ArticlePDF AvailableLiterature Review

Mesenchymal Stem/Stromal Cells Derived from Human and Animal Perinatal Tissues—Origins, Characteristics, Signaling Pathways, and Clinical Trials

Authors:

Abstract and Figures

Mesenchymal stem/stromal cells (MSCs) are currently one of the most extensively researched fields due to their promising opportunity for use in regenerative medicine. There are many sources of MSCs, of which cells of perinatal origin appear to be an invaluable pool. Compared to embryonic stem cells, they are devoid of ethical conflicts because they are derived from tissues surrounding the fetus and can be safely recovered from medical waste after delivery. Additionally, perinatal MSCs exhibit better self-renewal and differentiation properties than those derived from adult tissues. It is important to consider the anatomy of perinatal tissues and the general description of MSCs, including their isolation, differentiation, and characterization of different types of perinatal MSCs from both animals and humans (placenta, umbilical cord, amniotic fluid). Ultimately, signaling pathways are essential to consider regarding the clinical applications of MSCs. It is important to consider the origin of these cells, referring to the anatomical structure of the organs of origin, when describing the general and specific characteristics of the different types of MSCs as well as the pathways involved in differentiation.
Content may be subject to copyright.
cells
Review
Mesenchymal Stem/Stromal Cells Derived from Human and
Animal Perinatal Tissues—Origins, Characteristics, Signaling
Pathways, and Clinical Trials
Magdalena Kulus 1, Rafał Sibiak 2,3 , Katarzyna Stefa ´nska 2, Maciej Zdun 4, Maria Wieczorkiewicz 4,
Hanna Piotrowska-Kempisty 4,5, J˛edrzej M. Ja´skowski 6, Dorota Bukowska 6, Kornel Ratajczak 1, Maciej Zabel 7,
Paul Mozdziak 8and Bartosz Kempisty 1,2,8,9,*


Citation: Kulus, M.; Sibiak, R.;
Stefa´nska, K.; Zdun, M.;
Wieczorkiewicz, M.;
Piotrowska-Kempisty, H.; Ja´skowski,
J.M.; Bukowska, D.; Ratajczak, K.;
Zabel, M.; et al. Mesenchymal
Stem/Stromal Cells Derived from
Human and Animal Perinatal
Tissues—Origins, Characteristics,
Signaling Pathways, and Clinical
Trials. Cells 2021,10, 3278. https://
doi.org/10.3390/cells10123278
Academic Editor: Joni H. Ylostalo
Received: 5 October 2021
Accepted: 19 November 2021
Published: 23 November 2021
Publisher’s Note: MDPI stays neutral
with regard to jurisdictional claims in
published maps and institutional affil-
iations.
Copyright: © 2021 by the authors.
Licensee MDPI, Basel, Switzerland.
This article is an open access article
distributed under the terms and
conditions of the Creative Commons
Attribution (CC BY) license (https://
creativecommons.org/licenses/by/
4.0/).
1
Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun,
87-100 Torun, Poland; magdalena.kulus@umk.pl (M.K.); kornel@umk.pl (K.R.)
2Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland;
75094@student.ump.edu.pl (R.S.); k.stefanska94@o2.pl (K.S.)
3Division of Reproduction, Department of Obstetrics, Gynecology, and Gynecologic Oncology,
Poznan University of Medical Sciences, 60-535 Poznan, Poland
4Department of Basic and Preclinical Sciences, Institute of Veterinary Medicine,
Nicolaus Copernicus University in Torun, 87-100 Torun, Poland; maciejzdun@umk.pl (M.Z.);
maria.wieczorkiewicz@umk.pl (M.W.); hpiotrow@ump.edu.pl (H.P.-K.)
5Department of Toxicology, Poznan University of Medical Sciences, 60-631 Poznan, Poland
6Department of Diagnostics and Clinical Sciences, Institute of Veterinary Medicine,
Nicolaus Copernicus University in Torun, 87-100 Torun, Poland; jmjaskowski@umk.pl (J.M.J.);
dbukowska@umk.pl (D.B.)
7Division of Anatomy and Histology, University of Zielona Gora, 65-046 Zielona Gora, Poland;
m.zabel@wlnz.uz.zgora.pl
8Prestage Department of Poultry Science, North Carolina State University, Raleigh, NC 27695, USA;
pemozdzi@ncsu.edu
9Department of Anatomy, Poznan University of Medical Sciences, 60-781 Poznan, Poland
*Correspondence: bkempisty@ump.edu.pl
Abstract:
Mesenchymal stem/stromal cells (MSCs) are currently one of the most extensively re-
searched fields due to their promising opportunity for use in regenerative medicine. There are many
sources of MSCs, of which cells of perinatal origin appear to be an invaluable pool. Compared
to embryonic stem cells, they are devoid of ethical conflicts because they are derived from tissues
surrounding the fetus and can be safely recovered from medical waste after delivery. Additionally,
perinatal MSCs exhibit better self-renewal and differentiation properties than those derived from
adult tissues. It is important to consider the anatomy of perinatal tissues and the general description
of MSCs, including their isolation, differentiation, and characterization of different types of perinatal
MSCs from both animals and humans (placenta, umbilical cord, amniotic fluid). Ultimately, signaling
pathways are essential to consider regarding the clinical applications of MSCs. It is important to
consider the origin of these cells, referring to the anatomical structure of the organs of origin, when
describing the general and specific characteristics of the different types of MSCs as well as the
pathways involved in differentiation.
Keywords: perinatal mesenchymal stem/stromal cells; MSCs differentiation; signaling pathways
1. Human and Animal Perinatal Tissues—Anatomical, Histological, and
Cellular Characteristics
Perinatal tissues are an extremely rich source of various cell types. The placenta is an
organ that is irreplaceable for the development of the fetus, because it is a multicellular
barrier. The placenta is also unique in terms of the origin of the cells that form it, because it
is composed of cells of maternal and fetal origin, that are genetically distinct organisms [
1
].
Cells 2021,10, 3278. https://doi.org/10.3390/cells10123278 https://www.mdpi.com/journal/cells
Cells 2021,10, 3278 2 of 39
The necessary exchange of nutrients, metabolites, and endocrine regulation must take
place simultaneously with the maintenance of immunological tolerance between the two
organisms [
2
]. It replaces some of the inactive organs in the fetus (lungs, liver, endocrine
glands). The placenta is formed after successful fertilization and implantation of the
embryo in the mammalian uterus. There is a distinction between the maternal-uterine part,
which is the endometrium, and the fetal part. The membrane of maternal origin is referred
to as decidua. Decidualization occurs at different rates depending on the type of placenta,
and is stimulated by factors secreted by the blastocyst (histamine, prostaglandins). The
decidua basalis is the part of the endometrium immediately adjacent to the fetal bladder
and is involved in the formation of the chorioallantoic placenta. The decidua capsularis
separates the fetal bladder from the uterine cavity, and the decidua parietalis connects the
decidua basalis and decidua capsularis.
Fetal membranes arise from the zygote, and function as accessory organs. The fetal
membranes consist of the yolk sac (saccus vitellinus), amnion, chorion, and allantois. As the
early stage embryo migrates through the fallopian tube where fertilization occurred, the
cells divide to form a morula (morus) and then a blastocyst. The embryonic node (massa em-
bryonica) is where the embryo proper forms, and the peripherally arranged flattened cells
make up the trophoblast [
3
]. The trophoblast cells have microvilli where the cells of the
tubular epithelium of the endometrium fuse. The trophoblast cells contain proteolytic
enzymes (zinc-containing metalloproteases) that degrade the endometrial epithelium, al-
lowing the blastocyst to penetrate deep into the endometrium [
4
]. Initially, the trophoblast
has two layers: an inner layer (cytotrophoblastus) and an outer layer (syncytiotrophoblastus).
The inner layer consists of highly proliferating mononuclear cells, and the outer layer
is formed by cell fusion and has invasive capacity and is responsible for anchoring the
blastocyst in the uterus [
5
]. It will then develop into the chorion and participate in the
construction of the placenta.
The yolk sac forms as the first fetal membrane. The wall of the yolk sac is trilaminar
(saccus vitellinus trilaminaris) due to the ingrowth of the extraembryonic mesoderm between
the trophoblast and the extraembryonic endoderm. The yolk sac has hematopoietic func-
tions because the first blood cells and blood vessels are formed in it [
6
]. Primary germ
cells (gonocytes) also appear in the wall of the yolk sac, which then migrate to the gonadal
primordia [
7
]. In the further development of the embryo, the yolk sac usually disappears
and the established pedicle, together with the yolk vessels and the surrounding mesoderm,
become part of the umbilical cord.
The membrane that directly covers the embryo is the amnion, which is formed around
day 7 of embryonic development. The space between the amnion and the embryo, the
amniotic cavity, is filled with amniotic fluid (liquor amnioticus) [
8
]. The amnion consists
of ectodermal epithelium and mesenchymatous tissue and is generally not vascularized.
The amniotic epithelium is composed of large, polygonal flat cells, the surfaces of which
are covered with microvilli [
9
]. These cells may exfoliate into the amniotic fluid. Prenatal
diagnosis utilizes exfoliated epithelium by collecting fluid through a puncture of the
amniotic cavity (amniocentesis) [
10
]. Between individual amniotic epithelial cells, on
their lateral surfaces, intercellular spaces are formed. They are filled with microvilli
and protuberances, named amniotic water vacuoles because of their appearance. These
cells contain numerous lipid droplets and glycogen grains. The basement membrane of
the amniotic epithelium contains numerous reticular fibers, passing into mesenchymatic
tissue, which is rich in fibroblastic cells and collagen fibers [
11
]. This tissue shows great
strength. Amniotic fluid is produced by epithelial cells, and in addition serous fluid is
infiltrated from the mesenchyme through the intercellular spaces. The amount of fluid
changes during pregnancy, and the main components are water (99%), saccharides, proteins,
urea, and also exfoliated cells or fetal downy hairs (lanugo). Amniotic fluid is constantly
and rapidly exchanged. Resorption occurs by the amniotic epithelial cells and by the
fetus [
12
]. Amniotic fluid has important functions in providing the embryo with a watery
environment, protection from injury, amortization, and metabolism.
Cells 2021,10, 3278 3 of 39
The chorion lies in direct contact between the amnion and the endometrium, forming
an integral part of the placenta. It arises from the trophoblast, which merges with the
extraembryonic mesoderm [
13
]. The surface of the chorion is covered by characteristic
villi, which come into close physical contact with the uterine endometrium [
14
]. The shape
of these villi varies depending on the type of implantation and placenta, and the animal
species. The chorion does not produce blood vessels and vascularization comes from
the allantois or yolk sac. The chorion over a large area fuses with the allantois to form
the chorioallantois [15].
The last fetal membrane to form is the allantois, which is of endo- and mesodermal
origin. It arises from the posterior part of the primary intestine. Already at an early stage,
hematopoietic islands and blood vessels form in the wall of the allantois, which make up
the initial formation of the umbilical artery and vein [
16
]. The intraembryonic part of the
allantois merges with the bladder valve and then disappears to form the urachus and then
the median umbilical ligament [
17
]. The main role of the allantois is to supply blood vessels
to the chorion, forming the placental circulation. In some animals, the allantois also has
a role related to excretion of metabolic products, in which case it is well developed and
large in size [
18
]. In humans and rodents, it has a residual form as the diverticulum and
caulis allantoicus.
The umbilical cord (funiculus umbilicalis) extends from the ventral wall of the embryo
and connects the embryo to the placenta. It includes the yolk and umbilical blood vessels,
the yolk duct, and the caulis allantoicus, surrounded by the dermal cord. The yolk duct
quickly overgrows and becomes a solid string of cells with yolk vessels. The allantois duct
is lined with a flat monolayer of epithelium [
19
]. Anastomoses may be formed between
the vessels and the course of the vessels forms a spiral, providing great flexibility [
20
]. The
mesoderm of all the ducts running in the umbilical cord fuses together and develops into a
mucous connective tissue (tela mucoidea connectens), otherwise known as Wharton’s jelly.
It contains abundant intercellular substance, rich in glycosaminoglycans, collagen, elastic
and reticular fibers, and fibroblasts. Externally, the umbilical cord is surrounded by a thin
monolayer of epithelium (periderm) of ectodermal origin [
21
]. The umbilical cord may
contain nerves that receive sensory stimuli related to tissue tension [
22
]. The cord varies
in strength depending on the species, but is easily broken during birth. Bleeding from
the cord vessels is not abundant due to contraction of the strong arterial muscle layer and
rupture of the venous connection to the placenta.
Among mammals, a wide range of placental strategies can be observed based upon
the different gestational and environmental needs of the fetus [
23
]. Mammalian placentas
are mostly classified into two types: yolk sac placenta and chorioallantoic placenta. The
yolk sac placenta is a trilaminar yolk sac attached to the uterine tissue, which usually plays
a role during the early post-implantation period. In most mammals, with the exception
of rodents and rabbits, the yolk sac placenta becomes reduced after the first trimester of
pregnancy [
24
]. Thus, impaired structural and functional development of the yolk sac
contributes to embryo/fetal toxicity and teratogenicity in rats [
25
]. The chorioallantoic
placenta is formed from the endometrium of the mother and the trophectoderm of the
embryo, and is the principal placenta in mammals during middle to late gestation [24].
There are two main classifications of chorioallantoic placentas. The first, based on the
distribution of villi over the surface of the chorion, divides placentas into diffuse, multi-
cotyledonary, zonary, and discoid. In a diffuse type, such as those of pigs [
26
], camel [
27
],
lemurs, and lorises [
28
], the surface is covered with villi that interdigitate with crypts in the
uterine epithelium. The villi may aggregate into bundles, forming microcotyledons, such
as those observed in horses. In turn, most ruminants produce cotyledonary placentas. Each
cotyledon is a small disk, with their numbers varying from a few in deer (oligocotyledonary)
to many in bovids (polycotyledonary) [
29
]. Furthermore, the zonary placenta is typical for
carnivores, forming a belt around the chorionic sac [
24
]. Finally, the discoid placenta is
characterized by a roughly circular area. This type of placenta is found in most primates,
including humans, as well as in rodents and rabbits [24].
Cells 2021,10, 3278 4 of 39
The second classification is based on the number of tissues separating maternal and
fetal blood. In the hemochorial type of placenta, the trophoblast invades the uterine
epithelium, stroma, and maternal arterial walls to come into direct contact with maternal
blood [
23
]. There are hemomonochorial (higher primates, e.g., human; hystricomorph
rodents, e.g., guinea pig), hemodichorial (rabbits), and hemotrichorial (most myomorph
rodents, such as rats and mice) placentas, with one, two, and three trophoblast layers
in the interhaemal barrier, respectively [
24
,
28
]. Furthermore, there is a clear difference
between higher primates and lemurs or lorises (lower primates), with the former (similarly
to pigs) producing the epitheliochorial type of placenta. This is the most superficial type of
placenta, lacking significant invasion of the uterine lining [
26
,
28
]. Apart from these two
types, i.e., hemochorial and epitheliochorial, there is also the endotheliochorial type. In this
case, the maternal uterine epithelium and connective tissue disappear after implantation,
and the trophoblasts come into direct contact with the maternal endometrium. This type
occurs in all four major clades of eutherian mammals (Euarchontoglires, Laurasiatheria,
Xenarthra, and Afrotheria), including carnivores [24].
