu et al., 2013)


(Wu et al., 2014)
(Li et al., 2014)
and MSCs on severe AA without an HLA-identical sibling
donor was safe, effectively reduced the incidence of severe
References

GVHD, and improved patient survival [150]. Then another


clinical trial of AA investigated by our group and suggested
that the modified conditioning regimen without high-dose
immunosuppressive agents was sufficient to achieve sus-
Improved

Improved
Improved
Improved
Improved
Outcome

tained donor engraftment in which the third-party donor-


derived UC-MSCs may play an important role [151]. Although
cell dose is much less in our clinical trial (5  105/kg) than in
clinical trial (14  105) run by Le Blanc K et al., our clinical
32 months

58 months
78 months
2 years

3 years

trial also achieved ideal therapeutic effect. This is also


consistent to our recent data that show that PGE2 which plays
FP

a key role in immunomodulation was secreted much more in


cGVHD(%)

UC-MSCs than BM-MSCs. Therefore, PGE2 maybe can be


MSC:21.05

using as a biological effectiveness to evaluate immunomodu-


PBS: 31.6

lation potency of MSCs (not published). These results showed


14.2%

that MSCs derived from kinds of tissue may be safe and


61

32
35

effective for prevention of GVHD.


aGVHD(%)
Grade IIIeIV

5.2. MSC and acquired AA


PBS: 5.26
MSC: 0

23.5%
23.8

AA is mostly considered an immune-mediated BM failure


28

14

syndrome, characterized by hypoplasia and pancytopenia with


fatty BM and reduced angiogenesis. Previous investigations
aGVHD(%)

have demonstrated that acquired AA is manifested as abnor-


PBS: 47.36
MSC: 31.6
Grade ⅠeⅡ

malities of HSCs/HPCs and hematopoietic microenvironment


[152]. Lots of evidences have hinted that AA might be a
33.3
35.2
28
e

syndrome characterized by stem/progenitor-cell disorders


including HSCs/HPCs and BM-MSCs. BM-MSCs support
(2.87e10)  106/kg
(0.9e1.3)  106/kg

hematopoiesis and regulate almost overall immune cells


(1e5)  106/kg

function to maintain the hematopoietic and immune homeo-


5.0  105/kg
5.0  105/kg

stasis [97,139]. BM-MSCs can modulate the major immune


cell functions including T cells, B cells, monocytes, DCs,
Dose

NKTs and neutrophils [153,154]. BM-MSCs possess remark-


able immunosuppressive properties on Th1, Th17 and CTLs.
Allogeneic unbilical cord
Allogeneic unbilical cord
Allogeneic unbilical cord

BM-MSCs inhibit the proliferation of T cells, IFN-g and TNF-


Donor source of MSC

a secretion by Th1 cells while promoting IL-10 production by


Th2 cells and the expansion of Treg cells. However, recent
researches showed that BM-MSCs from AA patients had poor
proliferation and deficient immune suppression of MLR, PHA-
Clinical trials of using MSCs prophylaxis for GVHD.

induced T cell activation and IFN-g release [155,156]. Our


BM
BM

recent study showed that BM-MSCs from AA patients were


reduced in suppressing the proliferation and clonogenic po-
No. of patients

tential of CD4þ T cells while promoting Treg cells expansion.


BM-MSCs were also defective to suppress the production of
TNF-a and IFN-g by CD4þ cells. However, there was no
significant difference in regulating the production of IL-4, IL-
46
37

50
21
17

10 and IL-17 [157]. In addition, our research also showed that


hematologic malignancy
hematologic malignancy

hematologic malignancy

BM-MSCs from AA patients showed aberrant morphology,


decreased proliferation and clonogenic potential and increased
apoptosis than BM-MSCs from healthy controls. BM-MSCs
-: not observed.

from AA patients were susceptible to be induced to differen-


tiate into adipocytes but more difficult to differentiate into
Disease
Table 2

osteoblasts. Consistent with abnormal biological features, a


AA
AA

large number of genes implicated in cell cycle, cell division,


Q. Zhao et al. / Journal of Cellular Immunotherapy 2 (2016) 3e20 13

proliferation, chemotaxis and hematopoietic cell lineage mediated and humoral immunity [163]. Currently there is still
showed markedly decreased expression in BM-MSCs from no effective choice for MS treatment. Given MSCs putative
AA patients. Conversely, more related genes with apoptosis, roles in immunosuppression and neural repair, they have also
adipogenesis and immune response showed increased recently been proposed as treatment for MS. In addition, the
expression in BM-MSCs from AA patients. The gene results showed that patient MSCs exhibited phenotypic
expression profile of BM-MSCs further confirmed the changes, distinct transcriptional profile and functional defects
abnormal biological properties and provided significant evi- implicated in MSC immunomodulatory and immunosuppres-
dence for the possible mechanism of the destruction of the BM sive activity [164]. Currently, close to 17 clinical trials are
microenvironment in AA [158]. registered to use MSCs therapy for the treatment of MS (http://
MSCs may be a promising therapeutic candidate as a www.clinicaltrials.gov/). MSCs have well been tolerated and
treatment for AA due to following two quite important facts, were safe in patients with early phase clinical studies. Autol-
① hematopoiesis supporting and potent immunosuppressive ogous MSCs were safely given to patients with secondary
capability of MSC and ② biological characteristical difference progressive MS. The efficacy results demonstrated the evi-
and functional capability deficiency of BM-MSCs derived dence of structural, functional, and physiological improvement
from patients with AA. However, till today there is just one after treatment in some visual endpoints is suggestive of
clinical trials using MSCs alone was accomplished by Xiao neuroprotection [165]. Mohajeri et al. investigated the mo-
et al. Xiao et al. investigated that the safety and efficacy post lecular mechanism of MSCs infusion for MS treatment. They
intravenous administration of MSCs to 18 patients with AA. found that the expression of FoxP3 after intrathecal injection
BM-MSCs ((5.0e7.1)  105/kg body weight) were injected of MSCs was significantly higher than the levels prior to
intravenously to 18 patients, including 14 patients with non- treatment. Such significant enhanced expression of FoxP3
severe AA and four patients with severe AA who were re- associated with clinical stability [166]. Findings from this pilot
fractory to prior immunosuppressive treatment. Two patients study further support the potential of MSCs for treatment of
had injection-related adverse events, including transient fever MS patients. A patient with MS was transplanted with mul-
and headache. No major adverse events were reported during tiple allogeneic hUC-MSC. The results also showed allogeneic
the follow-up period. An immunological analysis revealed an hUC-MSC may be a safe, effective, and more practical source
increased proportion of CD4(þ)CD25(þ) FOXP3(þ) regula- of stem cells for the treatment of MS [167]. However, obvi-
tory T cells [159]. Therefore, their results showed that MSCs ously lots of effective trials still will be needed before MSCs
are safety and efficacy for AA treatment. In another recent can be used for MS clinical treatment in future.
study, Ozdogu H et al. summarized that the result of post-
transplant treatment with MSCs of a 26-year-old patient 5.4. MSCs and RA
with AA complicated by invasive sino-orbital aspergillosis.
The patient was treated with MSCs to benefit from the dual RA is the commonest autoimmune joint disease that has
effects of MSCs in immune reconstitution: suppression against achieved significant therapeutic advances in the past decades,
alloreactive T cells and facilitation of the re-engraftment but remains difficult to treat in a subset of cases. Therefore,
process. The patient did not develop acute or chronic Wang et al. firstly tried to treat RA by using MSCs infusion. In
GVHD. And the aspergillus infection healed completely. The a clinical trial for RA treatment by Wang, 172 patients with
engraftment failure was also ended without any complications active RA who had inadequate responses to traditional medi-
[160]. In addition, there are several reports pointed that the cation were enrolled. Patients were divided into two groups for
patients achieved rapid hematopoietic engraftment of donor different treatment: disease-modifying anti-rheumatic drugs
origin and lower rate of acute or chronic GVHD was observed (DMARDs) plus medium without UC-MSCs, or DMARDs
post cotransplantation MSCs with HSCs [161,162]. BM-MSCs plus UC-MSCs group (4  107 cells per time) via intravenous
in patients with AA are abnormal. However, allogenic MSCs injection. Their results showed that no serious adverse effects
will be eliminated soon after transplantation. The efficacy of were observed during or after infusion. The serum levels of
MSCs was mainly achieved through the secretion of couples TGF-а and IL-6 decreased after the first UC-MSCs treatment
of immune factors. Abnormal MSCs would not be replaced. (P < 0.05). The percentage of CD4(þ)CD25(þ)Foxp3(þ)
So it is totally possible the efficacy of MSCs in clinical regulatory T cells of peripheral blood was increased
treatment is transient. Therefore, according to our under- (P < 0.05). The therapeutic effects maintained for 3e6 months
standing MSCs should be applied for couple of times to without continuous administration, correlating with the
maintain the therapeutic effect. However, it is still need to be increased percentage of Treg cells of peripheral blood.
clear according to the controlled pilot or clinical trial. Repeated infusion after this period can enhance the therapeutic
efficacy. Therefore, treatment with DMARDs plus UC-MSCs
5.3. MSCs and MS may provide safe, significant, and persistent clinical benefits
for patients with active RA [62]. In another recent phase I
MS is the most common neurological disease in young clinical trial by Liang et al., four patients with persistently
adults, affecting approximately two million people worldwide. active RA underwent MSCs transplantation. Three of four
MS is an autoimmune disorder of the central nervous system patients received a reduction in erythrocyte sedimentation rate,
where myelin and oligodendrocytes are targeted by cell DAS-28, and visual analog scale pain score at 1 and 6 months
14 Q. Zhao et al. / Journal of Cellular Immunotherapy 2 (2016) 3e20