2. Mesenchymal Stem/Stromal Cells—Origin, Cellular and Molecular Characteristics,
and Signaling Pathways Involved in Differentiation
Cells with stem-like potential have been of continued interest to researchers in various
fields for many years. Division and classification are still being attempted, and molecular
characterization appears to be the most appropriate. Enabling mesenchymal stem/stromal
cells (MSCs) cells to be used in regenerative medicine, especially for musculoskeletal
diseases, degenerative diseases, or incurable conditions, is very promising. MSCs also
promote immunomodulation as they can both inhibit and stimulate the immune system and
express many immunosuppressors [
30
,
31
] and influence autophagy processes [
32
]. They
also exhibit anti-apoptotic [
33
] and antioxidant [
34
] effects, which promote the treatment
of neuromuscular soreness [
35
]. In recent years, there has been an abundance of progress
in research related to the isolation and culture of multipotent stem cells derived from
various human and animal tissues. The most commonly used sources of MSCs are: bone
marrow, adipose tissue, cord blood, peripheral blood, muscle tissue, placenta, and amniotic
fluid. According to the most general definition of MSCs, they need to exhibit the ability to
adhere to plastic surfaces; express specific differentiation clusters, such as CD73, CD90, and
CD105; and have the ability to differentiate into osteogenic, chondrogenic, or adipogenic
lineage cells
in vitro
. Unlike cells of the hematopoietic lineage, they do not express CD14,
CD34, CD45, and HLA-DR [
36
]. Additionally, MSCs can express other markers such as
nestin (Tuj-1) for neural cells [
37
], smooth muscle
α
-actin, smooth muscle myosin heavy
chain for muscle cells [
38
], or transforming growth factor-
β
(TGF-beta) receptor [
39
] and
integrins [
40
]. It should be emphasized that cells expressing neural specific markers in
in vitro
cultures often present only the transient neuron-like morphology. Inducing full
neuronal functionality remains an elusive goal. Cultured cells fail to generate functional
polarity and form new signals passing neuronal synapses [
41
]. Undoubtedly, significant
work remains to understand the biology of MSCs.
Initially, the discovery and identification of pluripotent embryonic stem cells (ESCs) [
42
]
revealed the existence of cells that can self-renew indefinitely and differentiate into all three
embryonic germ layers [
43
,
44
], revealing a wide field of applications. Ethical issues have
limited the availability of new ES cells lines [
45
]. An alternative source of stem cells has
proven to be adult tissues, which contain a certain pool of multipotent cells. The use of
adult stem cells is devoid of ethical considerations, they are widely available, and there
is a lower risk of tumorigenesis [
46
,
47
]. Hematopoietic stem cells (HSCs), which have
the ability to differentiate into all lineages of the blood and immune system, have been
distinguished. HSCs have found applications in the treatment of blood disorders and
leukemia [48].
A promising source consists of MSCs isolated from adult tissues, which
in vitro
differentiated into adipogenic [
49
], chondrogenic [
50
], osteogenic [
51
], or even neurogenic
lineages [
52
]. The cells taken from the patient are multiplied, differentiated, and have
Cells 2021,10, 3278 5 of 39
therapeutic applications. This would involve no need for xenotransplantation, as the
cells would come from the same patient. However, tissue harvesting itself is associated
with rather painful and invasive procedures and the possibility of infection at the harvest
point [
53
]. In addition, the patients themselves who require cell therapy are not in well
enough health to perform the procedure. The clinical condition of donor patients is also
not without significance. It appears that the exhaustion of physiologically occurring MSCs
in patients with primary osteoarthritis occurs. In biomechanical joint damage, subchondral
bone populations of MSCs are not impaired [
54
56
]. It is also important to establish
the ability to proliferate and differentiate, which are key processes in the harvesting of
MSCs. It appears that cells obtained from adult tissues exhibit different potentials [
57
],
and the capacity for proliferation and differentiation decreases during
in vitro
culture
after successive passages [
58
]. Differentiation of cells toward the tissues from which
they originate is overall more successful, as evidenced by the superior mechanisms of
tissue-specific MSCs [
59
]. It was also found that the differentiation capacity, as well as the
self-renewal potential, is dependent on the physiological state of the donor (age, health
status), its genetics, and the influence of environmental conditions [
60
64
]. For example,
the MSCs obtained from young rats presented faster growth correlating with the levels of
proliferating cell nuclear antigen and higher glucose utilization compared to older ones [
65
].
The impact of telomere erosion in MSCs was not insignificant here either [
66
]. MSCs can
be derived from perinatal tissues, which include the placenta [
67
], umbilical cord [
68
70
],
or the cord blood itself [
71
]. In addition, fetal tissue [
72
] and the surrounding amniotic
fluid [
73
] are promising sources for stem cell derivation. In principle, stem cells derived
from perinatal and fetal tissues should have a greater potential for self-renewal and the
ability to proliferate and differentiate; however, many sources have reported that they
show considerable diversity [
53
,
74
], the details of which are given in the following sections
describing the characteristics of individual perinatal MSCs.
Many authors have suggested that an attempt should be made to rename the MSC
because the term “mesenchymal stem cells” has been overused by groups commercializing
administration of “MSCs” for therapeutic purposes. However, the commercial entities do
not perform differentiation of the “MSCs”, and the therapeutic effect of the administered
compound is based mainly on local action and secretion of active factors [
75
]. Indeed,
traumatically altered tissues call induce repair mechanisms. MSCs respond to these signals,
migrate [
76
], and act by secreting active factors (immunomodulatory, trophic, regener-
ative) [
77
,
78
]. This translates into a local therapeutic effect that is, in fact, based on the
specific stem cells of a given patient that have been activated by the exogenous MSCs
administered [
79
,
80
]. Therefore, the first proposal for a new name for these cells in general
is “medicinal signaling cells”, which reduces the misleading of patients that they are re-
ceiving typical stem cells that will produce new tissue [
81
]. There has also been a proposal
(from the International Society for Cellular Therapy (ISCT)) that fibroblast-like cells, which
are plastic-adherent, should be called multipotent mesenchymal stromal cells regardless
of origin [
82
]. Attempts to undertake a uniform classification and nomenclature change
related to MSCs have been ongoing for several years. The acronym “MSCs” is proposed to
remain in the nomenclature, but the authors of individual studies should carefully specify
the origin and demonstrate the functional properties of MSCs [
83
]. However, it is important
to consider that stem/stromal cells remain the most widely used terms to describe MSCs.
Nevertheless, the wide variation in differentiation potential, the lack of standardized acqui-
sition procedures, and the absence of a single universal marker significantly limit standard
clinical application. A major step is the collection itself, the isolation of MSCs from a donor,
which influences the final cell population [
84
]. Most often, these are simple procedures in-
volving mechanical fragmentation of the harvested tissue or enzymatic digestion, followed
by seeding and attachment of the cell suspension into culture dishes. The explant-derived
method involves breaking the tissue fragments into small pieces to facilitate diffusion of
nutrients and gases, and placing these fragments in the culture medium. MSCs proliferate
and colonize on the surface of the bottom of the dish. Enzyme-mediated isolation involves
Cells 2021,10, 3278 6 of 39
incubation and enzymatic digestion of the harvested tissue to release individual cells from
the extracellular matrix (ECM), followed by centrifugation and placement of the resulting
cell pellet in the culture medium. Enzymatic isolations are considered to be more efficient,
but the explant method provides higher homology with better proliferation and viability
rates [
85
,
86
]. It is suggested that is the superior properties of the explant-derived cells are
associated with less stress on the cells [87].
The therapeutic effects of MSCs are related to the secretion of paracrine factors, in-
cluding cytokines, growth factors, mRNAs, miRNAs, and signaling lipids. Much recent
research has focused on nanoparticles derived from the cell membrane of MSCs, called
exosomes [
88
,
89
]. These vesicles are secreted outside the cell, mediating cell-to-cell com-
munication. MSC-derived exosomes are a tantalizing possibility for innovative cell-free
therapies, which could have advantages over whole-cell therapies. Extracellular vesicles
(EVs) can occur as exosomes released extracellularly by fusing with the cell membrane. EVs
are also microvesicles that bud from the plasma membrane, and sometimes occur as apop-
totic bodies of varying sizes [
90
]. In relation to secretory capacity, therapeutic properties are
associated with immunomodulatory and trophic effects at the site of injury/disease [
91
],
and EVs are able to penetrate the blood–brain barrier [92].
With recent advances in the study of exosome-based therapies with MSCs, it is impor-
tant to assess their localization, tracking, and monitoring after transplantation
in vivo
[
93
].
One method is magnetic resonance imaging (MRI), which allows the localization of ex-
osomes labeled with contrast agents (such as ultra-small superparamagnetic iron oxide
nanoparticles) [94].
2.1. Different Origins of MSCs—Sources, Cell Characteristics, Possible Applications
MSCs can be isolated from any vascularized tissue, and studies have indicated that
pericytes (perivascular cells) may be the source, as the gene expression profile of adipose
tissue pericytes is remarkably similar to that of adipose tissue stem cells [
95
,
96
]. Bone
marrow is rich in stem cells that show potential to differentiate into osteoblasts. Initiating
the differentiation process through the release of transforming growth factor β1 (TGF-β1)
leads to the development of osteocytes [
97
]. Research on the clinical application of various
procedures using MSCs in bone regeneration has been ongoing and advanced for several
years [
98
100
]. Supplementation of growth factors to promote bone repair by MSCs is also
currently being investigated, particularly in relation to tissue-engineering scaffolds [
101
].
MSCs also provide opportunities for the treatment of intervertebral disc degeneration.
Although they exhibit high regenerative potential, chondrogenic differentiation, and anti-
inflammatory effects, the specific nature of the intervertebral disc structure (including lack
of blood vessels, low pH and glucose levels, and hypoxia) largely limits the application of
this therapy [102].
Stem cells derived from adipose tissue have great potential for treating orthopedic
conditions [
103
,
104
]. Adipose-derived stem cells (ASCs) have been shown to have multidi-
rectional differentiation properties, including into adipocytes, osteocytes, and chondrocytes,
and they have been shown to express MSCs markers (CD 29, CD44, CD90) with a neg-
ative expression of hematopoietic markers (CD31, CD34, CD45). The demonstration of
cartilage repair in an animal model was also significant [
105
]. MSCs can also be isolated
from oral tissues such as dental pulp, gingiva, dental follicles, alveolar ligaments, and
others. These cells are used in tissue engineering as they show potential for multidirec-
tional differentiation and are easy to obtain and show regenerative potential [
106
]. MSCs
derived from dental tissues, in addition to their ability to differentiate into the mesodermal
lineage, have been shown to undergo ectodermal-neurocyte and endodermal-hepatocyte
differentiation [
107
]. MSCs also show remarkable potential in regenerative processes and
wound healing. MSCs extracted from the basal layer of the epidermis and hair follicles
have been shown to promote skin healing, new blood vessel formation, and endothelial
transformation. The mechanisms of these processes are not entirely clear [
108
]. MSCs
in the treatment of cutaneous wounds inhibit inflammation, promote angiogenesis, and
Cells 2021,10, 3278 7 of 39
accelerate wound closure, and their action is based on paracrine mechanisms [
109
]. MSCs
have also been shown to affect extracellular matrix remodeling [
110
,
111
]. Standard sources
of MSCs (adipose tissue or bone marrow), as well as perinatal tissues, are effective sources
to treat wounds [112,113].
Recently, there has also been research into the possibility of using MSCs from different
sources to treat female infertility of various backgrounds [
114
]. Additionally, numerous
literature data have indicated a high stemness potential of human [
115
], as well as ani-
mal [
116
], ovarian granulosa cells (GCs). GCs, co-forming the ovarian follicle, are classified
as mulipotent cells [
117
,
118
] and play a key role in oocyte maturation through their regular
contact. Their ability to differentiate into osteoblasts, chondroblasts [
119
], mioblasts [
120
],
or even cells from the neurogenic lineage [
121
] has been experimentally demonstrated.
However, a recent study showed that although there was a comparatively high expression
of stemness markers of cells isolated from ovarian follicles, the osteogenic and adipogenic
differentiation capacity in elderly patients was inferior to young ones [122].
Due to the many limitations in the use of adult MSCs, perinatal MSCs could be appli-
cable. They show higher plasticity and proliferation capacity than adult MSCs [
123
]. They
also differ from embryonic stem cells (ESCs) in that they express pluripotent markers and
have active telomerase, but at a much lower level than ESCs [
123
]. As a result, their use is
not associated with the risk of tumorigenesis, as in the case of ESCs [
124
,
125
]. Immunolog-
ical aspects of the transplanted cells are also important [
126
]. Perinatal MSCs should be
immunologically neutral due to the absence of intracellular HLA class II and poor expres-
sion of HLA class I compared to adult MSCs [
127
]. Perinatal cells are also distinguished
from adult MSCs by their greater capacity for osteogenesis
in vitro
, as well as
in vivo
[
128
],
exhibiting greater osteogenic potency [
129
]. Once perinatal MSCs are obtained, they are
centrifuged and cultured in a medium containing serum. Upon adhesion to the plastic
substrate, they form colonies and the cells assume a spindle shape, resembling fibroblasts.
Perinatal MSCs should not express hematopoietic and endothelial markers [
130
]. How-
ever, these are only general definitions of MSCs, which have been verified by subsequent
findings in the stemness field. Simply showing expression of molecular markers should
not determine that the cells in concern are pluripotent. Proteins, meaning products rather
than the genes themselves, are actually responsible for the mechanisms and abilities of
pluripotency [
131
]. For example, it is the amount of Oct-4 protein that determines the main-
tenance of pluripotency and also the direction of cell differentiation [
132
,
133
]. Therefore,
phenotypic characteristics, as well as the degree and efficiency of differentiation of the cells
studied, should be used to demonstrate their pluripotent abilities.
2.2. Differentiation of MSCs—Regulatory Factors and Signaling Pathways
Genetic regulation and transcription factors are involved in the differentiation of MSCs.
In addition, the microenvironment itself can promote proliferation and differentiation, in
addition to providing conditions for growth [
134
136
]. Differentiation towards osteogen-
esis, adipogenesis, or chondrogenesis is, in theory, straightforward for MSCs as they are
derived from the same embryonic lineage. However, theory alone does not correlate to the
actual laboratory results. The differentiation abilities are highly dependent on the source of
the starting cells, the primary cell population, and also the direction of differentiation [
137
].
A mixture of dexa-methasone (Dex) isobutyl-methylxanthine (IBMX) and indomethacin
(IM) is used to induce adipogenesis (Figure 1). Confirmation of a properly occurring differ-
entiation process has been provided by Oil Red O staining of neutral triglycerides and lipids
in cells and visualization of lipid droplets [
138
]. Osteogenic differentiation is performed
by stimulation with dexa-methasone (Dex),
β
-glycerophosphate (
β
-GP), and ascorbic acid
phosphate (aP) [
139
,
140
]. Alkaline activity and calcium accumulation were analyzed to
confirm the process. TGF-
β
2 and TGF-
β
1 are used for chondrogenesis [
141
]. Pathways
involving TGF-
β
, PPAR-gamma, Smad3, and SOX9 are involved in differentiation into
the mesodermal lineage [
142
144
], whereas ectodermal differentiation into the neurogenic
lineage involves the Notch-1 pathway and the protein kinase A (PKA) pathway [
145
147
].
Cells 2021,10, 3278 8 of 39
Hydrocortisone, DMSO, BHA, and KCL, as well as bFGF, NT-3,
β
-mercaptoethanol (
β
-ME),
BDNF, and NGF are used to induce differentiation [
148
,
149
]. Evaluation of differentiation
was based on Tuj-1,
γ
-aminobutyric acid (GABA), MAP-2, neurofilament 200, among oth-
ers [
150
]. Signaling pathways involving TGF-
β
, fibroblast growth factor (FGF), and bone
morphogenetic protein (BMP) are involved in endodermal differentiation of MSCs [151].
Figure 1.
Human mesenchymal stem/stromal cells (MSCs) differentiation and signaling pathways. Abbreviations:
aP—ascorbic
acid phosphate;
β
-GP—
β
-glycerophosphate;
β
-ME—
β
-mercaptoethanol; BDNF—brain-derived neurotrophic
factor; bFGF—basic fibroblast growth factor; BHA—butylated hydroxyanisole; BMP—bone morphogenetic protein;
Dex—dexa
-methasone; DMSO—dimethyl sulfoxide; FGF—fibroblast growth factor; IBMX—isobutyl-methylxanthine; IM—
indomethacin; KCL—potassium chloride; NGF—nerve growth factor; Notch1—notch homolog 1;
NT-3—neurotrophin-3
;
PPAR—gamma-peroxisome proliferator-activated receptor gamma; Smad3—mothers against decapentaplegic homolog 3;
SOX9—transcription factor SOX9; TGF-
β
—transforming growth factor-
β
. Created with BioRender.com (accessed on
5 October 2021).
Research continues to refine the procedures involved in the processes of stem cell
differentiation. Transcription factors for differentiation, such as osterix/Sp7 (Osx), runt-
related transcription factor 2 (RUNX2), and Dlx5 are known to play an important role
in osteogenic differentiation [
152
,
153
]. They influence the course of various signaling
pathways that play a role in osteogenic differentiation processes, including WNT [
154
],
Cells 2021,10, 3278 9 of 39
BMP [
155
], and Akt [
156
]. They may act as regulators as they both activate and inhibit
the differentiation of MSCs. Additionally, a recently published study demonstrated the
accelerating effect of using nanocomposites that activate the WNT/
β
-catenin pathway in
osteogenic differentiation of MSCs [157].
It is noteworthy that the signaling pathways and factors involved in cell differentiation
in a specific cell line described above are not obligatory and may proceed in different ways.
For example, the study by Brady et al. [
158
] compared factors that promote chondrogenesis
using human MSCs derived from perinatal and adult bone marrow. TGF
β
3 was shown
to induce SMAD3 phosphorylation in adult BM-MSCs but not in fetal BM-MSCs. It was
also observed that the induction of chondrogenesis in adult BM-MSCs occurred under
the influence of TGF
β
3 but not by BMP2. Conversely, differentiation of fetal BM-MSCs
was induced by BMP2 but not TGF
β
3. Further, fetal BM-MSCs stimulated chondrogenesis
simultaneously when TGF
β
3 and BMP2 were used. It was shown that they produced
tissue with proteoglycan and type II collagen content similar to that produced by adult
BM-MSCs treated with TGFβ3 alone [158].
3. Mesenchymal Stem/Stromal Cells Derived from Human and Animal Perinatal Tissues
MSCs are present in tissues of fetal origin in both animals and humans. Over time, their
isolation from tissues of fetal origin has become feasible using well-described laboratory
protocols. Isolation of stem cells from fetal material does not raise ethical dilemmas due
to the fact that these tissues are treated as medical waste immediately after delivery. As
a result, the material for laboratory analyses is readily available [
159
]. Research material
can be taken from tissues obtained from invasive diagnostic and treatment procedures
throughout the pregnancy, planned terminations, and after the full-term vaginal delivery
or cesarean section. The studies focused on perinatal stem cells are conducted worldwide.
Several countries, such as the U.S. or China, released their specific cellular and gene
therapy guidance and established regenerative medicine products’ regulations [
160
,
161
].
Each European Union member state currently has its own specific regulations targeted
on embryonic stem cells and perinatal tissues research. Perinatal stem cell clinical trials
must be approved by the local bioethical commissions and be conducted in line with the
EU clinical trials registration law [
162
,
163
]. Cells of fetal origin, also known as perinatal
stem cells, are derived from extraembryonic structures, such as the placenta, umbilical
cord, and amniotic fluid [
164
,
165
]. Perinatal tissues are also widely used as a reservoir of
hematopoietic progenitor cells obtained from the umbilical cord blood [
166
]. Furthermore,
it is believed that the perinatal stem cells obtained at the very early stages of pregnancy
(first trimester) possess a higher regenerative potency compared to cells isolated from
full-term or near-term feto-maternal tissues. Several authors reported that they detected
the higher expression of pluripotency genes in the samples collected at the earlier stages
of pregnancy [
167
169
]. However, the others found no differences in their expression
throughout the gestation [170].
Over the recent years, numerous studies have focused on perinatal stem cells appli-
cations in clinical practice and analyzed their characteristics. In most cases, to reach the
appropriate number of cells before the MSCs transplantation, they should be cultured
in vitro
for several passages. It raises the concern about the occurrence of replicative senes-
cence, which could affect the clinical effectiveness of cellular therapies. Importantly, it
has been noted that the perinatal MSCs seem to be less susceptible to senescence, along
with the passaging, in comparison to MSCs derived from other adult tissues—i.e., bone
marrow-derived MSCs. That feature is in favor of their application in more extensive
clinical trials [171173].
The terminology used to describe particular cellular populations is unclear and needs
to be standardized. Most authors have proposed their own nomenclature based on the
origin of the cells. Silini et al. introduced a novel broad definition of perinatal derivatives
(PnD), which includes all birth-associated tissues, the cells they are composed of, and all
the factors secreted by the mentioned cells along with the conditioned media [174].
Cells 2021,10, 3278 10 of 39
3.1. Animal Perinatal Mesenchymal Stem/Stromal Cells
The studies conducted on animals paved the way for a better understanding of
perinatal MSCs, enabling the first clinical trials in humans. Bailo et al. began the era of
perinatal MSCs in modern medicine by isolating and transplanting the human placenta-
derived MSCs to swine and rats [175].
Subsequently, many further studies were undertaken to determine the conditions for
isolation and to characterize the morphology, immunophenotypes, and other properties of
animal perinatal MSCs. Bartholomew et al. confirmed that the collection of umbilical cord
tissue and umbilical cord blood for stem cell isolation is a safe procedure for mare and foal
pairs in the equine model. They did not observe any changes in time to stand and nurse,
nor in hematological parameters in foals or time to pass the placenta for mares [
176
]. It
was found that the equine umbilical cord-derived MSCs are highly proliferative, spindle-
shaped cells with the potential to differentiate to osteogenic, chondrogenic, and adipogenic
derivatives [
177
179
]. Furthermore, their immunophenotype analyses revealed that they
are positive for vimentin, osteonectin, smooth muscle actin, and MHC I and do not express
CD31, CD18, MHC II, and the T-cell co-stimulatory molecule CD86 [177].
Shaw et al. examined the amniotic fluid obtained from pregnant ewes in early ges-
tation. They detected the presence of AFMSCs in all of the nine collected specimens (less
than 20,000 cells in 10 mL of amniotic fluid). Collected cells were cultured and harvested
for up to 20 passages. Cells doubled their count every 36 to 48 h. Analyzed ovine AFMSCs
presented the expression of CD44, CD58, and CD166 and were negative for the hematopoi-
etic markers CD14, CD31, and CD45 [
180
]. Colosimo et al. examined the properties of
in vitro
cultured ovine AFMSCs. The cells were cultured for 12 passages. They reported
that the ovine AFMSCs retained a high proliferation rate up to six passages. However, cells
maintained the prolonged expression of CD29, CD58, and CD166 surface molecules, as
well as Oct-4, TERT, NANOG, and Sox-2 pluripotency markers (up to 12 passages) [
181
]. It
was discovered that the ovine placental cotyledons are the next readily available source of
spindle-shaped, colony-forming, and plastic-adherent ovine perinatal MSCs. Cultured cells
showed the features of chondrogenic and osteogenic differentiation. They expressed the
CD29, CD44, and CD166 surface markers and were negative for hematopoietic progenitor
cell markers [182].
The studies conducted on dogs confirmed that the plastic-adherent perinatal MSCs
could be successfully isolated from the canine placental tissue, umbilical cord, and amniotic
membrane [
183
186
]. All isolated MSCs displayed a fibroblast-like shape in
in vitro
culture
conditions. Saulnier et al. reported that the placenta-derived MSCs exhibited the highest
proliferation rate in comparison to other types of perinatal MSCs. They discovered that all
cellular populations presented the expression of CD29, CD44, CD73, CD90, CD105, and
Sox-2, but did not show the expression of CD34, CD45, MHC II, NANOG, and Oct-4 [
185
].
The average
in vitro
canine perinatal MSC population doubling time ranged from 21–42 h
and depended on the tissue of their origin [
183
]. It is postulated that canine MSCs are
non-tumorigenic, as Borghesi et al. found no tumor formation features in nude mice after
the transplantation of MSCs derived from canine amniotic membranes [
184
]. Finally, in
strictly controlled conditions, all cell populations have the potential to differentiate into
adipocytes, osteo-, and chondroblasts [183185].
3.2. Human Perinatal Mesenchymal Stem/Stromal Cells
Silini et al. have also proposed the systematized nomenclature and classification of
human perinatal tissues and cells. According to their definition, based on their location,
the following human perinatal MSCs could be distinguished: human amniotic membrane
mesenchymal stromal cells (hAMSC), human placental amniotic membrane mesenchymal
stromal cells (hPAMSC), human reflected amniotic membrane mesenchymal stromal cells
(hRAMSC), human chorionic mesenchymal stromal cells (hCMSC), human chorionic plate
mesenchymal stromal cells (hCP-MSC), human chorionic plate mesenchymal stromal
cells derived from blood vessels (hCP-MSC-bv), human chorionic villi mesenchymal
Cells 2021,10, 3278 11 of 39
stromal cells (hCV-MSC), human chorion leave mesenchymal stromal cells (hCL-MSC). The
umbilical cord-derived MSCs: human umbilical cord amniotic mesenchymal stromal cells
(hUC-AMSC), human umbilical cord Wharton’s jelly mesenchymal stromal cells (hUC-
WJ-MSC), human umbilical cord sub-amnion Wharton’s jelly mesenchymal stromal cells
(hUC-saWJ-MSC), and a lineage of human umbilical cord intermediate Wharton’s jelly
mesenchymal stromal cells (hUC-iWJ-MSC). Finally, the population of human amniotic
fluid mesenchymal stromal cells (hAF-MSC), human basal decidua mesenchymal stromal
cells (hBD-MSC), and human parietal decidua mesenchymal stromal cells (hPD-MSC) [
174
]
were also distinguished.
3.2.1. Placenta-Derived Mesenchymal Stem/Stromal Cells
Historically, participants of the First International Workshop on Stem Cells from
Placenta (2007) proposed the first systematic classification of human placental stem cells
based on their origin and characteristics. They distinguished the following placenta-
derived stem cell subpopulations: (1) human amniotic epithelial cells (hAEC), (2) human
amniotic mesenchymal stromal cells (hAMSC), (3) human chorionic mesenchymal stromal
cells (hCMSC), and (4) human chorionic trophoblastic cells (hCTC) [
187
]. Those cellular
subpopulations emerged from the amniotic and chorionic membrane tissue. The same
experts established the minimal criteria for the definitions of hAMSC and hCMCS. Both cell
lineages: (1) have a capacity to adhere to plastic in
in vitro
conditions; (2) form fibroblast
colony-forming units; (3) from
in vitro
passages 2 to 4; are positive for CD90, CD73, and
CD105 antigens and do not express the CD45, CD34, CD14, and HLA-DR surface antigens;
(4) have the potential to differentiate into one or more lineages, including osteogenic,
adipogenic, chondrogenic, and endothelial; and (5) have a fetal origin [
187
]. Subsequent
studies revealed that placenta-derived MSCs could also be isolated from chorionic villi
samples [188191] and maternal decidua basalis [192,193].
In standard collection protocols, the samples of placental tissue are surgically dis-
sected. The chorionic and amniotic membranes are manually separated and minced into
small pieces. Subsequently, cells are isolated from the tissue by the enzymatic digestion
(dispase II and collagenase 2). In the next step, the solution is filtered and transferred
into culture dishes. Finally, the authors treat isolated cells with various basal culture
media supplemented with different fetal bovine serum concentrations (10–20%) and other
supplements, such as epidermal growth factor and antibiotics (Figure 2) [194198].
Figure 2.
Perinatal mesenchymal stem/stromal cells: isolation and preparation techniques. Created
with BioRender.com (accessed on 5 October 2021).
Cells 2021,10, 3278 12 of 39
It has been reported that the hAMSCs have a fibroblast-like cell shape and do not
change their morphology in
in vitro
culture conditions up to five passages. Moreover,
they can be easily distinguished from the hAEC because they are three times larger than
hAEC [
199
]. Tested hAMSC cultures reached the senescence after 5–15 passages
[199201]
.
Their immunophenotype analyses revealed the presence of the following antigens: i.a.
CD29, CD44, CD73, CD90, CD105, CD166, SSEA-3/4, CK18, HCAM-1, and HLA ABC,
and were negative for the CD14, CD34, CD45, TRA-1-60, VCAM-1, PECAM-1, and HLA-
DR—shown in Table 1[
199
201
]. Moreover, they exhibited the expression of the Oct-3/4,
GATA-4
, Rex-1, BMP-4, SCF, NCAM, nestin, HFN-4alpha, CK18, and vimentin genes,
but did not show the expression of BMP2, FGF-5, Pax-6, and telomerase reverse tran-
scriptase [
199
,
200
]. To date, no significant differences between the hAMSC obtained from
different regions of amniotic membranes have been found [174].
Table 1. Expression of cellular markers in various populations of perinatal mesenchymal stem/stromal cells (MSCs).
Cells Positive Expression (+) Negative Expression () References
Amniotic MSCs
CD29, CD44, CD73, CD90, CD105, CD166,
SSEA-3/4, CK18, HCAM-1, HLA ABC,
Oct-3/4, GATA-4, Rex-1, BMP-4, SCF,
NCAM, nestin, HFN-4alpha,
CK18, vimentin
CD14, CD34, CD45, TRA-1-60,
VCAM-1, PECAM-1,
HLA-DR, BMP2, FGF-5, Pax-6,
TERT
[199201]
Chorionic membrane MSCs CD13, CD29, CD44, CD54, CD73,
CD105, CD166
CD3, CD14, CD34,
CD45, CD31 [202]
Chorionic villi MSCs CD44, CD73, CD90, CD105,
HLA-ABC, Sox-2
CD45, CD34, CD19, HLA-DR,
CD14, CD40, CD56, CD80,
CD83, CD86, CD275
[36,202]
Chorionic plate MSCs CD44, CD73, CD90, CD105, CD166, Oct-4,
NANOG, Sox-2
CD14, CD19, CD34, CD45,
HLA-DR [203205]
Decidua MSCs CD44, CD90, CD105, CD146, CD166,
HLA-ABC
CD40, CD80, CD83, CD86,
HLA-DR [193]
Umbilical cord MSCs
CD13, CD29, CD44, CD73, CD90, CD105,
C10, CD49b-e, CD146, CD166, HLA-ABC,
NANOG, Rex-1, Sox-2
Uncertain expression:
Oct-3/4, SSEA-3, SSEA-4,
STRO-1, TRA-1-60, TRA-1-81
CD14, CD31, CD34, CD45,
CD51/61, CD64, CD106,
HLA-DR
[206211]
Amniotic fluid MSCs CD73, CD90, CD105, Oct-4 CD45, CD34, CD31 [212]
Amniotic fluid stromal cells
CD29, CD44, CD73, CD90, CD105, SSEA-4,
Oct-4, MHC-I, NANOG, SSEA-3,
TRA-1-60, TRA-1-81
MHC-II, CD80, CD86 [167,213]
Human chorionic membrane, chorionic plate, chorionic villi, and chorionic leave
MSCs have been distinguished due to the place of their origin. However, these cellular
populations have similar cellular characteristics and immunophenotypes consistent with
classification criteria established by Parolini et al. [
187
]. Chorionic membrane MSCs present
a fibroblast-like morphology and plastic adherence capacity. The comparative analysis
revealed some morphological differences between the various types of perinatal MSC.
Araujo et al. reported that the chorionic MSCs are smaller than other perinatal MSCs
isolated from amniotic membranes, umbilical cord, or the decidua; however, they exhibited
similar proliferation capacity until passage 8 [
214
]. Chorionic membrane MSCs show the
expression of CD13, CD29, CD44, CD54, CD73, CD105, CD166 surface markers, and the
absence of CD3, CD14, CD34, CD45, and CD31, and are characterized by high cellular
plasticity [
202
]. Chorionic villi MSCs meet the minimal MSC criteria proposed by the
International Society for Cellular Therapy—specifically, they show an expression of CD44,
CD73, CD90, CD105, and HLA-ABC and lack expression of CD45, CD34, CD19, and HLA-
Cells 2021,10, 3278 13 of 39
DR surface molecules [
36
]. Moreover, they are non-immunogenic, meaning they do not
express CD14, CD40, CD56, CD80, CD83, CD86, CD275 immune markers. The expression of
Sox-2 is the only detected pluripotency feature of CV-MSCs [
215
]. Perinatal MSCs isolated
from the chorionic plate are positive for CD44, CD73, CD90, CD105, and CD166; do not
exhibit the expression of CD14, CD19, CD34, CD45, and HLA-DR surface antigens [
203
,
204
];
and express the Oct-4, NANOG, and Sox-2 pluripotent stem cell markers [
205
]. It has been
reported that chorionic plate-derived stem cells possess significantly higher migration and
proliferation properties compared with other perinatal stem cells [203,205].