after transplantation. No one had achieved the DAS-28-defined in the pathogenesis of SLE. And numerous studies have shown
remission in the follow-up period. No serious adverse events that p16 [179], Wnt/b-catenin and p53/p21 pathway plays critical
were reported. Their results demonstrated that allogeneic role in the senescent of SLE BM-MSCs [180e182]. A pilot
MSCT is a one of safe treatment choice for severe and resis- single-center clinical studies reported the safety and efficacy of
tant RA [168]. allogeneic BM-MSCs and UC-MSCs in treating drug-resistant
SLE patients, and the clinical results have been encouraging
5.5. MSCs and CD [183]. In addition, Li et al. investigated that the roles of MSCs
transplantation in SLE patients with refractory cytopenia. Thirty-
CD is a chronic inflammatory bowel disease characterized five SLE patients with refractory cytopenia were enrolled in a
by a relapsing-remitting clinical behavior. The main feature of MSCs transplantation trial. The results suggested that MSCs
CD is intestinal inflammation. In recent years several groups transplantation could reverse hematological aberration in SLE
tried to use MSCs to treat CD and obtained some promising patients with refractory cytopenia, which might be associated
results. The results of a phase I study showed that adminis- with reconstitution of Treg and Th17 cells [184]. Wang et al.
tration of autologous BM-MSCs appears safe and feasible in observed the long-term safety and efficacy of allogeneic MSCs
the treatment of refractory CD [169]. Ciccocioppo et al. transplantation in treatment-resistant SLE patients. Eighty-seven
enrolled 12 consecutive outpatients with CD refractory or patients with persistently active SLE who were refractory to
unsuitable for current available therapies. Ten patients (two standard treatment or had life-threatening visceral involvement
refused) received intrafistular MSCs injections. All patients were enrolled. Allogeneic BM-MSCs and UC-MSCs were
who underwent MSCs infusion sustained complete closure intravenously. These results showed that allogeneic MSCs
(seven cases) or incomplete closure (3 cases) of fistula tracks transplantation resulted in the induction of clinical remission and
with a parallel reduction of CD and perianal disease activity improvement in organ dysfunction in drug-resistant SLE patients
indexes, and rectal mucosal healing were induced by treat- [185]. A recent multicenter clinical study by Wang et al. found
ment. In addition, the percentage of mucosal and circulating that intravenous UC-MSCs transplantation resulted in clinical
Treg cells significantly increased during the treatment and disease remission and systemic amelioration in lupus patients
remained stable until the end of follow up [170]. The results who are refractory to other. However, some patients had disease
from two phase I/IIa clinical trial demonstrated that allogeneic relapses after 6 months; therefore, they suggested that a repeated
AT-MSCs also is a simple, safe, and beneficial therapy for MSCs infusion is feasible and necessary after 6 months to avoid
perianal fistula in CD patients [171,172]. In another phase 2 disease relapse [186].
study, administration of allogeneic MSCs reduced CD activity
index and CD endoscopic index of severity scores in patients 5.7. MSCs and inflammatory disease
with CD luminal refractory to biologic therapy [173].
MSCs are also used in human clinical trials for the treat-
5.6. MSCs and SLE ment of inflammatory. MSCs modulate inflammation by
decreasing the immune cells and products of the inflammatory
SLE is a common and potentially fatal autoimmune disease response [187]. Skrahin et al. accomplished an open-label
characterized by autoantibodies associated with multiorgan phase 1 safety trial. They infused autologous MSCs as
injury, including the renal, cardiovascular, neural, musculoskel- adjunct treatment in patients with multidrug and extensively
etal and cutaneous systems. Although disease severity and organ drug-resistant tuberculosis. Their results showed that MSCs as
involvement vary significantly among SLE patients, abnormal- an adjunct therapy are safe and can now be explored further
ities of T and B lymphocytes are universal [174,175]. Conven- for the treatment of patients with MDR or XDR tuberculosis in
tional immunosuppressive therapies, such as cyclophosphamide combination with standard drug regimens [188].
and mycophenolate mofetil can control disease in most, but not
all, patients with lupus nephritis. There is a subset of lupus 5.8. MSCs and other immune disease
nephritis patients whose disease either does not respond or re-
lapses, and their prognosis remains poor. In addition, progressive Report showed that UC-MSCs could restore behavioral
immunosuppressive therapy may lead to the development of functions and attenuate the histopathological deficits of
serious infection, cumulative drug toxicity, and an increased risk experimental autoimmune encephalomyelitis over the long
of cardiovascular disease and malignancy [176]. Recent research term (i.e., 50 days) by suppression of perivascular immune cell
showed that there were abnormalities in actin cytoskeleton, cell infiltrations and reduction in both demyelination and axonal
cycling regulation, BMP/TGF-b, and MAPK signaling pathways injury in the spinal cord [189]. Zhang et al. tried to infuse UC-
in BM-MSCs from SLE patients [177]. Tang et al. found that MSCs to treat patients with HIV-1-infected immunological
activated NF-kB pathway in SLE derived BM-MSCs inhibits the nonresponders (INRs). Their results showed that all patients
BMP-2-induced osteoblastic differentiation through BMP/Smad tolerated the UC-MSCs transfusions well throughout the trial.
signaling pathway, suggesting that the impaired osteoblastic The UC-MSCs transfusions preferentially increased circulating
differentiation may participate in the pathology of osteoporosis in naive and central memory CD4 T-cell counts and restored HIV-
SLE patients [178]. Another research also found that BM-MSCs 1-specific IFN-g and IL-2 production in the INRs. These en-
from SLE patients exhibited senescent behavior and are involved hancements in immune reconstitution were also associated
Q. Zhao et al. / Journal of Cellular Immunotherapy 2 (2016) 3e20 15

with the reduction of systemic immune activation and inflam- Natural Science Foundation of China (81000196 and
mation in vivo. The data suggested that MSCs transplantation 81330015) and Tianjin Research Program of Application
may be used as a novel immunotherapeutic approach to Foundation and Advanced Technology (12JCZDJC25000).
reversing immune deficiency in HIV-1-infected INRs [190].