It is also postulated that tissue samples obtained from different individuals have
specific cellular characteristics. Tai et al. analyzed the differences in properties of placenta-
derived MSCs collected from various areas of the placenta—chorionic plate, amniotic
membrane, and decidual plate—in five patients. They discovered only a moderate hetero-
geneity in osteogenic and adipogenic differentiation potentials in samples obtained from
different placental regions in enrolled individuals. Similar heterogeneity was observed in
the tubulin acetylation measured in different samples [
216
]. Moreover, they discovered that
only the chorionic plate MSCs could decrease the proliferation of peripheral blood mononu-
clear cells (PBMCs) triggered by the phytohemagglutinin. All cell lineages decreased the
proportions of CD3+/CD8-/IFN-
γ
+ Th1 and CD3+/CD8-/IL17+ Th17 cells and elevated
the proportion of Treg in PBMCs [
216
]. Placenta-derived MSCs regulate trophoblast func-
tioning by promoting increased cell survival and protecting mitochondria from the effects
of oxidative stress and, as a result, facilitate trophoblast invasion in humans [217,218].
Decidua MSCs originate from the maternal part of the placenta. They could be
isolated from both decidua basalis and decidua parietalis tissue. It was found that MSCs
isolated from term decidua basalis are capable of differentiating into three mesenchymal cell
lineages [
193
]. Furthermore, Macias et al. reported that decidua-derived MSCs differentiate
into derivatives of all germ layers [
192
]. Decidua MSCs form a monolayer of plastic
adherent, fibroblast-like cells positive for MSC surface markers (CD44, CD90, CD105,
CD146, CD166) and HLA-ABC, and negative for hematopoietic and endothelial markers,
as well as the following molecules—CD40, CD80, CD83, CD86, and HLA-DR [193].
3.2.2. Umbilical Cord-Derived Mesenchymal Stem/Stromal Cells
MSCs could be isolated from both the umbilical cord blood and umbilical cord tis-
sue [
174
]. Yang et al. identified the population of plastic-adherent, fibroblast-like, umbilical
cord blood-derived MSCs in ~25% of primary cultures. The detected cells were positive
for CD13, CD29, CD44, CD73 (SH3, SH4), CD90, and CD105 (SH2) and negative for CD14,
CD31, CD34, CD45, CD51/61, CD64, CD106, and HLA-DR. The isolated cells presented
features of adipogenic, osteogenic, and chondrogenic differentiation [
219
]. Nonetheless,
the low presence of MSCs in the umbilical cord blood makes their isolation and cultures
non-effective for clinical use [219222].
Isolation protocols for MSCs derived from umbilical cord tissue are similar to those
used to isolate chorionic and amniotic membrane MSCs. At first, the umbilical cord tissue—
Wharton’s Jelly—is manually separated from the cord blood vessels and cut into smaller
pieces. Then, the collected samples are directly placed in culture flasks (explant cultures)
or enzymatically digested (collagenase I/II, hyaluronidase, trypsin). In the next step,
the processed solution is filtered or centrifuged and transferred into culture dishes and
cultured in various media at 37
C in a 5% CO
2
atmosphere [
197
,
223
225
]. Fortunately,
the isolation of MSCs from the umbilical cord tissue is much more efficient. Some authors
postulated that the two embryologically different MSCs populations (hUC-AMSC and
hUC-WJ-MSC) are present in the umbilical cord tissue. However, the direct connection
between the amniotic membrane and the inner connective tissue makes it difficult to isolate
the individual cellular subpopulations [
174
,
226
]. Cells isolated from Wharton’s Jelly share
the features of other perinatal MSCs. Interestingly, two morphologically distinct types of
hUC-WJ-MSC (small-sized subpopulation with a flat cell body, large-sized subpopulation)
have been visualized in primary cultures. It was found that the subpopulation of small-
Cells 2021,10, 3278 14 of 39
sized cells displayed higher expression of several surface antigens (CD44, CD73, CD90,
and CD105) [
206
]. Furthermore, those two cellular subpopulations exhibited different
cytoplasmic filament profiles (vimentin and cytokeratin filaments) [
207
]. Other reports
indicate that the umbilical cord-derived MSCs show a mostly fibroblastic morphology
and are positive for other cellular markers, i.e., C10, CD13, CD29, CD49b-e, CD51, CD146,
CD166, and HLA-ABC. In addition, umbilical cord MSCs express the NANOG, Rex-1, and
Sox-2 pluripotency gens. However, the expression of Oct-3/4, SSEA-3, SSEA-4, STRO-1,
Tra-1-60, and Tra-1-81 is uncertain due to the contrary results of previous studies [
208
211
].
Similar to other subpopulations of perinatal MSCs, umbilical cord-derived MSCs possess
high plasticity, and, under specific environmental conditions, could differentiate into all
germ layer derivatives [227231].
3.2.3. Amniotic Fluid-Derived Mesenchymal Stem/Stromal Cells
The amniotic fluid is the next rich source of fetal-origin stem cells collected during
diagnostic amniocentesis, therapeutic amnioreduction, or cesarean section [
232
235
]. Stem
cells suspended in the amniotic fluid form a heterogeneous group of cells with distinct
properties and cellular characteristics. Similar to placental tissues, amniotic fluid is abun-
dant in multipotent amniotic fluid mesenchymal stem cells that shed from the placenta
and umbilical cord. It was shown that these cells are able to differentiate into osteoblas-
tic, bone-forming cells, demonstrated through alkaline phosphatase (ALP) activity and
calcium deposition in the extracellular matrix. The MSCs possess the ability to adhere to
plastic flasks, which makes them possible to isolate from the second and third trimester
amniotic fluid samples in standard culture conditions [
236
]. In addition, differentiation
toward osteoblasts was more efficient on a gelatin scaffold compared to monolayer cul-
ture [
237
]. The experimental observations confirmed the presence of highly proliferative,
colony-forming, spindle-shaped cells in amniotic fluid cultures obtained from full-term
cesarean sections. Immunophenotype analysis revealed that the hAF-MSC did not express
the hematopoietic and endothelial markers (CD45, CD34, CD31), and expressed the MSC
markers (CD73, CD90). The expression of CD105 was detectable, but, significantly lower
than observed in other MSC lineages. The expression of Oct-4 was significantly increased in
the freshly obtained samples in comparison to
in vitro
cultured cells [
212
]. Moraghebi et al.
have also reprogrammed the term hAF-MSC into the pluripotent stem cell with a similar
expression of Oct-4 and NANOG to human embryonic stem cell lines. The reprogrammed
cells had the potential to form teratomas and differentiate into hematopoietic and neural
cell lineages [212].
Amniotic fluid can be used as a source of cells with higher potency, known as the
amniotic fluid stromal cells (AFSCs). AFSCs have the capacity to differentiate into cells of all
three embryonic germ layers without forming tumors. That ability places them somewhere
between ESCs and MSCs. The plastic adherent AFSCs are isolated by the positive selection
for CD117 surface antigen [
232
,
238
]. Cloned, CD117-positive cells express CD29, CD44,
CD73, CD90, CD105, SSEA-4, Oct-4, and MHC-I molecules, and are negative for MHC-II,
CD80, and CD86 antigens. Whereas the first trimester AFSCs could express NANOG,
SSEA-3, TRA-1-60, and TRA-1-81, their expression was not detected in the second trimester
cells [
167
,
213
]. In addition, AFSCs have the potential to be reprogrammed into the induced
pluripotent stem cells [
232
,
239
]. The regenerative properties of amniotic fluid-derived
stem cells mainly depend on the significant paracrine activity of numerous peptides and
cytokines released into the surrounding of damaged tissues [240].
4. Signaling Pathways Involved in Fetal-Derived MSC Development and Differentiation
All trophoblast lineages are derived from the trophoectoderm cells of the blasto-
cyst [
241
]. After the implantation in the uterus, trophoectodermal cells become cytotro-
phoblasts. Human trophoblastic cells can be distinguished into three subpopulations. Both
extravillus cytotrophoblasts and syncytiotrophoblasts are derived from the undifferentiated
cytotrophoblast cells [242]. Trophoblastic cells were demonstrated to differentiate in vitro
Cells 2021,10, 3278 15 of 39
from human embryonic stem cells forming embryoid bodies [
243
]. Bone morphogenetic
protein 4 (BMP4), a member of the transforming growth factor
β
(TGF-
β
) superfamily, in-
duced such differentiation, as indicated by Xu et al. [
244
]. Transcriptomic studies revealed
that BMP4-treated embryonic stem cells exhibited increased expression of trophoblastic
markers, such as CG-
α
, CG-
β
(subunits of human chorionic gonadotropin), placental
growth factor, glial cells missing 1 (GCM1), the non-classical HLA class I molecule HLA-G1,
and CD9. On the contrary, the expression of genes associated with pluripotency, such as
POU domain class 5 transcription factor (POU5F1) or telomerase reverse transcriptase
(TERT), was decreased [244].
Cytotrophoblast proliferation is associated with hypoxic conditions (2% O
2
), as indi-
cated by
in vitro
studies, however, low levels of oxygen do not induce syncytialization [
245
].
Effects exerted by hypoxia are mediated by the hypoxia inducible factor 1 (HIF-1), which
is regulated by the tumor suppressor protein von Hippel-Lindau (VHL) via complex for-
mation. Under normoxic conditions, HIF-1/VHL complex is degraded [
242
]. Apart from
that, hypoxia induces expression of several genes, such as cyclin B1, focal adhesion kinase
(FAK), α5β1 integrin, p53, BAX, TGF-β, or MMP-2 [246].
Syncytialization is a process occurring at implantation when cytotrophoblast cells fuse,
which may be influenced by various factors, as indicated in
in vitro
studies, such as epider-
mal growth factor (EGF), granulocyte-macrophage stimulating factor (GM-CSF), human
chorionic gonadotropin (hCG), glucocorticoids, or estradiol. Syncytin, encoded by an enve-
lope gene of a defective endogenous human retrovirus, HERV-W, induces cell fusion and
syncytiotrophoblast formation, as indicated by
in vitro
studies by Frendo et al. [
247
]. Con-
nexin 43 was demonstrated to participate in syncytiotrophoblast formation as well [248].
Extravillus cytotrophoblasts comprise several subtypes based on their location, such
as cytotrophoblasts of cell columns, interstitial cytotrophoblasts, or endovascular cytotro-
phoblasts. Differentiation towards extravillus cytotrophoblasts occurs along the invasive
pathway, where the cells invade the endometrium [242].
Transcriptomic studies performed by Okae et al. [
249
] revealed that the genes related
to the wingless/integrated (Wnt) and epidermal growth factor (EGF) signaling pathways
were overexpressed in cytotrophoblast cells isolated from first-trimester placentas. This
led to the establishment of proliferative human cytotrophoblast cells in
in vitro
culture
via activation of Wnt and EGF and inhibition of TGF-
β
, histone deacetylase (HDAC),
and Rho-associated protein kinase (ROCK). Such cultured cells were able to give rise to
the three major trophoblastic lineages and, therefore, were designated ‘trophoblast stem
cells’. Their differentiation towards extravillus cytotrophoblast cells was dependent on
the addition of neuregulin 1 (NRG1), A83-01 (an TGF-
β
inhibitor), and Matrigel
®
to the
culture, which resulted in epithelial-mesenchymal transition and expression of HLA-G.
Trophoblast stem cells were also successfully differentiated towards syncytiotrophoblast
and the addition of forskolin (cyclic AMP agonist), EGF, and 3D culture conditions were
vital for this purpose [
249
]. In contrast, the derivation of mouse trophoblast stem cells is
dependent on activation of FGF and TGF-βand inhibition of Wnt and ROCK [250].
The development of the human umbilical cord starts after the implantation of the
blastocyst. Initially, the embryo is connected to endometrium through the trophoblast,
which develops into the connecting stalk, constituting the earliest sign of the umbilical
cord [
22
,
251
]. The umbilical cord’s connective tissue originates from the extraembryonic
mesoblast. Between 28 and 40 days post coitum, the expanding amniotic cavity compresses
the connecting stalk, the allantois, and the yolk sac and covers them with the amniotic
epithelium, forming the cord. Fetal blood vessels originate from the allantois around the
third week post coitum and subsequently develop into umbilical vessels [251].
Although stem cells are located in various compartments of the umbilical cord, the
stromal tissue, called Wharton’s jelly, provides cells that are the richest in stemness proper-
ties [
252
]. Moreover, these cells are mostly located in the proximity of umbilical vessels
in Wharton’s jelly, therefore, it seems that the perivascular region is a source of precursor
cells, Figure 3[253,254].
Cells 2021,10, 3278 16 of 39
Figure 3.
Umbilical cord-derived MSCs—location and characteristics. Created with BioRender.com
(accessed on 5 October 2021).
This hypothesis, related to Wharton’s jelly MSCs, is consistent with the results ob-
tained by Crisan et al. [
255
]. The authors aimed to investigate the presence of multilineage
progenitors among perivascular cells, mostly pericytes, and isolated them from skeletal
muscle, pancreas, adipose tissue, placenta, umbilical cord, and other tissues. These cells
expressed NG2 (neural/glial antigen 2), CD146, and PDGFR
β
(platelet-derived growth
factor receptor
β
), while not expressing endothelial cell markers. When cultured over
a prolonged time period, perivascular cells exhibited expression of markers typical for
MSCs, such as CD44, CD73, CD90, and CD105, and were able to differentiate towards
chondrocytes, adipocytes, and osteocytes, suggesting that MSCs are derived from perivas-
cular cells [
255
]. Sarugaser et al. [
256
] isolated a nonhematopoietic human umbilical cord
perivascular cell population capable of bone nodule formation and expressing markers
typical of MSCs. Interestingly, umbilical cord perivascular cells are also PDGFR
β
+ and may
be recruited through the PDGFB signaling pathway, especially since amniotic fluid has been
demonstrated to contain both PDGFA and PDGFB, which could influence their migration
from the vasculature [
257
,
258
]. Takashima et al. [
259
] demonstrated that the earliest wave
of PDGFR
α
+ MSCs in the embryonic trunk was generated from Sox1+ neuroepithelium
partially through a neural crest pathway. Nevertheless, MSCs may also be isolated from
non-perivascular regions of the umbilical cord, such as the umbilical cord lining, however,
they could have simply migrated away from the vasculature [260].
Moreover, Wharton’s jelly-derived MSCs exhibit properties of both fibroblasts (the expres-
sion of vimentin) and smooth muscle cells (the expression of desmin, actin, and myosin) [
253
]
and, therefore, are regarded as myofibroblasts. As indicated by Nanaev et al. [
21
], the
level of differentiation of stromal cells towards myofibroblasts is dependent on the stage
of gestation, and the most differentiated cells are located in the perivascular zone of
Wharton’s jelly.
The amnion adheres to the umbilical cord and fetal skin and is extended from the edge
of the placenta. This fetal membrane is composed of an epithelial monolayer contacting the
amniotic fluid and four layers of connective tissue of mesodermal origin. The connective
tissue layer is composed of fibroblast-like mesenchymal cells and a collagenous extracellular
matrix. Importantly, the amniotic membrane regulates the volume and composition of
amniotic fluid, which consists of amniotic fluid-derived MSCs [
261
]. However, the cellular
component of the amniotic fluid changes during gestation, receiving cells shed from the
fetus or possibly containing cells derived from the placenta or the inner cell mass of the
Cells 2021,10, 3278 17 of 39
morula [
262
]. It has also been hypothesized that embryonic cell mass releases a variety of
stem cell types into the amniotic cavity, which are transported by the amniotic fluid and
implant various tissues [263].
Torricelli et al. [
264
] obtained small nucleated round cells from the amniotic fluid
before the 12th week of gestation, which were identified as hematopoietic progenitor cells
originating from the yolk sac. This is consistent with the results of Pieternella et al. [
265
],
who demonstrated that amniotic fluid-derived cells are of fetal origin, as indicated by
molecular HLA typing. Moreover, these cells exhibited properties of MSCs, such as
multilineage differentiation potential towards fibroblasts, adipocytes, osteocytes [
265
], and
chondrogenic lineage [
266
]. Ovine mesenchymal amniocytes were shown to give rise to
smooth and skeletal muscle cells after the treatment with promyogenic medium, which
resulted in expression of transgelin, calponin, and α-actin [267].
However, recently, third-trimester amniotic fluid was demonstrated to contain MSCs
of renal origin [
268
]. These cells were not only positive for pluripotency markers, such
as SSEA-4 (stage-specific embryonic antigen 4), c-kit, or TRA-1-60, but also expressed
the master renal progenitor markers: SIX2 (SIX homeobox 2) and CITED1 (CBP/p300-
interacting transactivator 1), as well as renal proteins, including PODXL (podocalyxin
like), LHX1 (LIM homeobox 1), BRN1 (POU class 3 homeobox 3), and PAX8. Moreover,
these cells exhibited renal functions, as demonstrated by albumin endocytosis assays,
and gene ontology terms revealed their involvement in pathways associated with kidney
morphogenesis [268].
Both renal and osteoblastic differentiation was reported to be dependent on mTOR
(mechanistic target of rapamycin) signaling cascade. Importantly, this pathway was demon-
strated to be fully active in amniotic fluid stem cells by Siegel et al. [
269
]. Blocking intercellu-
lar activity of mTOR via the inhibitor rapamycin or through siRNA resulted in diminished
embryonic body formation by amniotic fluid stem cells, which constitutes the principal
step in differentiation of pluripotent embryonic stem cells. Specifically, embryonic body
formation was reported to be dependent on two complexes, namely mTORC1, which regu-
lates mRNA translation via kinase phosphorylation, and mTORC2, which phosphorylates
and subsequently activates AKT [270].
5. Animal Models and Clinical Applications
Human and animal stem cells of fetal origin have been the subjects of numerous
studies that aimed to find their possible applications in daily clinical practice. The first
reports about the unique properties of cells derived from human amniotic membranes were
released almost two decades ago. Balio et al. described a breakthrough in the field of peri-
natal MSC transplantation. They successfully transplanted the human amnion and chorion
cells, obtained from term placentas, to neonatal swine and rats. They discovered that
the obtained cells did not induce allogeneic or xenogeneic lymphocyte proliferation [
175
],
making them viable therapeutic candidates.
5.1. Animal Models
Laboratory animals have been used in medical research for many years. Veterinary
medicine provides a tool to study transplantation mechanisms between basic science and
clinical human medicine. Most human diseases also affect animals, hence the etiopatho-
genesis and treatment are similar. Veterinary medicine, thus, represents a valuable field,
especially in regenerative medicine, where the development of animal-based protocols
could be transferred to human medicine. A key issue is the collaboration of researchers
from different fields, including physicians, veterinarians, biologists, geneticists, and others,
and working together in accordance with the “One Health” mindset [271].
5.1.1. Bone and Cartilage Diseases
With regard to bone repair processes, SCID mice were used that were subcuta-
neously implanted with MSCs from human placental chorion and MSCs from the decidua
Cells 2021,10, 3278 18 of 39
basalis [
272
]. In both cases, ectopic bone formation was observed at 8 weeks. The expression
of the markers osteopontin (OPN), osteocalcin (OCN), biglikan (BGN), and bone sialopro-
tein (BSP), which are characteristic of bone tissue, was also demonstrated. Placenta-derived
MSCs are, therefore, cells with bone-forming potential [
272
]. Similar findings were pre-
sented in a Wistar rat model, where, among MSCs from different sources (bone marrow,
Wharton’s jelly, umbilical cord, placenta, adipose tissue), human placental MSCs showed
the highest osteogenic potential and complete bone regeneration [273].
Human amniotic MSCs were differentiated into chondrocytes, as confirmed by the
expression of SOXs and BMPs, and then transplanted into non-cartilage tissues in mice
and on a collagen scaffold into defects in rat bone. Morphological changes to the trans-
planted MSCs, along with deposition of type II collagen, were observed, suggesting their
potential use in the treatment of osteoarthritis [
274
]. A rabbit model was also used to study
the treatment of cartilage damage, where placental MSCs were applied to a silk fibroin
biomaterial scaffold [
275
]. Damaged femoral condyles lacking articular cartilage were
analyzed after implantation of MSCs. Defect repair occurred within 4–12 weeks and no
more degeneration or inflammatory cell infiltration was observed thereafter. Another study
using a rabbit model for cartilage damage in the knee joint showed similar results [
276
].
MSCs from amniotic fluid were differentiated on chondrogenic medium with TGF
β
3 and
BMP2 and xenotransplantation allowed in vivo survival for 8 weeks [276].
5.1.2. Cardiac Diseases
Due to the fact that adult cardiomyocytes do not regenerate after injury, treatment of
heart failure continues to pose problems. Zhao et al. [
277
] investigated the feasibility of
using human amniotic MSCs to treat heart injury in a rat model. MSCs were cultured with
neonatal rat heart explants and then transplanted into infarcted rat hearts. During culture,
MSCs were stimulated with bFGF and activin A and shown to express Nkx2.5 and atrial
natriuretic peptide (specific for cardiomyocytes). The cardiac-specific myosin alpha heavy
chain gene was also detected. When cultured together with explants, MSCs integrated
and differentiated into the host tissue. After transplantation, MSCs were maintained for
2 months as similar to cardiomyocytes [
277
]. Animal models have also been used to study
the effects of MSCs on myocardial infarction healing [
278
,
279
]. MSCs from Wharton’s
jelly were transplanted into mouse [
279
] and mini-swine [
278
] models. Both studies
demonstrated decreased apoptosis in injured myocardium, cardioprotective effects, and
increased capillary density. Nevertheless, studies in the mini-swine model demonstrated
differentiation of MSCs into cardiomyocytes and endothelial cells. Human placenta-derived
MSCs were also used to study myocardial infarction in a porcine model. MSCs were
preconditioned with hyaluronan mixed with butyric and retinoic acid ester. Implantation
of these MSCs reduced scar size and increased capillary density and myocardial perfusion,
along with a reduction in fibrous tissue [280].
5.1.3. Neurological Disorders
MSCs isolated from Wharton’s jelly have been used to study Parkinson’s disease
using a rat model. The animals were induced to have forebrain lesions, causing movement
disorders. A reduction in motor deficits was observed in rats after MSC administration,
explained by the protection of dopaminergic neurons by growth and neurotrophic factors
secreted by MSCs [
281
,
282
]. Human placenta-derived MSCs were also used in studies
of Parkinson’s disease treatment in a rat model [
283
]. Almost normal motor function
was observed 24 weeks after MSC transplantation. Through immunohistochemical and
positron emission tomography (PET) analyses, dopaminergic differentiation of progenitors
was demonstrated, indicating that neuronal progenitors can differentiate eventually
in vivo
and alleviate motor defects [
283
]. A mouse model of Alzheimer’s disease (AD) was used
to study the effects of MSCs from human placenta [
284
]. These cells were given to mice in-
travenously and the first effects observed were improved spatial learning ability correlated
with fewer A
β
plaques in the brain. There was also a decrease in pro-inflammatory and
Cells 2021,10, 3278 19 of 39
an increase in anti-inflammatory cytokines in mice after MSC administration compared to
animals receiving saline. Improvement of AD pathology through paracrine processes and
immune modulation was indicated [284].
A rat model was also recently used to study the anti-inflammatory effects of Wharton’s
jelly MSCs in spinal cord injury [
285
]. The study used real-time polymerase chain reaction,
Western blotting, and ELISA and determined the expression levels of NLRP1, ASC, active
caspase-1, interleukin-1beta (IL-1
β
) and IL-18, and TNF-
α
. These factors are responsible
for the local inflammatory response. The results indicated decreased expression in rats
with injured spinal cords that received MSC transplantation and, in addition, the motor
functions of the animals were improved [
285
]. Interesting studies have also been conducted
in rodent models for the treatment of spinal cord injury using umbilical cord MSCs. The
spleen was shown to be an important organ mediating the effects of MSCs, through their
immunomodulatory action, stimulating, for example, specific inflammatory cytokines
that recruit immune cells. Splenectomized animals lost the ability to reduce spinal cord
hemorrhage and did not increase systemic IL-10 levels after MSC administration [
286
]. In
contrast, another study, also in a rat model with a damaged cord, compared the effects of
MSCs alone with Wharton’s jelly and conditioned medium [
287
]. Cells in culture exhibit
paracrine activity, hence the potential action of factors contained in conditioned medium
(CM). Rats with compressive lesions were administered MSCs and CM intrathecally. In
both cases, improvement and increased expression of genes related to axonal growth were
observed. However, when MSCs were used, expression of inflammatory markers was
demonstrated, which is a result of the inflammatory response to the transplant. In the
case of CM, no inflammatory response was shown, and, in addition, axonal sprouting was
improved and the number of reactive astrocytes decreased [
287
]. Similarly, treatment of
spinal cord injury in a rat model was studied using Wharton’s jelly MSCs. In the study
described here, MSCs were administered intrathecally at different concentrations and
repetitions. The results showed a positive but dose-dependent effect of MSCs on spinal
cord regeneration [288].
The experimental animals modeling multiple sclerosis were administered human
placenta-derived MSCs (hPMSCs) and low and high doses of extracellular vesicles from
hPMSCs (hPMSCs-EVs), as well as saline. High doses of hPMSCs-EVs resulted in improved
motor function. Both hPMSCs-EVs and hPMSCs reduced DNA damage in oligodendroglia
and also increased myelination in the spinal cord [
289
]. Research on the use of MSCs to
treat multiple sclerosis has also been conducted previously in a mouse model [
290
]. Human
placental MSCs (hPMSCs) were administered intracerebrally to mice with experimental
autoimmune encephalomyelitis in an MS model. Both survival and reduced disease severity
were observed, and the effects were attributed to a reduction in the anti-inflammatory
protein TSG-6 [290].
The rat model has also been used to study MSC therapy in neural tissue ischemia.
MSCs from Wharton’s jelly were transplanted into rats intracerebrally and were shown
to differentiate into glial cells and neuronal cells and also showed increased angiogenesis.
The regulation of beta1-integrin was shown to include an important role in the processes
that promote plasticity of MSCs when transplanted intracerebrally [
150
]. The effect of
human umbilical cord-derived stem cells on the treatment of neonatal hypoxic–ischemic
encephalopathy was also studied in a rat model [
33
]. Motor and cognitive functions were
shown to be improved and caspase-3 and Beclin-2 expression was decreased, suggesting
the potential for MSCs in the treatment of hypoxic–ischemic encephalopathy.
5.1.4. Organ Disorders
The rat and mouse model was also used to study the effect of MSCs from Wharton’s
jelly on liver [
291
] and lung [
292
] fibrosis. In both cases, the amount of collagen was reduced,
which improved organ function, and inflammation was also shown to be reduced. Similar
studies were also conducted later using intravenously administered human placental
MSCs with green fluorescent protein (GFP) expression in the treatment of liver fibrosis
Cells 2021,10, 3278 20 of 39
in a rat model [
293
]. Alleviation of liver fibrosis, reduction of collagen area, reduction
of TGF-
β
1 and
α
-SMA (markers of fibrosis) expression and improvement of rat organ
function were obtained. Liver regeneration by MSCs was examined using a rat model with
carbon-tetrachloride-damaged liver tissue [
294
]. Human MSCs derived from chorionic
platelets were used in this study. The expression of markers related to autophagy, apoptosis,
cell survival, and liver regeneration were analyzed. The results indicated that MSCs
induced tissue repair through HIF-1
α
-mediated mechanisms and autophagy [
294
]. The
effect of human MSCs from amniotic membrane on CCl
4
-induced cirrhosis in mice was
studied [
295
]. The isolated MSCs were injected into the spleens of mice, which were then
sacrificed after 4 weeks. Alanine aminotransferase (ALT) and aspartate aminotransferase
(AST) levels in the blood of mice were evaluated and histological analysis of the liver was
performed. Reduced areas of liver fibrosis and improved blood parameters (ALT, AST)
were observed in mice after MSC injection compared to the control group. Activation
of hepatic stellate cells and apoptosis of hepatocytes were decreased, while regenerative
processes were promoted. The injected MSCs showed expression of hepatocyte-specific
markers (
α
-fetoproteinran and human albumin) [
295
]. Due to the high genetic similarity
of pigs to humans, as well as similar organ size, clinical studies on a porcine model are
particularly valuable. The study by Cao et al. [
296
] used Chinese miniature pigs with
acute liver failure that were implanted with human placental MSCs. Histological analysis
showed a reduction in liver inflammation, liver denaturation, and necrosis and also showed
promotion of liver regeneration; however, these observations applied only to the group of
pigs injected with MSCs via the portal vein. Those that received administration of MSCs
through the jugular vein or treatment of MSCs with X-rays prior to injection and the control
group did not show these changes. The authors suggested that the portal vein infusion
route with the help of B-ultrasound is more favorable compared to the jugular vein [296].
The isolated MSCs from cord blood were used to study colitis in a mouse model
[297,298]
.
The first study used NOD2-activated MSCs from cord blood that were injected intraperi-
toneally. It was shown that MSCs inhibited the inflammatory response and activated the
anti-inflammatory response in the colon [
297
]. In turn, the second study analyzed extracts
from MSCs injected intraperitoneally into mice. It was shown that the extracts strongly
inhibited the inflammatory process and increased the body weight of the animals. A shift
in the functional phenotype of macrophages from M1 to M2 was observed [298].
The mouse model was also used to study the effect of MSCs from human amniotic
fluid on the treatment of stress urinary incontinence [
299
]. MSCs were first differentiated
in vitro
for the myogenic direction, as confirmed by the expression of PAX7, MYOD, and
dystrophin. MSCs were labeled with silica-coated magnetic nanoparticles containing
rhodamine B isothiocyanate and then injected transurethrally into mice with gluteal nerve
injury. Nerve regeneration and neuromuscular junction formation were demonstrated by
expression of neuronal markers and acetylcholine receptor. Transurethral injection of MSCs
resulted in a return to normal histological structure of the urethral sphincter and also a
definite improvement in function, while lacking tumorigenicity and immunogenicity [
299
].
5.1.5. Ischemia and Wound Healing
MSCs isolated from the umbilical cord were tested in a mouse model of hind limb
ischemia. MSCs were isolated and cultured in vitro and then differentiated in endothelial
differentiation medium with VEGF and bFGF. Transplanted cells into the mouse hind limb
differentiated into endothelial cells, indicating the potential use of these cells in promoting
angiogenesis and reendothelialization [230].
Animal models involving mice were used to study wound healing using Wharton’s
jelly MSCs that were placed on a scaffold decellularized from amniotic membrane. Re-
sults showed accelerated wound healing, reduced scarring, and also hair growth on the
treated skin [300].
Cells 2021,10, 3278 21 of 39
5.2. The Application of Perinatal MSC in Human Clinical Trials
The PubMed database was searched for the relevant references from the last five
years until April 2021 to summarize the latest reports on the perinatal MSC application
in the completed clinical trials. We searched the PubMed database using the following
terms: “placenta mesenchymal stem cells”, “umbilical cord mesenchymal stem cells”, and
“amniotic fluid stem cells”. We set the article type to “Clinical Trial” and the species to
“Humans” for additional searching filters.
5.2.1. Placenta-Derived MSCs
Regenerative medicine opens novel perspectives for the clinical application of placenta-
derived MSCs. Soltani et al. analyzed the safety and efficacy of intra-articular injection of
placenta-derived MSCs in knee osteoarthritis treatment. Patients did not present any acute
and long-term severe adverse effects of MSC therapy. The first results were promising—
eight weeks after the procedure, a significant increase in knee flexion range of motion
and pain reduction was noted. Patients also reported a significant improvement in their
daily activity and quality of life. However, they did not observe any significant long-term
clinical improvements and chondral regeneration (24 weeks after the procedure) [
301
]. The
list of the most recent human perinatal MSC clinical trials is shown in Table 2. Winkler
et al. investigated the profile of safety and the potential dose-dependent benefits from the
intramuscular injection of placenta-derived MSCs in the patients who underwent the hip
arthroplasty that was used as a standardized injury model. The MSC administration
was well tolerated and did not cause any adverse effects. Interestingly, the patients
who received the lower dose of placenta-derived MSCs had better post-operative results.
They had significantly improved muscle strength and volume compared with placebo.