6. Perspective References

MSCs have become a subject of clinical research interest [1] Friedenstein AJ, Piatetzky II S, Petrakova KV. Osteogenesis in transplants
of bone marrow cells. J Embryol Exp Morphol 1966;16(3):381e90.
due to their easy isolation and in vitro large scale cultivated
[2] Friedenstein AJ, Chailakhyan RK, Latsinik NV, Panasyuk AF, Keiliss-
amplification, attractive potential for multi-lineage differenti- Borok IV. Stromal cells responsible for transferring the microenviron-
ation and hematopiesis supporting, growth factors production ment of the hemopoietic tissues. Cloning in vitro and retransplantation
and cytokines secretion, and potential immunomodulatory in vivo. Transplantation 1974;17(4):331e40.
capacity. In addition, MSCs is definitely safe and well-toler- [3] Zuk PA, Zhu M, Ashjian P, De Ugarte DA, Huang JI, Mizuno H, et al.
Human adipose tissue is a source of multipotent stem cells. Mol Biol
ated for use in cell therapy, which provide a striking candidate
Cell 2002;13(12):4279e95.
for degenerative diseases and immune mediated diseases. [4] Zhang HJ, Miao ZC, He ZP, Yang YX, Wang Y, Feng MF. The existence
Increased clinical evidence suggests that MSCs may have of epithelial-to-mesenchymal cells with the ability to support hemato-
great potential in the treatment and prevention of GVHD, AA, poiesis in human fetal liver. Cell Biol Int 2005;29(3):213e9.
MS, CD, SLE and some other immune mediated diseases. [5] Tondreau T, Meuleman N, Delforge A, Dejeneffe M, Leroy R,
Massy M, et al. Mesenchymal stem cells derived from CD133-positive
Nevertheless, clinical trials yielded ambiguous results on the
cells in mobilized peripheral blood and cord blood: proliferation, Oct4
effects of MSCs. These ambiguous findings might result from expression, and plasticity. Stem cells 2005;23(8):1105e12.
insufficient standardization during the MSCs extraction, [6] Zheng CL, Yang SG, Guo ZX, Dong CL, Liang L, Chen XJ, et al.
expansion and administration procedures and interindividual Human Multipotent mesenchymal stromal cells from fetal lung
MSCs donor differences. Therefore, to obtain maximal clinical expressing pluripotent markers and differentiating into cell types of
three germ layers. Cell Transplant 2009;18(10):1093e109.
benefit, lots of problems, including tissue sources, numbers of
[7] Fukuchi Y, Nakajima H, Sugiyama D, Hirose I, Kitamura T, Tsuji K.
cells, the optimal passage time in culture before use, cell Human placenta-derived cells have mesenchymal stem/progenitor cell
subpopulation and standardized process of cell production, potential. Stem cells 2004;22(5):649e58.
need to be solved before MSCs as a medicine applied widely [8] Sarugaser R, Lickorish D, Baksh D, Hosseini MM, Davies JE. Human
to treat patients. In addition, it is absolutely important to umbilical cord perivascular (HUCPV) cells: a source of mesenchymal
progenitors. Stem Cells 2005;23(2):220e9.
optimize process of treatment and be able to effectively
[9] Lu LL, Liu YJ, Yang SG, Zhao QJ, Wang X, Gong W, et al. Isolation
monitor and communicate the benefits and risks of a cell and characterization of human umbilical cord mesenchymal stem cells
therapy to a patient. Furthermore, a clear and rigorous phar- with hematopoiesis-supportive function and other potentials. Haema-
macokinetics and pharmacodynamics model of the mechanism tologica 2006;91(8):1017e26.
of action of transient cell therapies is absolutely necessary. [10] Huang GT, Gronthos S, Shi S. Mesenchymal stem cells derived from
dental tissues vs. those from other sources: their biology and role in
Expanding our understanding of the molecular mechanisms
regenerative medicine. J Dent Res 2009;88(9):792e806.
governing immunomodulatory properties of MSCs will enable [11] Hermida-Gomez T, Fuentes-Boquete I, Gimeno-Longas MJ, Muinos-
us to greatly improve their clinical efficacy. Most importantly, Lopez E, Diaz-Prado S, de Toro FJ, et al. Quantification of cells
further well-designed, randomized and controlled clinical tri- expressing mesenchymal stem cell markers in healthy and osteoarthritic
als should be designed for better understanding of the under- synovial membranes. J Rheumatol 2011;38(2):339e49.
[12] Park JC, Kim JM, Jung IH, Kim JC, Choi SH, Cho KS, et al. Isolation
lying biology and utilizing MSCs therapy for immune
and characterization of human periodontal ligament (PDL) stem cells
mediated disease in the future. In addition, the cell dose that (PDLSCs) from the inflamed PDL tissue: in vitro and in vivo evalua-
was using in clinical treatment is still totally different in tions. J Clin Periodontol 2011;38(8):721e31.
different group study. We suggest that we should determine the [13] Schwab KE, Hutchinson P, Gargett CE. Identification of surface
MSCs cell dose according to the biological effectiveness of the markers for prospective isolation of human endometrial stromal colony-
forming cells. Human Reprod 2008;23(4):934e43.
MSCs. Meanwhile, it is really urgent to develop the biological
[14] Sakaguchi Y, Sekiya I, Yagishita K, Ichinose S, Shinomiya K,
effectiveness of various MSCs biological functions. Muneta T. Suspended cells from trabecular bone by collagenase
digestion become virtually identical to mesenchymal stem cells ob-
Conflict of interst tained from marrow aspirates. Blood 2004;104(9):2728e35.
[15] Zhu H, Guo ZK, Jiang XX, Li H, Wang XY, Yao HY, et al. A protocol
for isolation and culture of mesenchymal stem cells from mouse
The authors wish to confirm that there are no conflicts of
compact bone. Nat Protoc 2010;5(3):550e60.
interest associated with this publication and there has been no [16] Wang Y, Han ZB, Ma J, Zuo C, Geng J, Gong W, et al. A toxicity study
significant financial support for this work that could have of multiple-administration human umbilical cord mesenchymal stem
influenced its outcome. cells in cynomolgus monkeys. Stem Cells Dev 2012;21(9):1401e8.
[17] Wang Y, Zhang Z, Chi Y, Zhang Q, Xu F, Yang Z, et al. Long-term cultured
mesenchymal stem cells frequently develop genomic mutations but do not
Acknowledgments
undergo malignant transformation. Cell Death Dis 2013:4. e950.
[18] Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R,
This work was supported by The National Basic Research Mosca JD, et al. Multilineage potential of adult human mesenchymal
Program of China (973 Program) (2011CB964802), National stem cells. Science 1999;284(5411):143e7.
16 Q. Zhao et al. / Journal of Cellular Immunotherapy 2 (2016) 3e20