Furthermore, the administration of placenta-derived MSCs reduced the levels of surgery-
related inflammatory biomarkers [
302
]. Norgren et al. published the protocol of their
planned clinical trial that aims to assess the safety and the efficiency of placenta-derived
MSC intramuscular injections performed in patients with atherosclerotic critical limb
ischemia who are unsuitable for the standard revascularization [
303
]. Levy et al. reported
that the intrapenile placenta-derived MSC injection increased the penile peak systolic
velocity at 3 and 6 months after the procedure. At 3 months after the procedure, 3 out of
8 patients were able to achieve erection [
304
]. Finally, Zeng et al. described a successful
single-case report of placenta-derived MSC hydrogel application to treat the diabetic
foot ulcer [305].
Immunomodulatory properties of placenta-derived MSCs could be applied to the
treatment of chronic inflammatory diseases. Haller et al. performed the new drug’s
clinical trial for patients with chronic obstructive pulmonary disease. The drug contains
a mixture of anti-inflammatory molecules obtained from the exosomes isolated from
the placenta-derived MSCs. After three (one per week) inhalations with the new drug,
treated patients were found to have significantly increased spirometry parameters (FEV1,
PEF) and improvements in CT pulmonary images [
306
]. Hashemian et al. focused on
the possible implementation of the perinatal MSC transplantations in the treatment of
patients with COVID-19-induced ARDS. They discovered that the cell therapy was safe
and could be associated with reduced dyspnea and increased SpO
2
within 48–96 h after
the intervention [
307
]. Ringden et al., in their non-randomized trial, reported that the
decidua-derived MSCs could be used as a promising new tool for the treatment of severe
acute graft-versus-host disease [308].
5.2.2. Umbilical Cord and Amniotic Fluid-Derived MSCs
Similar to placenta-derived MSCs, umbilical MSCs have found numerous applications
in regenerative medicine. Several clinical trials focused on their utility in the treatment
of osteoarthritis. Patients treated with MSCs via intra-articular injections experienced a
significant decrease in knee pain and improvements in knee joint function and clinical
parameters, such as Western Ontario and Mc Master Arthritis Indexes. The MSC therapy
Cells 2021,10, 3278 22 of 39
was more efficient than the hyaluronic acid injections—its effect was stable over seven
years of follow-up [
309
312
]. He et al. analyzed the efficacy of intramyocardial grafting
of collagen scaffolds covered with umbilical cord-derived MSCs in patients with chronic
ischemic heart disease. They did not observe any significant benefits (reduction of the in-
farct size) from the cellular therapy compared with the control group [
313
]. Interestingly, it
was reported the umbilical cord MSC infusion was connected with the significant improve-
ment in the left ventricular ejection fraction in patients with heart failure [
314
]. Hashemi
et al. noted that the human umbilical cord-derived MSCs seeded on the acellular amniotic
membrane scaffolds improved the process of healing of chronic skin ulcers in patients with
diabetes [
315
]. In addition, the umbilical cord-derived MSC conditioned media could be
used in the novel protocols of regenerative medicine. Kim et al. reported that the topical
drugs with umbilical cord-derived MSC conditioned media improved the skin stratum
corneum and strengthened the skin barrier in patients with atopic dermatitis [
316
]. Finally,
it was found that the application of umbilical cord blood-derived MSC conditioned media
containing cream or serum reduced the total area of microcrusts and erythema in patients
treated with ablative CO2fractional laser [317].
The immunomodulatory properties of the umbilical cord-derived MSCs may success-
fully modify the course of chronic inflammatory and degenerative diseases. Therefore,
several studies have aimed to investigate their feasibility and efficacy in the treatment
of rheumatoid arthritis. MSC therapy was found to be safe and associated with reduced
levels of inflammatory parameters and disease activity [
318
321
]. Other factors, like cervus
and cucumis peptides and interferon-
γ
combined with MSCs, could improve the therapy
effects [
318
,
319
]. Large analysis revealed that the umbilical cord-derived MSC transplanta-
tions are safe and do not cause serious adverse effects in patients with chronic autoimmune
rheumatoid diseases, such as systemic lupus erythematosus, Sjögren’s syndrome, and
systemic sclerosis. However, Deng et al. declared that the MSC therapy has no signif-
icant positive effects in patients with lupus nephritis [
322
]. Riordan et al. reported no
serious adverse effects of MSCs therapy in patients with multiple sclerosis. Assessment
performed one month after the treatment revealed that the treated patients experienced
positive changes in the bladder, bowel, non-dominant hand, and sexual functions, as
well as in the results of walk tests and general quality of life [
323
]. Allogenic umbilical
cord-derived MSCs were also used to treat individuals with cerebral palsy and autism
spectrum disorders. The therapy combined with rehabilitation improved the gross motor
and comprehensive function in children with cerebral palsy, and was associated with
lower levels of inflammatory markers and better clinical outcomes in patients with autism
disorders [324,325].
Moreover, allogeneic umbilical cord-derived MSC infusions were found to be safe
in patients with Crohn’s disease. The randomized controlled trial outcomes revealed
that patients treated with MSCs had significantly reduced disease activity index and
corticosteroid dosage, compared with the controls, at 12 months after the cell therapy [
326
].
Some authors speculated that the unique immunomodulatory properties of umbilical
MSCs could modulate the immune response in allogeneic graft recipients. MSCs were
administered as supplementary induction therapy in patients who underwent kidney
transplantation. The applied procedure was found to be safe for enrolled patients. However,
there was no evidence of its efficacy in comparison with the control group treated with
standard immunosuppressive drugs [
327
]. However Shi et al. discovered that umbilical
cord-derived MSC modifies the course of acute liver allograft rejection. Their analyses
revealed that the MSCs transplantation increased the T-regulatory/T-helper 17 cells ratio,
TGF-β1, and prostaglandin E2 levels [328].
It was discovered that the umbilical cord-derived MSC infusions are safe and well-
tolerated in patients with severe COVID-19 infection. The results of the first clinical trials
indicate that the MSCs therapy could be potentially efficient in the management of COVID-
19 cases. The authors observed several improvements in the treated patients compared
with the placebo—in the CT imaging outcomes and clinical parameters, such as the results
Cells 2021,10, 3278 23 of 39
of 6 min walk tests. However, further large phase III clinical trials are needed to confirm
those mostly positive tendencies [
307
,
329
331
]. He et al. administered a single infusion
of umbilical cord-derived MSCs in patients with severe sepsis. They reported that the
experimental procedure was safe and well-tolerated. Nonetheless, to investigate its efficacy
in that indication, further studies are still needed [332].
Even though the animal models brought us the first promising outcomes, there have
been no studies that assessed the safety and efficacy of amniotic fluid-derived stem cells
in experimental treatment protocols in humans [
232
]. It was demonstrated that after the
appropriate preparation, the population of amniotic fluid-derived MSCs could be used as a
model in the studies focused on human genetic diseases. Squillaro et al. reported that they
silenced glucocerebrosidase (GBA) and alpha-galactosidase A (GLA) genes in the amniotic
fluid-derived MSCs, creating the models of Gaucher and Fabry diseases. They found that
GBA and GLA silencing provoked impaired autophagy and DNA repair mechanisms and,
as a consequence, promoted apoptosis and increased senescence in the investigated cellular
populations. Moreover, the authors concluded that the perinatal cells collected from fetuses
affected by genetic diseases could be used as a readily available source of experimental
materials alternative to animal models [333].
Multiple phase I/II clinical trials confirmed the safety of perinatal MSC administration
in the regenerative medicine protocols and the treatment of various chronic diseases.
However, their efficacy should be investigated in further large randomized controlled trials.
Table 2. Perinatal MSCs in human clinical trials.
Condition/
Procedures Type of Study Number of
Participants Material First Author; Year;
Reference
Knee osteoarthritis
Randomized, double-blind,
placebo-controlled
clinical trial
20 Allogenic
placenta-derived MSCs Soltani; 2019; [301]
Hip arthroplasty
Randomized, double blind,
placebo-controlled,
phase I/IIa
clinical trial
20 Allogenic
placenta-derived MSCs Winkler; 2018; [302]
Erectile dysfunction
Prospective,
non-randomized,
single-arm
clinical trial
8Allogenic
placenta-derived MSCs Levy; 2016; [304]
Chronic obstructive
pulmonary disease
Prospective,
non-randomized,
single-arm
clinical trial
30
Allogenic placenta-derived
MSCs-derived product:
Exo-d-MAPPS drug
Harrell; 2020; [306]
COVID-19-induced
ARDS
Prospective,
non-randomized,
single-arm
clinical trial
11
Allogenic
placenta-derived MSCs
(5 cases)
Umbilical
cord-derived MSCs
(6 cases)
Hashemian; 2021; [307]
Acute graft-versus-host
disease
Prospective,
non-randomized,
single-arm
clinical trial
38 Allogenic decidua-derived
MSCs Ringden; 2018; [308]
Knee osteoarthritis
Single-arm,
open-label
clinical trial
29 Allogenic umbilical
cord-derived MSCs Dilogo; 2020; [309]
Cells 2021,10, 3278 24 of 39
Table 2. Cont.
Condition/
Procedures Type of Study Number of
Participants Material First Author; Year;
Reference
Knee osteoarthritis
Randomized,
placebo-controlled,
phase I/II
clinical trial
26 Allogenic umbilical
cord-derived MSCs Matas; 2019; [310]
Knee osteoarthritis
Open-label,
single-arm,
single-center,
phase I/II clinical trial with
7-year extended follow-up
7
Allogeneic human
umbilical cord
blood-derived MSCs
Park; 2017; [311]
Knee osteoarthritis
Randomized,
placebo-controlled
clinical trial
36 Allogenic umbilical
cord-derived MSCs Wang; 2016; [312]
Chronic ischemic
heart disease
Randomized, double-blind
clinical trial 115
Collagen scaffolds covered
with umbilical
cord-derived MSCs
He; 2020; [313]
Heart failure
Randomized, controlled,
phase I/II
clinical trial
30 Allogenic umbilical
cord-derived MSCs Bartolucci; 2017; [314]
Chronic diabetic
skin ulcers Randomized, clinical trial 5
Allogenic umbilical
cord-derived MSCs seeded
on biological scaffold
Hashemi; 2019; [315]
Atopic dermatitis
Prospective,
non-randomized,
single-arm
clinical trial
28
Topical drugs with
allogenic umbilical
cord-derived MSCs
conditioned media
Kim; 2020; [316]
Ablative CO
2
fractional
laser treatment
Randomized,
double-blinded, controlled
clinical trial
23
Umbilical cord
blood-derived MSCs
conditioned media
containing serum
Kim; 2020; [317]
Rheumatoid arthritis Randomized, controlled
clinical trial 119 Allogenic umbilical
cord-derived MSCs Qi; 2020; [319]
Rheumatoid arthritis
Randomized, controlled,
phase I/II
clinical trial
63 Allogenic umbilical
cord-derived MSCs He; 2020; [318]
Rheumatoid arthritis
Prospective,
phase I/II
clinical trial
64 Allogenic umbilical
cord-derived MSCs Wang; 2019; [320]
Rheumatoid arthritis
Open-label,
single-arm,
single-center,
phase Ia clinical trial
9Allogenic umbilical cord
blood-derived MSCs Park; 2018; [321]
Lupus nephritis
Randomized,
double-blind,
placebo-controlled
clinical trial
18 Allogenic umbilical cord
blood-derived MSCs Deng; 2017; [322]
Cells 2021,10, 3278 25 of 39
Table 2. Cont.
Condition/
Procedures Type of Study Number of
Participants Material First Author; Year;
Reference
Multiple sclerosis
Prospective,
non-randomized,
single-arm
clinical trial
20 Allogenic umbilical
cord-derived MSCs Riordan; 2018; [323]
Cerebral palsy Randomized, controlled
clinical trial 39 Allogenic umbilical
cord-derived MSCs Gu; 2020; [324]
Autism spectrum
disorder
Single-arm,
phase I/II
clinical trial
20 Allogenic umbilical
cord-derived MSCs Riordan; 2019; [325]
Crohn’s disease Randomized, controlled
clinical trial 82 Allogenic umbilical
cord-derived MSCs Zhang; 2018; [326]
Kidney transplantation
Multicenter, randomized,
controlled
clinical trial
42 Allogenic umbilical
cord-derived MSCs Sun; 2018; [327]
Acute liver
allograft rejection
Randomized, controlled,
clinical trial 27 Allogenic umbilical
cord-derived MSCs Shi; 2017; [328]
COVID-19
Randomized, double-blind,
placebo-controlled,
phase II
clinical trial
101 Allogenic umbilical
cord-derived MSCs Shi; 2021; [329]
COVID-19
Parallel assigned
controlled,
non-randomized, phase I
clinical trial
18 Allogenic umbilical
cord-derived MSCs Meng; 2020; [330]
COVID-19
Single-center
open-label, individually
randomized, standard
treatment-controlled
clinical trial
41 Allogenic umbilical
cord-derived MSCs Shu; 2020; [331]
Sepsis
Single-center,
open-label,
dose-escalation phase 1
clinical trial
15 Allogenic umbilical
cord-derived MSCs He; 2018; [332]
6. Conclusions
Due to the indicated properties of differentiation into other cell types, MSCs show great
potential for application in regenerative medicine. Additionally, the simplicity of obtaining
cells from perinatal tissues during routine procedures and minimal ethical concerns make
perinatal tissues one of the most valuable sources for MSCs. Promising results from clinical
trials indicate possible therapeutic use. It seems, therefore, necessary to describe a universal
and simple isolation protocol for the standard use of MSCs in medicine.
Funding: This research received no external funding.
Acknowledgments:
Support in part by the National Institute of Food and Agriculture, United States
Department of Agriculture Animal Health Project NC7082.
Conflicts of Interest: The authors declare no conflict of interest.
Cells 2021,10, 3278 26 of 39
References
1.
Cross, J.; Werb, Z.; Fisher, S. Implantation and the placenta: Key pieces of the development puzzle. Science
1994
,266, 1508–1518.
[CrossRef] [PubMed]
2. Siiteri, P.K.; Stites, D.P. Immunologic and Endocrine Interrelationships in Pregnancy. Biol. Reprod. 1982,26, 1–14. [CrossRef]
3.
Bartel, H. Embriologia, Textbook for Medical Students, 2nd ed.; Medical Publishing House PZWL: Warsaw, Poland, 1999;
ISBN 9788320035933.
4.
Zhang, S.; Mesalam, A.; Joo, M.-D.; Lee, K.-L.; Hwang, J.-Y.; Xu, L.; Song, S.-H.; Koh, P.-O.; Yuan, Y.-G.; Lv, W.; et al. Matrix
metalloproteinases improves trophoblast invasion and pregnancy potential in mice. Theriogenology
2020
,151, 144–150. [CrossRef]
[PubMed]
5.
Pijnenborg, R.; Dixon, G.; Robertson, W.B.; Brosens, I. Trophoblastic invasion of human decidua from 8 to 18 weeks of pregnancy.
Placenta 1980,1, 3–19. [CrossRef]
6.
Palis, J.; McGrath, K.E.; Kingsley, P.D. Initiation of hematopoiesis and vasculogenesis in murine yolk sac explants. Blood
1995
,86,
156–163. [CrossRef] [PubMed]
7.
Ross, C.; Boroviak, T.E. Origin and function of the yolk sac in primate embryogenesis. Nat. Commun.
2020
,11, 3760. [CrossRef]
[PubMed]
8. Hoyes, A.D. Structure and function of the amnion. Obstet. Gynecol. Annu. 1975,4, 1–38. [PubMed]
9. Knezevic, V. Differentiation potential of rat amnion. J. Anat. 1996,189, 1–7. [PubMed]
10. Nizard, J. Amniocentesis: Technique and education. Curr. Opin. Obstet. Gynecol. 2010,22, 152–154. [CrossRef] [PubMed]
11.
Biela´nska-Osuchowska, Z. Embriologia, Textbook for Medical Students, 4th ed.; National Agricultural and Forestry Publishing
House: Warsaw, Poland, 2001; ISBN 8309017480.
12. Modena, A.B.; Fieni, S. Amniotic fluid dynamics. Acta Bio-Med. Atenei Parm. 2004,75, 11–13.
13.
King, B.F. Developmental changes in the fine structure of the chorion laeve (Smooth chorion) of the rhesus monkey placenta.
Anat. Rec. 1981,200, 163–175. [CrossRef]
14.
Watson, E.D.; Cross, J.C. Development of Structures and Transport Functions in the Mouse Placenta. Physiology
2005
,20, 180–193.
[CrossRef] [PubMed]
15.
Minh, H.-N.; Douvin, D.; Smadja, A.; Orcel, L. Fetal membrane morphology and circulation of the liquor amnii. Eur. J. Obstet.
Gynecol. Reprod. Biol. 1980,10, 213–223. [CrossRef]
16.
Downs, K.M.; Temkin, R.; Gifford, S.; McHugh, J. Study of the Murine Allantois by Allantoic Explants. Dev. Biol.
2001
,233,
347–364. [CrossRef] [PubMed]
17.
Spurway, J.; Logan, P.; Pak, S. The development, structure and blood flow within the umbilical cord with particular reference to
the venous system. Australas. J. Ultrasound Med. 2012,15, 97–102. [CrossRef]
18.
Downs, K.M.; Harmann, C. Developmental potency of the murine allantois. Development
1997
,124, 2769–2780. [CrossRef]
[PubMed]
19.
Di Naro, E.; Ghezzi, F.; Raio, L.; Franchi, M.; D’Addario, V. Umbilical cord morphology and pregnancy outcome. Eur. J. Obstet.
Gynecol. Reprod. Biol. 2001,96, 150–157. [CrossRef]
20.
Raio, L.; Ghezzi, F.; di Naro, E.; Franchi, M.; Bruhwiler, H. Prenatal assessment of the Hyrtl anastomosis and evaluation of its
function: Case report. Hum. Reprod. 1999,14, 1890–1893. [CrossRef] [PubMed]
21.
Nanaev, A.; Kohnen, G.; Milovanov, A.P.; Domogatsky, S.P.; Kaufmann, P. Stromal differentiation and architecture of the human
umbilical cord. Placenta 1997,18, 53–64. [CrossRef]
22.
Davies, J.E.; Walker, J.T.; Keating, A. Concise Review: Wharton’s Jelly: The Rich, but Enigmatic, Source of Mesenchymal Stromal
Cells. Stem Cells Transl. Med. 2017,6, 1620–1630. [CrossRef]
23. Turco, M.Y.; Moffett, A. Development of the human placenta. Development 2019,146, dev163428. [CrossRef]
24.
Furukawa, S.; Kuroda, Y.; Sugiyama, A. A Comparison of the Histological Structure of the Placenta in Experimental Animals.
J. Toxicol. Pathol. 2014,27, 11–18. [CrossRef] [PubMed]
25.
Furukawa, S.; Tsuji, N.; Sugiyama, A. Morphology and physiology of rat placenta for toxicological evaluation. J. Toxicol. Pathol.
2019,32, 1–17. [CrossRef]
26.
Leiser, R.; Dantzer, V. Initial vascularisation in the pig placenta: II. Demonstration of gland and areola-gland subunits by histology
and corrosion casts. Anat. Rec. Adv. Integr. Anat. Evol. Biol. 1994,238, 326–334. [CrossRef] [PubMed]
27.
Abd-Elnaeim, M.M.; Pfarrer, C.; Saber, A.S.; Abou-Elmagd, A.; Jones, C.J.; Leiser, R. Fetomaternal attachment and anchorage
in the early diffuse epitheliochorial placenta of the camel (Camelus dromedarius). Light, transmission, and scanning electron
microscopic study. Cells Tissues Organs 1999,164, 141–154. [CrossRef] [PubMed]
28.
Carter, A.M.; Enders, A.C. Comparative aspects of trophoblast development and placentation. Reprod. Biol. Endocrinol.
2004
,2, 46.
[CrossRef]
29.
Carter, A.M. Evolution of Placental Function in Mammals: The Molecular Basis of Gas and Nutrient Transfer, Hormone Secretion,
and Immune Responses. Physiol. Rev. 2012,92, 1543–1576. [CrossRef] [PubMed]
30. Jiang, W.; Xu, J. Immune modulation by mesenchymal stem cells. Cell Prolif. 2019,53, e12712. [CrossRef]
31.
Popis, M.; Konwerska, A.; Partyka, M.; Wieczorkiewicz, M.; Ciesiółka, S.; Stefa´nska, K.; Spaczy´nska, J.; Golkar-Narenji, A.; Jeseta,
M.; Bukowska, D.; et al. Mesenchymal stem cells and their secretome -candidates for safe and effective therapy for systemic
lupus erythematosus. Med. J. Cell Biol. 2021,9, 110–122. [CrossRef]
Cells 2021,10, 3278 27 of 39
32.
Ceccariglia, S.; Cargnoni, A.; Silini, A.R.; Parolini, O. Autophagy: A potential key contributor to the therapeutic action of
mesenchymal stem cells. Autophagy 2019,16, 28–37. [CrossRef] [PubMed]
33.
Xu, J.; Feng, Z.; Wang, X.; Xiong, Y.; Wang, L.; Ye, L.; Zhang, H. hUC-MSCs Exert a Neuroprotective Effect via Anti-apoptotic
Mechanisms in a Neonatal HIE Rat Model. Cell Transplant. 2019,28, 1552–1559. [CrossRef] [PubMed]
34.
Stavely, R.; Nurgali, K. The emerging antioxidant paradigm of mesenchymal stem cell therapy. STEM CELLS Transl. Med.
2020
,9,
985–1006. [CrossRef] [PubMed]
35.
van Buul, G.M.; Siebelt, M.; Leijs, M.J.C.; Bos, P.K.; Waarsing, J.H.; Kops, N.; Weinans, H.; Verhaar, J.A.N.; Bernsen, M.R.; van Osch,
G.J.V.M. Mesenchymal stem cells reduce pain but not degenerative changes in a mono-iodoacetate rat model of osteoarthritis.
J. Orthop. Res. 2014,32, 1167–1174. [CrossRef] [PubMed]
36.
Dominici, M.; Le Blanc, K.; Mueller, I.; Slaper-Cortenbach, I.; Marini, F.C.; Krause, D.S.; Deans, R.J.; Keating, A.; Prockop, D.J.;
Horwitz, E.M. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular
Therapy position statement. Cytotherapy 2006,8, 315–317. [CrossRef] [PubMed]
37.
Tondreau, T.; Lagneaux, L.; Dejeneffe, M.; Massy, M.; Mortier, C.; Delforge, A.; Bron, D. Bone marrow–derived mesenchymal stem
cells already express specific neural proteins before any differentiation. Differentiation 2004,72, 319–326. [CrossRef] [PubMed]
38.
Cho, S.-W.; Kim, I.-K.; Lim, S.H.; Kim, D.-I.; Kang, S.-W.; Kim, S.H.; Kim, Y.H.; Lee, E.Y.; Choi, C.Y.; Kim, B.-S. Smooth muscle-like
tissues engineered with bone marrow stromal cells. Biomaterials 2004,25, 2979–2986. [CrossRef]
39.
Hung, S.-C.; Chen, N.-J.; Hsieh, S.-L.; Li, H.; Ma, H.-L.; Lo, W.-H. Isolation and Characterization of Size-Sieved Stem Cells from
Human Bone Marrow. STEM CELLS 2002,20, 249–258. [CrossRef] [PubMed]
40.
Hung, S.-C.; Chang, C.-F.; Ma, H.-L.; Chen, T.-H.; Ho, L.L.-T. Gene expression profiles of early adipogenesis in human mesenchy-
mal stem cells. Gene 2004,340, 141–150. [CrossRef] [PubMed]
41.
George, S.; Hamblin, M.R.; Abrahamse, H. Differentiation of Mesenchymal Stem Cells to Neuroglia: In the Context of Cell
Signalling. Stem Cell Rev. Rep. 2019,15, 814–826. [CrossRef] [PubMed]
42.
Martin, G.R. Isolation of a pluripotent cell line from early mouse embryos cultured in medium conditioned by teratocarcinoma
stem cells. Proc. Natl. Acad. Sci. USA 1981,78, 7634–7638. [CrossRef] [PubMed]
43. Carpenter, M.K.; Rößler, E.; Rao, M.S. Characterization and Differentiation of Human Embryonic Stem Cells. Cloning Stem Cells
2003,5, 79–88. [CrossRef] [PubMed]
44.
Hoffman, L.M.; Carpenter, M.K. Characterization and culture of human embryonic stem cells. Nat. Biotechnol.
2005
,23, 699–708.
[CrossRef] [PubMed]
45.
Goldring, C.E.; Duffy, P.A.; Benvenisty, N.; Andrews, P.W.; Ben-David, U.; Eakins, R.; French, N.; Hanley, N.A.; Kelly, L.;
Kitteringham, N.R.; et al. Assessing the Safety of Stem Cell Therapeutics. Cell Stem Cell 2011,8, 618–628. [CrossRef] [PubMed]
46.
Ren, G.; Chen, X.; Dong, F.; Li, W.; Ren, X.; Zhang, Y.; Shi, Y. Concise Review: Mesenchymal Stem Cells and Translational
Medicine: Emerging Issues. STEM CELLS Transl. Med. 2012,1, 51–58. [CrossRef] [PubMed]
47.
Prockop, D.J.; Brenner, M.; Fibbe, W.E.; Horwitz, E.; Le Blanc, K.; Phinney, D.G.; Simmons, P.J.; Sensebe, L.; Keating, A. Defining
the risks of mesenchymal stromal cell therapy. Cytotherapy 2010,12, 576–578. [CrossRef] [PubMed]
48.
Handgretinger, R.; Schilbach, K. The potential role of
γδ
T cells after allogeneic HCT for leukemia. Blood
2018
,131, 1063–1072.
[CrossRef] [PubMed]
49.
Hemmingsen, M.; Vedel, S.; Skafte-Pedersen, P.; Sabourin, D.; Collas, P.; Bruus, H.; Dufva, M. The Role of Paracrine and Autocrine
Signaling in the Early Phase of Adipogenic Differentiation of Adipose-derived Stem Cells. PLoS ONE
2013
,8, e63638. [CrossRef]
[PubMed]
50.
Moghadam, F.H.; Tayebi, T.; Dehghan, M.; Eslami, G.; Nadri, H.; Moradi, A.; Vahedian-Ardakani, H.; Barzegar, K. Differentiation
of Bone Marrow Mesenchymal Stem Cells into Chondrocytes after Short Term Culture in Alkaline Medium. Int. J. Hematol. Stem
Cell Res. 2014,8, 12–19.
51.
Wang, J.-J.; Ye, F.; Cheng, L.-J.; Shi, Y.-J.; Bao, J.; Sun, H.-Q.; Wang, W.; Zhang, P.; Bu, H. Osteogenic differentiation of mesenchymal
stem cells promoted by overexpression of connective tissue growth factor. J. Zhejiang Univ. Sci. B 2009,10, 355–367. [CrossRef]
52.
Tropel, P.; Platet, N.; Platel, J.-C.; Noël, D.; Albrieux, M.; Benabid, A.-L.; Berger, F. Functional Neuronal Differentiation of Bone
Marrow-Derived Mesenchymal Stem Cells. STEM CELLS 2006,24, 2868–2876. [CrossRef] [PubMed]
53.
Hass, R.; Kasper, C.; Böhm, S.; Jacobs, R. Different populations and sources of human mesenchymal stem cells (MSC): A
comparison of adult and neonatal tissue-derived MSC. Cell Commun. Signal. 2011,9, 12. [CrossRef]
54.
Camernik, K.; Miheliˇc, A.; Mihaliˇc, R.; Haring, G.; Herman, S.; Presen, D.M.; Janež, A.; Trebše, R.; Marc, J.; Zupan, J. Comprehen-
sive analysis of skeletal muscle- and bone-derived mesenchymal stem/stromal cells in patients with osteoarthritis and femoral
neck fracture. Stem Cell Res. Ther. 2020,11, 146. [CrossRef] [PubMed]
55.
ˇ
Camernik, K.; Miheliˇc, A.; Mihaliˇc, R.; Presen, D.M.; Janež, A.; Trebše, R.; Marc, J.; Zupan, J. Increased Exhaustion of the
Subchondral Bone-Derived Mesenchymal Stem/Stromal Cells in Primary Versus Dysplastic Osteoarthritis. Stem Cell Rev. Rep.
2020,16, 742–754. [CrossRef]
56.
Zupan, J.; Strazar, K.; Kocijan, R.; Nau, T.; Grillari, J.; Presen, D.M. Age-related alterations and senescence of mesenchymal
stromal cells: Implications for regenerative treatments of bones and joints. Mech. Ageing Dev. 2021,198, 111539. [CrossRef]
57.
Pevsner-Fischer, M.; Levin, S.; Zipori, D. The Origins of Mesenchymal Stromal Cell Heterogeneity. Stem Cell Rev. Rep.
2011
,7,
560–568. [CrossRef] [PubMed]
Cells 2021,10, 3278 28 of 39
58.
Russell, A.L.; Lefavor, R.; Durand, N.; Glover, L.; Zubair, A.C. Modifiers of mesenchymal stem cell quantity and quality.
Transfusion 2018,58, 1434–1440. [CrossRef] [PubMed]
59.
Pizzute, T.; Lynch, K.; Pei, M. Impact of Tissue-Specific Stem Cells on Lineage-Specific Differentiation: A Focus on the Muscu-
loskeletal System. Stem Cell Rev. Rep. 2015,11, 119–132. [CrossRef]
60.
Mattiucci, D.; Maurizi, G.; Leoni, P.; Poloni, A. Aging- and Senescence-associated Changes of Mesenchymal Stromal Cells in
Myelodysplastic Syndromes. Cell Transplant. 2018,27, 754–764. [CrossRef]
61.
Malgieri, A.; Kantzari, E.; Patrizi, M.P.; Gambardella, S. Bone marrow and umbilical cord blood human mesenchymal stem cells:
State of the art. Int. J. Clin. Exp. Med. 2010,3, 248–269. [PubMed]
62.
D’Ippolito, G.; Schiller, P.C.; Ricordi, C.; Roos, B.A.; Howard, G.A. Age-Related Osteogenic Potential of Mesenchymal Stromal
Stem Cells from Human Vertebral Bone Marrow. J. Bone Miner. Res. 1999,14, 1115–1122. [CrossRef] [PubMed]
63.
Ahmed, A.; Sheng, M.H.; Wasnik, S.; Baylink, D.J.; Lau, K.-H.W. Effect of aging on stem cells. World J. Exp. Med.
2017
,7, 1–10.
[CrossRef] [PubMed]
64.
Fukada, S.-I.; Ma, Y.; Uezumi, A. Adult stem cell and mesenchymal progenitor theories of aging. Front. Cell Dev. Biol.
2014
,2, 10.
[CrossRef] [PubMed]
65.
Babenko, V.A.; Silachev, D.N.; Danilina, T.I.; Goryunov, K.V.; Pevzner, I.B.; Zorova, L.D.; Popkov, V.A.; Chernikov, V.P.; Plotnikov,
E.Y.; Sukhikh, G.T.; et al. Age-Related Changes in Bone-Marrow Mesenchymal Stem Cells. Cells
2021
,10, 1273. [CrossRef]
[PubMed]
66. Fehrer, C.; Lepperdinger, G. Mesenchymal stem cell aging. Exp. Gerontol. 2005,40, 926–930. [CrossRef] [PubMed]
67.
Miao, Z.; Jin, J.; Chen, L.; Zhu, J.; Huang, W.; Zhao, J.; Qian, H.; Zhang, X. Isolation of mesenchymal stem cells from human
placenta: Comparison with human bone marrow mesenchymal stem cells. Cell Biol. Int.
2006
,30, 681–687. [CrossRef] [PubMed]
68.
Beeravolu, N.; Khan, I.; McKee, C.; Dinda, S.; Thibodeau, B.; Wilson, G.; Perez-Cruet, M.; Bahado-Singh, R.; Chaudhry, G.R.
Isolation and comparative analysis of potential stem/progenitor cells from different regions of human umbilical cord. Stem Cell
Res. 2016,16, 696–711. [CrossRef] [PubMed]
69.