[19] Simmons PJ, Torok-Storb B. Identification of stromal cell precursors in [39] Krampera M, Pizzolo G, Aprili G, Franchini M. Mesenchymal stem
human bone marrow by a novel monoclonal antibody, STRO-1. Blood cells for bone, cartilage, tendon and skeletal muscle repair. Bone
1991;78(1):55e62. 2006b;39(4):678e83.
[20] Barry FP, Boynton RE, Haynesworth S, Murphy JM, Zaia J. The [40] Noack S, Seiffart V, Willbold E, Laggies S, Winkel A, Shahab-
monoclonal antibody SH-2, raised against human mesenchymal stem Osterloh S, et al. Periostin secreted by mesenchymal stem cells supports
cells, recognizes an epitope on endoglin (CD105). Biochem Biophys tendon formation in an ectopic mouse model. Stem Cells Dev
Res Commun 1999;265(1):134e9. 2014;23(16):1844e57.
[21] Barry F, Boynton R, Murphy M, Haynesworth S, Zaia J. The SH-3 and [41] Dodin G, Andrieux M, al Kabbani H. Binding of ellipticine to beta-
SH-4 antibodies recognize distinct epitopes on CD73 from human lactoglobulin. A physico-chemical study of the specific interaction of an
mesenchymal stem cells. Biochem Biophys Res Commun antitumor drug with a transport protein. Eur J Biochem
2001;289(2):519e24. 1990;193(3):697e700.
[22] Martinez C, Hofmann TJ, Marino R, Dominici M, Horwitz EM. Human [42] Labovsky V, Hofer EL, Feldman L, Fernandez Vallone V, Garcia
bone marrow mesenchymal stromal cells express the neural ganglioside Rivello H, Bayes-Genis A, et al. Cardiomyogenic differentiation of
GD2: a novel surface marker for the identification of MSCs. Blood human bone marrow mesenchymal cells: Role of cardiac extract from
2007;109(10):4245e8. neonatal rat cardiomyocytes. Differentiation 2010;79(2):93e101.
[23] Xu J, Liao WB, Gu DS, Liang L, Du WT, Liu PX, et al. Neural [43] Wang JF, Wang LJ, Wu YF, Xiang Y, Xie CG, Jia BB, et al. Mesen-
ganglioside GD2 identifies a subpopulation of MSC in human umbilical chymal stem/progenitor cells in human umbilical cord blood as support
cord. Cell Physiol Biochem 2009;23(4-6):415e24. for ex vivo expansion of CD34(+) hematopoietic stem cells and for
[24] Majumdar MK, Keane-Moore M, Buyaner D, Hardy WB, chondrogenic differentiation. Haematologica 2004;89(7):837e44.
Moorman MA, McIntosh KR, et al. Characterization and functionality [44] Zhou SB, Wang J, Chiang CA, Sheng LL, Li QF. Mechanical stretch
of cell surface molecules on human mesenchymal stem cells. J upregulates SDF-1alpha in skin tissue and induces migration of circu-
Biomedical Sci 2003;10(2):228e41. lating bone marrow-derived stem cells into the expanded skin. Stem
[25] Majumdar MK, Thiede MA, Mosca JD, Moorman M, Gerson SL. Cells 2013;31(12):2703e13.
Phenotypic and functional comparison of cultures of marrow-derived [45] Padovan CS, Jahn K, Birnbaum T, Reich P, Sostak P, Strupp M, et al.
mesenchymal stem cells (MSCs) and stromal cells. J Cell Physiol Expression of neuronal markers in differentiated marrow stromal cells
1998;176(1):57e66. and CD133+ stem-like cells. Cell Transplant 2003;12(8):839e48.
[26] Le Blanc K, Tammik C, Rosendahl K, Zetterberg E, Ringden O. HLA [46] Kotton DN, Ma BY, Cardoso WV, Sanderson EA, Summer RS,
expression and immunologic properties of differentiated and undiffer- Williams MC, et al. Bone marrow-derived cells as progenitors of lung
entiated mesenchymal stem cells. Exp Hematol 2003a;31(10):890e6. alveolar epithelium. Development 2001;128(24):5181e8.
[27] Zhao Q, Ren H, Li X, Chen Z, Zhang X, Gong W, et al. Differentiation [47] Li Y, Xu W, Yan J, Xia Y, Gu C, Ma Y, et al. Differentiation of human
of human umbilical cord mesenchymal stromal cells into low immu- amniotic fluid-derived mesenchymal stem cells into type II alveolar
nogenic hepatocyte-like cells. Cytotherapy 2009;11(4):414e26. epithelial cells in vitro. Int J Mol Med 2014b;33(6):1507e13.
[28] Klyushnenkova E, Mosca JD, Zernetkina V, Majumdar MK, Beggs KJ, [48] Chien CC, Yen BL, Lee FK, Lai TH, Chen YC, Chan SH, et al. In vitro
Simonetti DW, et al. T cell responses to allogeneic human mesenchymal differentiation of human placenta-derived multipotent cells into hepa-
stem cells: immunogenicity, tolerance, and suppression. J Biomed Sci tocyte-like cells. Stem Cells 2006;24(7):1759e68.
2005;12(1):47e57. [49] Sam R. The role of epithelial to mesenchymal transdifferentiation in
[29] Weiss ML, Anderson C, Medicetty S, Seshareddy KB, Weiss RJ, end-stage renal disease. Int J Artif Organs 2004;27(9):744e50.
Vander-Werff I, et al. Immune properties of human umbilical cord [50] Singaravelu K, Padanilam BJ. In vitro differentiation of MSC into cells
Wharton's jelly-derived cells. Stem cells 2008;26(11):2865e74. with a renal tubular epithelial-like phenotype. Ren Fail
[30] Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini F, 2009;31(6):492e502.
Krause D, et al. Minimal criteria for defining multipotent mesenchymal [51] Karaoz E, Okcu A, Unal ZS, Subasi C, Saglam O, Duruksu G. Adipose
stromal cells. The International Society for Cellular Therapy position tissue-derived mesenchymal stromal cells efficiently differentiate into
statement. Cytotherapy 2006;8(4):315e7. insulin-producing cells in pancreatic islet microenvironment both in
[31] Wislet-Gendebien S, Hans G, Leprince P, Rigo JM, Moonen G, Rogister B. vitro and in vivo. Cytotherapy 2013;15(5):557e70.
Plasticity of cultured mesenchymal stem cells: switch from nestin-positive [52] Moriscot C, de Fraipont F, Richard MJ, Marchand M, Savatier P,
to excitable neuron-like phenotype. Stem Cells 2005;23(3):392e402. Bosco D, et al. Human bone marrow mesenchymal stem cells can ex-
[32] Battula VL, Treml S, Bareiss PM, Gieseke F, Roelofs H, de Zwart P, press insulin and key transcription factors of the endocrine pancreas
et al. Isolation of functionally distinct mesenchymal stem cell subsets developmental pathway upon genetic and/or microenvironmental
using antibodies against CD56, CD271, and mesenchymal stem cell manipulation in vitro. Stem Cells 2005;23(4):594e603.
antigen-1. Haematologica 2009;94(2):173e84. [53] Alviano F, Fossati V, Marchionni C, Arpinati M, Bonsi L, Franchina M,
[33] Yang ZX, Han ZB, Ji YR, Wang YW, Liang L, Chi Y, et al. CD106 et al. Term Amniotic membrane is a high throughput source for mul-
identifies a subpopulation of mesenchymal stem cells with unique tipotent Mesenchymal Stem Cells with the ability to differentiate into
immunomodulatory properties. PLoS One 2013;8(3). e59354. endothelial cells in vitro. BMC Dev Biol 2007;7:11.
[34] Bruder SP, Kurth AA, Shea M, Hayes WC, Jaiswal N, Kadiyala S. Bone [54] Bell E, Richardson GD, Jahoda CA, Gledhill K, Phillips HM,
regeneration by implantation of purified, culture-expanded human Henderson D, et al. Dermal stem cells can differentiate down an
mesenchymal stem cells. J Orthop Res 1998;16(2):155e62. endothelial lineage. Stem Cells Dev 2012;21(16):3019e30.
[35] Noel D, Gazit D, Bouquet C, Apparailly F, Bony C, Plence P, et al. Short- [55] Bernardo ME, Zaffaroni N, Novara F, Cometa AM, Avanzini MA,
term BMP-2 expression is sufficient for in vivo osteochondral differenti- Moretta A, et al. Human bone marrow derived mesenchymal stem cells
ation of mesenchymal stem cells. Stem Cells 2004;22(1):74e85. do not undergo transformation after long-term in vitro culture and do
[36] Gross-Aviv T, Vago R. The role of aragonite matrix surface chemistry not exhibit telomere maintenance mechanisms. Cancer Res
on the chondrogenic differentiation of mesenchymal stem cells. Bio- 2007;67(19):9142e9.
materials 2009;30(5):770e9. [56] Meza-Zepeda LA, Noer A, Dahl JA, Micci F, Myklebost O, Collas P.
[37] Lee HK, Lee BH, Park SA, Kim CW. The proteomic analysis of an High-resolution analysis of genetic stability of human adipose tissue
adipocyte differentiated from human mesenchymal stem cells using stem cells cultured to senescence. J Cell Mol Med 2008;12(2):553e63.
two-dimensional gel electrophoresis. Proteomics 2006;6(4):1223e9. [57] Poloni A, Maurizi G, Babini L, Serrani F, Berardinelli E, Mancini S,
[38] Wall ME, Bernacki SH, Loboa EG. Effects of serial passaging on the et al. Human mesenchymal stem cells from chorionic villi and amniotic
adipogenic and osteogenic differentiation potential of adipose-derived fluid are not susceptible to transformation after extensive in vitro
human mesenchymal stem cells. Tissue Eng 2007;13(6):1291e8. expansion. Cell Transplant 2011;20(5):643e54.
Q. Zhao et al. / Journal of Cellular Immunotherapy 2 (2016) 3e20 17

[58] Chen G, Yue A, Ruan Z, Yin Y, Wang RZ, Ren Y, et al. Human um- [76] Krampera M, Glennie S, Dyson J, Scott D, Laylor R, Simpson E, et al.
bilical cord-derived mesenchymal stem cells do not undergo malignant Bone marrow mesenchymal stem cells inhibit the response of naive and
transformation during long-term culturing in serum-free medium. PLoS memory antigen-specific T cells to their cognate peptide. Blood
One 2014;9(6). e98565. 2003;101(9):3722e9.
[59] Feng M, Li Y, Han Q, Bao X, Yang M, Zhu H, et al. Preclinical safety [77] Kim JH, Lee YT, Hong JM, Hwang YI. Suppression of in vitro murine
evaluation of human mesenchymal stem cell transplantation in cere- T cell proliferation by human adipose tissue-derived mesenchymal stem
brum of nonhuman primates. Int J Toxicol 2014;33(5):403e11. cells is dependent mainly on cyclooxygenase-2 expression. Anat Cell
[60] Francois S, Usunier B, Douay L, Benderitter M, Chapel A. Long-term biol 2013;46(4):262e71.
quantitative biodistribution and side effects of human mesenchymal [78] Engela AU, Hoogduijn MJ, Boer K, Litjens NH, Betjes MG, Weimar W,
stem cells (hMSCs) engraftment in NOD/SCID mice following irradi- et al. Human adipose-tissue derived mesenchymal stem cells induce
ation. Stem Cells Int 2014;2014:939275. functional de-novo regulatory T cells with methylated FOXP3 gene
[61] Shi M, Zhang Z, Xu R, Lin H, Fu J, Zou Z, et al. Human mesenchymal DNA. Clin Exp Immunol 2013;173(2):343e54.
stem cell transfusion is safe and improves liver function in acute-on- [79] Chen K, Wang D, Du WT, Han ZB, Ren H, Chi Y, et al. Human um-
chronic liver failure patients. Stem Cells Transl Med bilical cord mesenchymal stem cells hUC-MSCs exert immunosup-
2012;1(10):725e31. pressive activities through a PGE2-dependent mechanism. Clin
[62] Wang LM, Wang LH, Cong XL, Liu GY, Zhou JJ, Bai B, et al. Human Immunol 2010;135(3):448e58.
umbilical cord mesenchymal stem cell therapy for patients with active [80] Zhang W, Ge W, Li C, You S, Liao L, Han Q, et al. Effects of
rheumatoid arthritis: safety and efficacy. Stem Cells Dev mesenchymal stem cells on differentiation, maturation, and function of
2013;22(24):3192e202. human monocyte-derived dendritic cells. Stem Cells Dev
[63] Rodrigo SF, van Ramshorst J, Hoogslag GE, Boden H, Velders MA, 2004;13(3):263e71.
Cannegieter SC, et al. Intramyocardial injection of autologous bone [81] Hegyi B, Kudlik G, Monostori E, Uher F. Activated T-cells and pro-
marrowderived ex vivo expanded mesenchymal stem cells in acute inflammatory cytokines differentially regulate prostaglandin E2 secre-
myocardial infarction patients is feasible and safe up to 5 years of tion by mesenchymal stem cells. Biochem Biophys Res Commun
follow-up. J Cardiovasc Transl Res 2013;6(5):816e25. 2012;419(2):215e20.
[64] Gupta PK, Chullikana A, Parakh R, Desai S, Das A, Gottipamula S. A [82] Krampera M, Cosmi L, Angeli R, Pasini A, Liotta F, Andreini A, et al.
double blind randomized placebo controlled phase I/II study assessing Role for interferon-gamma in the immunomodulatory activity of human
the safety and efficacy of allogeneic bone marrow derived mesenchymal bone marrow mesenchymal stem cells. Stem Cells
stem cell in critical limb ischemia. J Transl Med 2013;11(1). 143. 2006a;24(2):386e98.
[65] Lee JW, Lee SH, Youn YJ, Ahn MS, Kim JY, Yoo BS, et al. A ran- [83] Ling W, Zhang J, Yuan Z, Ren G, Zhang L, Chen X, et al. Mesenchymal
domized, open-label, multicenter trial for the safety and efficacy of stem cells use IDO to regulate immunity in tumor microenvironment.
adult mesenchymal stem cells after acute myocardial infarction. J Cancer Res 2014;74(5):1576e87.
Korean Med Sci 2014;29(1):23e31. [84] Sotiropoulou PA, Perez SA, Gritzapis AD, Baxevanis CN,
[66] Pak J, Chang JJ, Lee JH, Lee SH. Safety reporting on implantation of Papamichail M. Interactions between human mesenchymal stem cells
autologous adipose tissue-derived stem cells with platelet-rich plasma and natural killer cells. Stem Cells 2006;24(1):74e85.
into human articular joints. BMC Musculoskelet Disord 2013;14:337. [85] Yen BL, Yen ML, Hsu PJ, Liu KJ, Wang CJ, Bai CH, et al. Multipotent
[67] Le Blanc K, Tammik L, Sundberg B, Haynesworth SE, Ringden O. human mesenchymal stromal cells mediate expansion of myeloid-
Mesenchymal stem cells inhibit and stimulate mixed lymphocyte cul- derived suppressor cells via hepatocyte growth factor/c-Met and STAT3.
tures and mitogenic responses independently of the major histocom- Stem Cell Rep 2013;1(2):139e51.
patibility complex. Scand J Immunol 2003b;57(1):11e20. [86] Xu CL, Yu PF, Han XY, Du LM, Gan JH, Wang Y, et al. TGF-beta
[68] Tse WT, Pendleton JD, Beyer WM, Egalka MC, Guinan EC. Sup- promotes immune responses in the presence of mesenchymal stem cells.
pression of allogeneic T-cell proliferation by human marrow stromal J Immunol 2014;192(1):103e9.
cells: implications in transplantation. Transplantation [87] Rafei M, Campeau PM, Aguilar-Mahecha A, Buchanan M, Williams P,
2003;75(3):389e97. Birman E, et al. Mesenchymal stromal cells ameliorate experimental
[69] Di Nicola M, Carlo-Stella C, Magni M, Milanesi M, Longoni PD, autoimmune encephalomyelitis by inhibiting CD4 Th17 T cells in a CC
Matteucci P, et al. Human bone marrow stromal cells suppress T- chemokine ligand 2-dependent manner. J Immunol
lymphocyte proliferation induced by cellular or nonspecific mitogenic 2009;182(10):5994e6002.
stimuli. Blood 2002;99(10):3838e43. [88] Shi Y, Hu G, Su J, Li W, Chen Q, Shou P, et al. Mesenchymal stem
[70] Djouad F, Plence P, Bony C, Tropel P, Apparailly F, Sany J, et al. cells: a new strategy for immunosuppression and tissue repair. Cell Res
Immunosuppressive effect of mesenchymal stem cells favors tumor 2010;20(5):510e8.
growth in allogeneic animals. Blood 2003;102(10):3837e44. [89] Uccelli A, Pistoia V, Moretta L. Mesenchymal stem cells: a new
[71] Potian JA, Aviv H, Ponzio NM, Harrison JS, Rameshwar P. Veto-like strategy for immunosuppression? Trends Immunol 2007;28(5):219e26.
activity of mesenchymal stem cells: functional discrimination between [90] Zappia E, Casazza S, Pedemonte E, Benvenuto F, Bonanni I, Gerdoni E,
cellular responses to alloantigens and recall antigens. J Immunol et al. Mesenchymal stem cells ameliorate experimental autoimmune
2003;171(7):3426e34. encephalomyelitis inducing T-cell anergy. Blood 2005;106(5):1755e61.
[72] Bartholomew A, Sturgeon C, Siatskas M, Ferrer K, McIntosh K, Patil S, [91] Schurch CM, Riether C, Ochsenbein AF. Cytotoxic CD8+ T cells
et al. Mesenchymal stem cells suppress lymphocyte proliferation in stimulate hematopoietic progenitors by promoting cytokine release
vitro and prolong skin graft survival in vivo. Exp hematol from bone marrow mesenchymal stromal cells. Cell Stem Cell
2002;30(1):42e8. 2014;14(4):460e72.
[73] Aggarwal S, Pittenger MF. Human mesenchymal stem cells modulate [92] Godfrey DI, Stankovic S, Baxter AG. Raising the NKT cell family. Nat
allogeneic immune cell responses. Blood 2005;105(4):1815e22. Immunol 2010;11(3):197e206.
[74] Maccario R, Podesta M, Moretta A, Cometa A, Comoli P, Montagna D, [93] Spaggiari GM, Capobianco A, Becchetti S, Mingari MC, Moretta L.
et al. Interaction of human mesenchymal stem cells with cells involved Mesenchymal stem cell-natural killer cell interactions: evidence that
in alloantigen-specific immune response favors the differentiation of activated NK cells are capable of killing MSCs, whereas MSCs can
CD4+ T-cell subsets expressing a regulatory/suppressive phenotype. inhibit IL-2-induced NK-cell proliferation. Blood
Haematologica 2005;90(4):516e25. 2006;107(4):1484e90.
[75] Yan Z, Zhuansun Y, Chen R, Li J, Ran P. Immunomodulation of [94] Selmani Z, Naji A, Zidi I, Favier B, Gaiffe E, Obert L, et al. Human
mesenchymal stromal cells on regulatory T cells and its possible leukocyte antigen-G5 secretion by human mesenchymal stem cells is
mechanism. Exp Cell Res 2014;324(1):65e74. required to suppress T lymphocyte and natural killer function and to
18 Q. Zhao et al. / Journal of Cellular Immunotherapy 2 (2016) 3e20

induce CD4+CD25highFOXP3+ regulatory T cells. Stem Cells [114] Cao X, Han ZB, Zhao H, Liu Q. Transplantation of mesenchymal
2008;26(1):212e22. stem cells recruits trophic macrophages to induce pancreatic beta
[95] Prigione I, Benvenuto F, Bocca P, Battistini L, Uccelli A, Pistoia V. cell regeneration in diabetic mice. Int J Biochem Cell Biol
Reciprocal interactions between human mesenchymal stem cells and 2014;53:372e9.
gammadelta T cells or invariant natural killer T cells. Stem Cells [115] Gao S, Mao F, Zhang B, Zhang L, Zhang X, Wang M, et al. Mouse bone
2009;27(3):693e702. marrow-derived mesenchymal stem cells induce macrophage M2 po-
[96] Corcione A, Benvenuto F, Ferretti E, Giunti D, Cappiello V, Cazzanti F, larization through the nuclear factor-kappaB and signal transducer and
et al. Human mesenchymal stem cells modulate B-cell functions. Blood activator of transcription 3 pathways. Exp Bio Med (Maywood)
2006;107(1):367e72. 2014;239(3):366e75.
[97] Uccelli A, Moretta L, Pistoia V. Mesenchymal stem cells in health and [116] Abumaree MH, Al Jumah MA, Kalionis B, Jawdat D, Al Khaldi A,
disease. Nat Rev Immunol 2008;8(9):726e36. Abomaray FM, et al. Human placental mesenchymal stem cells
[98] Lee DS, Yi TG, Lee HJ, Kim SN, Park S, Jeon MS, et al. Mesenchymal (pMSCs) play a role as immune suppressive cells by shifting macro-
stem cells infected with Mycoplasma arginini secrete complement C3 to phage differentiation from inflammatory M1 to anti-inflammatory M2
regulate immunoglobulin production in b lymphocytes. Cell Death Dis macrophages. Stem Cell Rev 2013;9(5):620e41.
2014:5. e1192. [117] Cho DI, Kim MR, Jeong HY, Jeong HC, Jeong MH, Yoon SH, et al.
[99] Yan H, Wu M, Yuan Y, Wang ZZ, Jiang H, Chen T. Priming of Toll-like Mesenchymal stem cells reciprocally regulate the M1/M2 balance in
receptor 4 pathway in mesenchymal stem cells increases expression of mouse bone marrow-derived macrophages. Exp Mol Med 2014:46. e70.
B cell activating factor. Biochem Biophys Res Commun [118] Oh JY, Lee RH, Yu JM, Ko JH, Lee HJ, Ko AY, et al. Intravenous
2014;448(2):212e7. mesenchymal stem cells prevented rejection of allogeneic corneal
[100] Ungerer C, Quade-Lyssy P, Radeke HH, Henschler R, Konigs C, transplants by aborting the early inflammatory response. Mol Ther
Kohl U, et al. Galectin-9 is a suppressor of T and B cells and predicts 2012;20(11):2143e52.
the immune modulatory potential of mesenchymal stromal cell prepa- [119] Bassi EJ, Moraes-Vieira PM, Moreira-Sa CS, Almeida DC, Vieira LM,
rations. Stem Cells Dev 2014;23(7):755e66. Cunha CS, et al. Immune regulatory properties of allogeneic adipose-
[101] Rosado MM, Bernardo ME, Scarsella M, Conforti A, Giorda E, derived mesenchymal stem cells in the treatment of experimental
Biagini S, et al. Inhibition of B-cell proliferation and antibody pro- autoimmune diabetes. Diabetes 2012;61(10):2534e45.
duction by mesenchymal stromal cells is mediated by T cells. Stem [120] Anderson P, Souza-Moreira L, Morell M, Caro M, O'Valle F, Gonzalez-
Cells Dev 2014. Rey E, et al. Adipose-derived mesenchymal stromal cells induce
[102] Ji YR, Yang ZX, Han ZB, Meng L, Liang L, Feng XM, et al. Mesen- immunomodulatory macrophages which protect from experimental
chymal stem cells support proliferation and terminal differentiation of B colitis and sepsis. Gut 2013;62(8):1131e41.
cells. Cell Physiol Biochem 2012;30(6):1526e37. [121] Mathias LJ, Khong SM, Spyroglou L, Payne NL, Siatskas C,
[103] Spaggiari GM, Moretta L. Interactions between mesenchymal stem cells Thorburn AN, et al. Alveolar macrophages are critical for the inhibition
and dendritic cells. Adv Biochem Eng Biotechnol 2013;130:199e208. of allergic asthma by mesenchymal stromal cells. J Immunol
[104] Nauta AJ, Kruisselbrink AB, Lurvink E, Willemze R, Fibbe WE. 2013;191(12):5914e24.
Mesenchymal stem cells inhibit generation and function of both [122] Lee M, Jeong SY, Ha J, Kim M, Jin HJ, Kwon SJ, et al. Low immu-
CD34+-derived and monocyte-derived dendritic cells. J Immunol nogenicity of allogeneic human umbilical cord blood-derived mesen-
2006;177(4). 2080-7. chymal stem cells in vitro and in vivo. Biochem Biophys Res Commun
[105] English K, Barry FP, Mahon BP. Murine mesenchymal stem cells 2014b;446(4):983e9.
suppress dendritic cell migration, maturation and antigen presentation. [123] Zanotti L, Sarukhan A, Dander E, Castor M, Cibella J, Soldani C, et al.
Immunol Lett 2008;115(1):50e8. Encapsulated mesenchymal stem cells for in vivo immunomodulation.
[106] Jiang XX, Zhang Y, Liu B, Zhang SX, Wu Y, Yu XD, et al. Human Leukemia 2013;27(2):500e3.
mesenchymal stem cells inhibit differentiation and function of mono- [124] Gonzalez-Rey E, Anderson P, Gonzalez MA, Rico L, Buscher D,
cyte-derived dendritic cells. Blood 2005;105(10):4120e6. Delgado M. Human adult stem cells derived from adipose tissue protect
[107] Djouad F, Charbonnier LM, Bouffi C, Louis-Plence P, Bony C, against experimental colitis and sepsis. Gut 2009;58(7):929e39.
Apparailly F, et al. Mesenchymal stem cells inhibit the differentiation of [125] Hoogduijn MJ, Roemeling-van Rhijn M, Engela AU, Korevaar SS,
dendritic cells through an interleukin-6-dependent mechanism. Stem Mensah FK, Franquesa M, et al. Mesenchymal stem cells induce an
Cells 2007;25(8). 2025-32. inflammatory response after intravenous infusion. Stem Cells Dev
[108] Spaggiari GM, Abdelrazik H, Becchetti F, Moretta L. MSCs inhibit 2013;22(21):2825e35.
monocyte-derived DC maturation and function by selectively inter- [126] Liu D, Xu J, Liu O, Fan Z, Liu Y, Wang F, et al. Mesenchymal stem
fering with the generation of immature DCs: central role of MSC- cells derived from inflamed periodontal ligaments exhibit impaired
derived prostaglandin E2. Blood 2009;113(26):6576e83. immunomodulation. J Clin Periodontol 2012a;39(12):1174e82.
[109] Chiesa S, Morbelli S, Morando S, Massollo M, Marini C, Bertoni A, [127] Waterman RS, Tomchuck SL, Henkle SL, Betancourt AM. A new
et al. Mesenchymal stem cells impair in vivo T-cell priming by dendritic mesenchymal stem cell (MSC) paradigm: polarization into a pro-in-
cells. Proc Natl Acad Sci USA 2011;108(42):17384e9. flammatory MSC1 or an Immunosuppressive MSC2 phenotype. PloS
[110] Raffaghello L, Bianchi G, Bertolotto M, Montecucco F, Busca A, One 2010;5(4). e10088.
Dallegri F, et al. Human mesenchymal stem cells inhibit neutrophil [128] Liu L, Wang Y, Fan H, Zhao X, Liu D, Hu Y, et al. MicroRNA-181a
apoptosis: a model for neutrophil preservation in the bone marrow regulates local immune balance by inhibiting proliferation and immu-
niche. Stem Cells 2008;26(1):151e62. nosuppressive properties of mesenchymal stem cells. Stem Cells
[111] Cassatella MA, Mosna F, Micheletti A, Lisi V, Tamassia N, Cont C, 2012;30(8):1756e70.
et al. Toll-like receptor-3-activated human mesenchymal stromal cells [129] Xu C, Ren G, Cao G, Chen Q, Shou P, Zheng C, et al. miR-155 reg-
significantly prolong the survival and function of neutrophils. Stem ulates immune modulatory properties of mesenchymal stem cells by
Cells 2011;29(6):1001e11. targeting TAK1-binding protein 2. J Biol Chem 2013;288(16):11074e9.
[112] Maqbool M, Vidyadaran S, George E, Ramasamy R. Human mesen- [130] Ma J, Ning YN, Xu M, Hou Y, Wang N, Hou XY, et al. Thalidomide cor-
chymal stem cells protect neutrophils from serum-deprived cell death. rects impaired mesenchymal stem cell function in inducing tolerogenic
Cell Biol Int 2011;35(12):1247e51. DCs in patients with immune thrombocytopenia. Blood 2013;122(12).
[113] Nemeth K, Leelahavanichkul A, Yuen PS, Mayer B, Parmelee A, Doi K, 2074-82.
et al. Bone marrow stromal cells attenuate sepsis via prostaglandin [131] de Andrade AV, Riewaldt J, Wehner R, Schmitz M, Odendahl M,
E(2)-dependent reprogramming of host macrophages to increase their Bornhauser M, et al. Gamma irradiation preserves immunosuppressive
interleukin-10 production. Nat Med 2009;15(1):42e9. potential and inhibits clonogenic capacity of human bone marrow-
Q. Zhao et al. / Journal of Cellular Immunotherapy 2 (2016) 3e20 19

derived mesenchymal stromal cells. J Cell Mol Med [150] Li XH, Gao CJ, Da WM, Cao YB, Wang ZH, Xu LX, et al. Reduced
2014;18(6):1184e93. intensity conditioning, combined transplantation of haploidentical he-
[132] Zhuang Y, Li D, Fu JQ, Shi Q, Lu YY, Ju XL. Comparison of biological matopoietic stem cells and mesenchymal stem cells in patients with
properties of umbilical cord derived mesenchymal stem cells from early severe aplastic anemia. PLoS One 2014a;(3):9. e89666.
and late passages: immunomodulatory ability is enhanced in aged cells. [151] Wu Y, Cao Y, Li X, Xu L, Wang Z, Liu P, et al. Cotransplantation of
Mol Med Rep 2014;11(1):166e74. haploidentical hematopoietic and umbilical cord mesenchymal stem
[133] Engela AU, Baan CC, Peeters AMA, Weimar W, Hoogdujin MJ. cells for severe aplastic anemia: successful engraftment and mild
Interaction between adipose tissue-derived mesenchymal stem cells and GVHD. Stem Cell Res 2014;12(1):132e8.
regulatory T-cells. Cell Transplant 2013;22(1):41e54. [152] Li JP, Zheng CL, Han ZC. Abnormal immunity and stem/progenitor
[134] Ferrara JMF, Levine JE, Reddy P, Holler E. Graft-versus-host disease. cells in acquired aplastic anemia. Crit Rev Oncol Hematol
Lancet 2009;373(9674):1550e61. 2010;75(2):79e93.
[135] Baron F, Baker JE, Storb R, Gooley TA, Sandmaier BM, Maris MB, [153] Chen X, Armstrong MA, Li G. Mesenchymal stem cells in immuno-
et al. Kinetics of engraftment in patients with hematologic malignancies regulation. Immunol Cell Biol 2006;84(5):413e21.
given allogeneic hematopoietic cell transplantation after non- [154] Guo Z, Zheng C, Chen Z, Gu D, Du W, Ge J, et al. Fetal BM-derived
myeloablative conditioning. Blood 2004;104(8):2254e62. mesenchymal stem cells promote the expansion of human Th17 cells,
[136] Tabbara IA, Zimmerman K, Morgan C, Nahleh Z. Allogeneic he- but inhibit the production of Th1 cells. Eur J Immunol
matopoietic stem cell transplantation: complications and results. Arch 2009;39(10):2840e9.
Intern Med 2002;162(14):1558e66. [155] Bacigalupo A, Valle M, Podesta M, Pitto A, Zocchi E, De Flora A, et al.
[137] Kim SS. Treatment options in steroid-refractory acute graft-versus-host T-cell suppression mediated by mesenchymal stem cells is deficient in
disease following hematopoietic stem cell transplantation. Ann Phar- patients with severe aplastic anemia. Exp Hematol 2005;33(7):819e27.
macother 2007;41(9):1436e44. [156] Chao YH, Peng CT, Harn HJ, Chan CK, Wu KH. Poor potential of
[138] Le Blanc K, Rasmusson I, Sundberg B, Gotherstrom C, Hassan M, proliferation and differentiation in bone marrow mesenchymal stem
Uzunel M, et al. Treatment of severe acute graft-versus-host disease cells derived from children with severe aplastic anemia. Ann Hematol
with third party haploidentical mesenchymal stem cells. Lancet 2010;89(7):715e23.
2004;363(9419):1439e41. [157] Li J, Lu S, Yang S, Xing W, Feng J, Li W, et al. Impaired immuno-
[139] Le Blanc K, Samuelsson H, Gustafsson B, Remberger M, Sundberg B, modulatory ability of bone marrow mesenchymal stem cells on CD4 (+)
Arvidson J, et al. Transplantation of mesenchymal stem cells to enhance T cells in aplastic anemia. Results Immunol 2012a;2:142e7.
engraftment of hematopoietic stem cells. Leukemia [158] Li J, Yang S, Lu S, Zhao H, Feng J, Li W, et al. Differential gene
2007;21(8):1733e8. expression profile associated with the abnormality of bone marrow
[140] Ringden O, Uzunel M, Rasmusson I, Remberger M, Sundberg B, mesenchymal stem cells in aplastic anemia. PLoS One 2012b;7(11).
Lonnies H, et al. Mesenchymal stem cells for treatment of therapy- e47764.
resistant graft-versus-host disease. Transplantation 2006;81(10):1390e7. [159] Xiao Y, Jiang ZJ, Pang Y, Li L, Gao Y, Xiao HW, et al. Efficacy and
[141] Herrmann RP, Sturm MJ. Adult human mesenchymal stromal cells and safety of mesenchymal stromal cell treatment from related donors for
the treatment of graft versus host disease. Stem Cells Cloning; 7: patients with refractory aplastic anemia. Cytotherapy
45e52. 2013;15(7):760e6.
[142] Ball LM, Bernardo ME, Roelofs H, van Tol MJ, Contoli B, [160] Ozdogu H, Yeral M, Boga C, Kozanoglu I. Use of mesenchymal cells to
Zwaginga JJ, et al. Multiple infusions of mesenchymal stromal cells modulate immune suppression and immune reconstruction in a patient
induce sustained remission in children with steroid-refractory, grade III- with aplastic anemia complicated by invasive sino-orbital aspergillosis.
IV acute graft-versus-host disease. Br J Haematol 2013;163(4):501e9. Turk J Haematol 2014;31(2):181e3.
[143] Muroi K, Miyamura K, Ohashi K, Murata M, Eto T, Kobayashi N, et al. [161] Fang B, Li N, Song Y, Li J, Zhao RC, Ma Y. Cotransplantation of hap-
Unrelated allogeneic bone marrow-derived mesenchymal stem cells for loidentical mesenchymal stem cells to enhance engraftment of hemato-
steroid-refractory acute graft-versus-host disease: a phase I/II study. Int poietic stem cells and to reduce the risk of graft failure in two children
J Hematol 2013;98(2):206e13. with severe aplastic anemia. Pediatr Transplant 2009;13(4):499e502.
[144] Ringden O, Erkers T, Nava S, Uzunel M, Iwarsson E, Conrad R, et al. [162] Wang H, Wang Z, Xue M, Liu J, Yan H, Guo Z. Co-transfusion of
Fetal membrane cells for treatment of steroid-refractory acute graft- haplo-identical hematopoietic and mesenchymal stromal cells to treat a
versus-host disease. Stem Cells 2013;31(3):592e601. patient with severe aplastic. Cytotherapy 2012;12(4):563e5.
[145] Lazarus HM, Haynesworth SE, Gerson SL, Rosenthal NS, Caplan AI. [163] Keegan BM, Noseworthy JH. Multiple sclerosis. Ann Rev Med
Ex vivo expansion and subsequent infusion of human bone marrow- 2002;53:285e302.
derived stromal progenitor cells (mesenchymal progenitor cells): im- [164] de Oliveira GL, de Lima KW, Colombini AM, Pinheiro DG,
plications for therapeutic use. Bone Marrow Transplant Panepucci RA, Palma PV, et al. Bone marrow mesenchymal stromal
1995;16(4):557e64. cells isolated from multiple sclerosis patients have distinct gene
[146] Lazarus HM, Koc ON, Devine SM, Curtin P, Maziarz RT, Holland HK, expression profile and decreased suppressive function compared with
et al. Cotransplantation of HLA-identical sibling culture-expanded healthy counterparts. Cell Transplant 2013.
mesenchymal stem cells and hematopoietic stem cells in hematologic [165] Connick P, Kolappan M, Crawley C, Webber DJ, Patani R, Michell AW,
malignancy patients. Biol Blood Marrow Transplant et al. Autologous mesenchymal stem cells for the treatment of sec-
2005;11(5):389e98. ondary progressive multiple sclerosis: an open-label phase 2a proof-of-
[147] Kuzmina LA, Petinati NA, Parovichnikova EN, Lubimova LS, concept study. Lancet Neurol 2012;11(2):150e6.
Gribanova EO, Gaponova TV, et al. Multipotent Mesenchymal Stromal [166] Mohajeri M, Farazmand A. Mohyeddin Bonab M, Nikbin B, Minagar
Cells for the Prophylaxis of Acute Graft-versus-Host Disease-A Phase A. FOXP3 gene expression in multiple sclerosis patients pre- and post
II Study. Stem Cells Int 2012 2012:968213. mesenchymal stem cell therapy. Iran J Allergy Asthma Immunol
[148] Manochantr S, Y UP, Kheolamai P, Rojphisan S, Chayosumrit M, 2011;10(3):155e61.
Tantrawatpan C, et al. Immunosuppressive properties of mesenchymal [167] Hou ZL, Liu Y, Mao XH, Wei CY, Meng MY, Liu YH, et al. Trans-
stromal cells derived from amnion, placenta, Wharton's jelly and um- plantation of umbilical cord and bone marrow-derived mesenchymal
bilical cord. Intern Med J 2013;43(4):430e9. stem cells in a patient with relapsing-remitting multiple sclerosis. Cell
[149] Wu Y, Wang Z, Cao Y, Xu L, Li X, Liu P, et al. Cotransplantation of Adhes Migrat 2013;7(5):404e7.
haploidentical hematopoietic and umbilical cord mesenchymal stem [168] Liang J, Li X, Zhang H, Wang D, Feng X, Wang H, et al. Allogeneic
cells with a myeloablative regimen for refractory/relapsed hematologic mesenchymal stem cells transplantation in patients with refractory RA.
malignancy. Ann Hematol 2013;92(12):1675e84. Clin Rheumatol 2012;31(1):157e61.
20 Q. Zhao et al. / Journal of Cellular Immunotherapy 2 (2016) 3e20

[169] Duijvestein M, Vos AC, Roelofs H, Wildenberg ME, Wendrich BB, [180] Gu Z, Jiang J, Tan W, Xia Y, Cao H, Meng Y, et al. p53/p21 Pathway
Verspaget HW, et al. Autologous bone marrow-derived mesenchymal involved in mediating cellular senescence of bone marrow-derived
stromal cell treatment for refractory luminal Crohn's disease: results of a mesenchymal stem cells from systemic lupus erythematosus patients.
phase I study. Gut 2010;59(12):1662e9. Clin Dev Immunol 2013;2013:134243.
[170] Ciccocioppo R, Bernardo ME, Sgarella A, Maccario R, Avanzini MA, [181] Gu Z, Tan W, Feng G, Meng Y, Shen B, Liu H, et al. Wnt/beta-catenin
Ubezio C, et al. Autologous bone marrow-derived mesenchymal stro- signaling mediates the senescence of bone marrow-mesenchymal stem
mal cells in the treatment of fistulising Crohn's disease. Gut cells from systemic lupus erythematosus patients through the p53/p21
2011;60(6):788e98. pathway. Mol Cell Biochem 2014;387(1-2):27e37.
[171] Cho YB, Lee WY, Park KJ, Kim M, Yoo HW, Yu CS. Autologous [182] Nie Y, Lau C, Lie A, Chan G, Mok M. Defective phenotype of
adipose tissue-derived stem cells for the treatment of Crohn's fistula: a mesenchymal stem cells in patients with systemic lupus erythematosus.
phase I clinical study. Cell Transplant 2013;22(2):279e85. Lupus 2010;19(7):850e9.
[172] de la Portilla F, Alba F, Garcia-Olmo D, Herrerias JM, Gonzalez FX, [183] Alexander T, Thiel A, Rosen O, Massenkeil G, Sattler A, Kohler S,
Galindo A. Expanded allogeneic adipose-derived stem cells (eASCs) et al. Depletion of autoreactive immunologic memory followed by
for the treatment of complex perianal fistula in Crohn's disease: results autologous hematopoietic stem cell transplantation in patients with re-
from a multicenter phase I/IIa clinical trial. Int J Colorectal Dis fractory SLE induces long-term remission through de novo generation
2013;28(3):313e23. of a juvenile and tolerant immune system. Blood 2009;113(1):214e23.
[173] Forbes GM, Sturm MJ, Leong RW, Sparrow MP, Segarajasingam D, [184] Li X, Wang D, Liang J, Zhang H, Sun L. Mesenchymal SCT ameliorates
Cummins AG, et al. A phase 2 study of allogeneic mesenchymal refractory cytopenia in patients with systemic lupus erythematosus.
stromal cells for luminal Crohn's disease refractory to biologic therapy. Bone marrow Transplant 2013;48(4):544e50.
Clin Gastroenterol Hepatol 2014;12(1):64e71. [185] Wang D, Zhang H, Liang J, Li X, Feng X, Wang H, et al. Allogeneic
[174] Hoffman RW. T cells in the pathogenesis of systemic lupus erythema- mesenchymal stem cell transplantation in severe and refractory sys-
tosus. Clin Immunol 2004;113(1):4e13. temic lupus erythematosus: 4 years of experience. Cell Transplant
[175] Sanz I. Connective tissue diseases: targeting B cells in SLE: good news 2013a;22(12):2267e77.
at last! Nat Rev Rheumatol 2011;7(5):255e6. [186] Wang D, Li J, Zhang Y, Zhang M, Chen J, Li X, et al. Umbilical cord
[176] Bernatsky S, Boivin JF, Joseph L, Manzi S, Ginzler E, Gladman DD, mesenchymal stem cell transplantation in active and refractory systemic
et al. Mortality in systemic lupus erythematosus. Arthritis Rheum lupus erythematosus: a multicenter clinical study. Arthritis Res Ther
2006;54(8):2550e7. 2014;16(2). R79.
[177] Tang Y, Ma X, Zhang H, Gu Z, Hou Y, Gilkeson GS, et al. Gene [187] Bernardo ME, Fibbe WE. Mesenchymal stromal cells: sensors and
expression profile reveals abnormalities of multiple signaling pathways switchers of inflammation. Cell Stem Cell 2013;13(4):392e402.
in mesenchymal stem cell derived from patients with systemic lupus [188] Skrahin A, Ahmed RK, Ferrara G, Rane L, Poiret T, Isaikina Y, et al.
erythematosus. Clin Dev Immunol 2012;2012:826182. Autologous mesenchymal stromal cell infusion as adjunct treatment in
[178] Tang Y, Xie H, Chen J, Geng L, Chen H, Li X, et al. Activated patients with multidrug and extensively drug-resistant tuberculosis: an
NF-kappaB in bone marrow mesenchymal stem cells from sys- open-label phase 1 safety trial. Lancet Respir Med 2014;2(2):108e22.
temic lupus erythematosus patients inhibits osteogenic differenti- [189] Liu R, Zhang Z, Lu Z, Borlongan C, Pan J, Chen J, et al. Human
ation through downregulating Smad signaling. Stem Cells Dev umbilical cord stem cells ameliorate experimental autoimmune
2013;22(4):668e78. encephalomyelitis by regulating immunoinflammation and remyelina-
[179] Gu Z, Cao X, Jiang J, Li L, Da Z, Liu H, et al. Upregulation of tion. Stem Cells Dev 2013;22(7):1053e62.
p16INK4A promotes cellular senescence of bone marrow-derived [190] Zhang Z, Fu J, Xu X, Wang S, Xu R, Zhao M, et al. Safety and
mesenchymal stem cells from systemic lupus erythematosus patients. immunological responses to human mesenchymal stem cell therapy in
Cell Signal 2012;24(12):2307e14. difficult-to-treat HIV-1-infected patients. AIDS 2013;27(8):1283e93.

Вам также может понравиться