Beeravolu, N.; McKee, C.; Alamri, A.; Mikhael, S.; Brown, C.; Perez-Cruet, M.; Chaudhry, G.R. Isolation and Characterization of
Mesenchymal Stromal Cells from Human Umbilical Cord and Fetal Placenta. J. Vis. Exp. 2017,122, e55224. [CrossRef]
70.
Raileanu, V.N.; Whiteley, J.; Chow, T.; Kollara, A.; Mohamed, A.; Keating, A.; Rogers, I.M. Banking Mesenchymal Stromal Cells
from Umbilical Cord Tissue: Large Sample Size Analysis Reveals Consistency Between Donors. STEM CELLS Transl. Med.
2019
,
8, 1041–1054. [CrossRef]
71.
Maurice, S.; Srouji, S.; Livne, E. Isolation of progenitor cells from cord blood using adhesion matrices. Cytotechnology
2007
,52,
125–137. [CrossRef]
72.
Gucciardo, L.; Lories, R.; Ochsenbein-Kölble, N.; Done, E.; Zwijsen, A.; Deprest, J.; Ochsenbein-Kölble, N. Fetal mesenchymal
stem cells: Isolation, properties and potential use in perinatology and regenerative medicine. BJOG Int. J. Obstet. Gynaecol.
2008
,
116, 166–172. [CrossRef] [PubMed]
73.
Wouters, G.; Grossi, S.; Mesoraca, A.; Bizzoco, D.; Mobili, L.; Cignini, P.; Giorlandino, C. Isolation of amniotic fluid-derived
mesenchymal stem cells. J. Prenat. Med. 2007,1, 39–40.
74.
Kwon, A.; Kim, Y.; Kim, M.; Kim, J.; Choi, H.; Jekarl, D.W.; Lee, S.; Kim, J.M.; Shin, J.-C.; Park, I.Y. Tissue-specific Differentiation
Potency of Mesenchymal Stromal Cells from Perinatal Tissues. Sci. Rep. 2016,6, 23544. [CrossRef] [PubMed]
75. Caplan, A.I.; Correa, D. The MSC: An Injury Drugstore. Cell Stem Cell 2011,9, 11–15. [CrossRef]
76.
da Silva Meirelles, L.; Caplan, A.I.; Nardi, N.B. In Search of the In Vivo Identity of Mesenchymal Stem Cells. STEM CELLS
2008
,
26, 2287–2299. [CrossRef]
77.
da Silva Meirelles, L.; Fontes, A.M.; Covas, D.T.; Caplan, A.I. Mechanisms involved in the therapeutic properties of mesenchymal
stem cells. Cytokine Growth Factor Rev. 2009,20, 419–427. [CrossRef] [PubMed]
78.
Caplan, A.I.; Dennis, J.E. Mesenchymal stem cells as trophic mediators. J. Cell. Biochem.
2006
,98, 1076–1084. [CrossRef] [PubMed]
79.
Le Blanc, K.; Mougiakakos, D. Multipotent mesenchymal stromal cells and the innate immune system. Nat. Rev. Immunol.
2012
,
12, 383–396. [CrossRef] [PubMed]
80. Caplan, A.I. Adult Mesenchymal Stem Cells: When, Where, and How. Stem Cells Int. 2015,2015, 628767. [CrossRef] [PubMed]
81.
Caplan, A.I. Mesenchymal Stem Cells: Time to Change the Name! Stem Cells Transl. Med.
2017
,6, 1445–1451. [CrossRef] [PubMed]
82.
Horwitz, E.M.; Le Blanc, K.; Dominici, M.; Mueller, I.; Slaper-Cortenbach, I.; Marini, F.C.; Deans, R.J.; Krause, D.S.; Keating,
A. International Society for Cellular Therapy Clarification of the nomenclature for MSC: The International Society for Cellular
Therapy position statement. Cytotherapy 2005,7, 393–395. [CrossRef] [PubMed]
83.
Viswanathan, S.; Shi, Y.; Galipeau, J.; Krampera, M.; Leblanc, K.; Martin, I.; Nolta, J.; Phinney, D.G.; Sensebe, L. Mesenchymal
stem versus stromal cells: International Society for Cell & Gene Therapy (ISCT
®
) Mesenchymal Stromal Cell committee position
statement on nomenclature. Cytotherapy 2019,21, 1019–1024. [CrossRef] [PubMed]
84.
Mushahary, D.; Spittler, A.; Kasper, C.; Weber, V.; Charwat, V. Isolation, cultivation, and characterization of human mesenchymal
stem cells. Cytom. Part A 2018,93, 19–31. [CrossRef] [PubMed]
85.
Salehinejad, P.; Alitheen, N.B.; Ali, A.M.; Omar, A.R.; Mohit, M.; Janzamin, E.; Samani, F.S.; Torshizi, Z.; Nematollahi-Mahani,
S.N. Comparison of different methods for the isolation of mesenchymal stem cells from human umbilical cord Wharton’s jelly.
Vitr. Cell. Dev. Biol.-Anim. 2012,48, 75–83. [CrossRef]
Cells 2021,10, 3278 29 of 39
86.
Yoon, J.H.; Roh, E.Y.; Shin, S.; Jung, N.H.; Song, E.Y.; Chang, J.Y.; Kim, B.J.; Jeon, H.W. Comparison of Explant-Derived and
Enzymatic Digestion-Derived MSCs and the Growth Factors from Wharton’s Jelly. BioMed Res. Int.
2013
,2013, 1–8. [CrossRef]
[PubMed]
87.
Hendijani, F. Explant culture: An advantageous method for isolation of mesenchymal stem cells from human tissues. Cell Prolif.
2017,50, e12334. [CrossRef] [PubMed]
88.
Tkach, M.; Théry, C. Communication by Extracellular Vesicles: Where We Are and Where We Need to Go. Cell
2016
,164,
1226–1232. [CrossRef]
89.
Keerthikumar, S.; Chisanga, D.; Ariyaratne, D.; Al Saffar, H.; Anand, S.; Zhao, K.; Samuel, M.; Pathan, M.; Jois, M.; Chilamkurti,
N.; et al. ExoCarta: A Web-Based Compendium of Exosomal Cargo. J. Mol. Biol. 2016,428, 688–692. [CrossRef] [PubMed]
90.
EL Andaloussi, S.; Mäger, I.; Breakefield, X.O.; Wood, M.J.A. Extracellular vesicles: Biology and emerging therapeutic opportuni-
ties. Nat. Rev. Drug Discov. 2013,12, 347–357. [CrossRef] [PubMed]
91.
Cosenza, S.; Toupet, K.; Maumus, M.; Luz-Crawford, P.; Blanc-Brude, O.; Jorgensen, C.; Noël, D. Mesenchymal stem cells-derived
exosomes are more immunosuppressive than microparticles in inflammatory arthritis. Theranostics
2018
,8, 1399–1410. [CrossRef]
92.
Bang, O.Y.; Kim, E.H. Mesenchymal Stem Cell-Derived Extracellular Vesicle Therapy for Stroke: Challenges and Progress. Front.
Neurol. 2019,10, 211. [CrossRef] [PubMed]
93.
Wiklander, O.P.B.; Nordin, J.Z.; O’Loughlin, A.; Gustafsson, Y.; Corso, G.; Mäger, I.; Vader, P.; Lee, Y.; Sork, H.; Seow, Y.; et al.
Extracellular vesicle
in vivo
biodistribution is determined by cell source, route of administration and targeting. J. Extracell. Vesicles
2015,4, 26316. [CrossRef] [PubMed]
94.
Busato, A.; Bonafede, R.; Bontempi, P.; Scambi, I.; Schiaffino, L.; Benati, D.; Malatesta, M.; Sbarbati, A.; Marzola, P.; Mariotti, R.
Labeling and Magnetic Resonance Imaging of Exosomes Isolated from Adipose Stem Cells. Curr. Protoc. Cell Biol.
2017
,75, 3–44.
[CrossRef] [PubMed]
95.
da Silva Meirelles, L.; Malta, T.M.; de Deus Wagatsuma, V.M.; Palma, P.V.B.; Araújo, A.G.; Ribeiro Malmegrim, K.C.; Morato De
Oliveira, F.; Panepucci, R.A.; Silva, W.A.; Kashima Haddad, S.; et al. Cultured Human Adipose Tissue Pericytes and Mesenchymal
Stromal Cells Display a Very Similar Gene Expression Profile. Stem Cells Dev. 2015,24, 2822–2840. [CrossRef] [PubMed]
96.
da Silva Meirelles, L.; de Deus Wagatsuma, V.M.; Malta, T.; Palma, P.V.B.; Araújo, A.G.; Panepucci, R.A.; Silva, W.A.; Kashima, S.;
Covas, D. The gene expression profile of non-cultured, highly purified human adipose tissue pericytes: Transcriptomic evidence
that pericytes are stem cells in human adipose tissue. Exp. Cell Res. 2016,349, 239–254. [CrossRef]
97.
Tang, Y.; Wu, X.; Lei, W.; Pang, L.; Wan, C.; Shi, Z.; Zhao, L.; Nagy, T.R.; Peng, X.; Hu, J.; et al. TGF-beta1-induced migration of
bone mesenchymal stem cells couples bone resorption with formation. Nat. Med. 2009,15, 757–765. [CrossRef] [PubMed]
98.
Zhang, Z. Bone regeneration by stem cell and tissue engineering in oral and maxillofacial region. Front. Med.
2011
,5, 401–413.
[CrossRef] [PubMed]
99.
Gómez, S.; Vlad, M.D.; López, J.; Fernández, E. Design and properties of 3D scaffolds for bone tissue engineering. Acta Biomater.
2016,42, 341–350. [CrossRef] [PubMed]
100.
Paspaliaris, V.; Kolios, G. Stem cells in Osteoporosis: From Biology to New Therapeutic Approaches. Stem Cells Int.
2019
,2019,
1–16. [CrossRef]
101.
De Witte, T.-M.; Fratila-Apachitei, L.E.; Zadpoor, A.A.; Peppas, N.A. Bone tissue engineering via growth factor delivery: From
scaffolds to complex matrices. Regen. Biomater. 2018,5, 197–211. [CrossRef] [PubMed]
102.
Vadalà, G.; Ambrosio, L.; Russo, F.; Papalia, R.; Denaro, V. Interaction between Mesenchymal Stem Cells and Intervertebral Disc
Microenvironment: From Cell Therapy to Tissue Engineering. Stem Cells Int. 2019,2019, 1–15. [CrossRef]
103.
Estes, B.T.; Diekman, B.O.; Gimble, J.M.; Guilak, F. Isolation of adipose-derived stem cells and their induction to a chondrogenic
phenotype. Nat. Protoc. 2010,5, 1294–1311. [CrossRef] [PubMed]
104.
Orbay, H.; Devi, K.; Williams, P.A.; Dehghani, T.; Silva, E.A.; Sahar, D.E. Comparison of Endothelial Differentiation Capacities of
Human and Rat Adipose-Derived Stem Cells. Plast. Reconstr. Surg. 2016,138, 1231–1241. [CrossRef] [PubMed]
105.
Xu, T.; Yu, X.; Yang, Q.; Liu, X.; Fang, J.; Dai, X. Autologous Micro-Fragmented Adipose Tissue as Stem Cell-Based Natural
Scaffold for Cartilage Defect Repair. Cell Transplant. 2019,28, 1709–1720. [CrossRef] [PubMed]
106.
Zhou, L.-L.; Liu, W.; Wu, Y.-M.; Sun, W.-L.; Dörfer, C.E.; El-Sayed, K.M.F. Oral Mesenchymal Stem/Progenitor Cells: The
Immunomodulatory Masters. Stem Cells Int. 2020,2020, 1–16. [CrossRef] [PubMed]
107.
Gan, L.; Liu, Y.; Cui, D.; Pan, Y.; Zheng, L.; Wan, M. Dental Tissue-Derived Human Mesenchymal Stem Cells and Their Potential
in Therapeutic Application. Stem Cells Int. 2020,2020, 1–17. [CrossRef] [PubMed]
108.
Yang, R.; Wang, J.; Chen, X.; Shi, Y.; Xie, J. Epidermal Stem Cells in Wound Healing and Regeneration. Stem Cells Int.
2020
,2020,
1–11. [CrossRef]
109.
Lee, D.E.; Ayoub, N.; Agrawal, D.K. Mesenchymal stem cells and cutaneous wound healing: Novel methods to increase cell
delivery and therapeutic efficacy. Stem Cell Res. Ther. 2016,7, 1–8. [CrossRef] [PubMed]
110.
Jeon, Y.K.; Jang, Y.H.; Yoo, D.R.; Na Kim, S.; Lee, S.K.; Nam, M.J. Mesenchymal stem cells’ interaction with skin: Wound-healing
effect on fibroblast cells and skin tissue. Wound Repair Regen. 2010,18, 655–661. [CrossRef] [PubMed]
111.
Lee, S.H.; Jin, S.Y.; Song, J.S.; Seo, K.K.; Cho, K.H. Paracrine effects of adipose-derived stem cells on keratinocytes and dermal
fibroblasts. Ann. Dermatol. 2012,24, 136–143. [CrossRef]
Cells 2021,10, 3278 30 of 39
112.
Hsieh, J.-Y.; Wang, H.-W.; Chang, S.-J.; Liao, K.-H.; Lee, I.-H.; Lin, W.-S.; Wu, C.-H.; Lin, W.-Y.; Cheng, S.-M. Mesenchymal
Stem Cells from Human Umbilical Cord Express Preferentially Secreted Factors Related to Neuroprotection, Neurogenesis, and
Angiogenesis. PLoS ONE 2013,8, e72604. [CrossRef]
113.
I Arno, A.; Amini-Nik, S.; Blit, P.H.; Al-Shehab, M.; Belo, C.; Herer, E.; Tien, C.H.; Jeschke, M.G. Human Wharton’s jelly
mesenchymal stem cells promote skin wound healing through paracrine signaling. Stem Cell Res. Ther. 2014,5, 28. [CrossRef]
114.
Zhao, Y.-X.; Chen, S.-R.; Su, P.-P.; Huang, F.-H.; Shi, Y.-C.; Shi, Q.-Y.; Lin, S. Using Mesenchymal Stem Cells to Treat Female
Infertility: An Update on Female Reproductive Diseases. Stem Cells Int. 2019,2019, 1–10. [CrossRef]
115.
Kossowska-Tomaszczuk, K.; Geyter, C. De Cells with Stem Cell Characteristics in Somatic Compartments of the Ovary. Biomed
Res. Int. 2013,2013, 310859. [CrossRef]
116.
Hoang, S.N.; Ho, Q.C.; Nguyen, T.T.P.; Doan, C.C.; Tran, D.H.; Le, L.T. Evaluation of stemness marker expression in bovine
ovarian granulosa cells. Anim. Reprod. 2019,16, 277–281. [CrossRef] [PubMed]
117.
Varras, M.; Griva, T.; Kalles, V.; Akrivis, C.; Paparisteidis, N. Markers of stem cells in human ovarian granulosa cells: Is there a
clinical significance in ART? J. Ovarian Res. 2012,5, 36. [CrossRef] [PubMed]
118.
Kossowska-Tomaszczuk, K.; Pelczar, P.; Güven, S.; Kowalski, J.; Volpi, E.; De Geyter, C.; Scherberich, A. A Novel Three-
Dimensional Culture System Allows Prolonged Culture of Functional Human Granulosa Cells and Mimics the Ovarian Environ-
ment. Tissue Eng. Part A 2010,16, 2063–2073. [CrossRef] [PubMed]
119.
Kossowska-Tomaszczuk, K.; De Geyter, C.; De Geyter, M.; Martin, I.; Holzgreve, W.; Scherberich, A.; Zhang, H. The Multipotency
of Luteinizing Granulosa Cells Collected from Mature Ovarian Follicles. Stem Cells 2009,27, 210–219. [CrossRef] [PubMed]
120.
Brevini, T.A.L.; Pennarossa, G.; Rahman, M.M.; Paffoni, A.; Antonini, S.; Ragni, G.; Deeguileor, M.; Tettamanti, G.; Gandolfi, F.
Morphological and Molecular Changes of Human Granulosa Cells Exposed to 5-Azacytidine and Addressed Toward Muscular
Differentiation. Stem Cell Rev. Rep. 2014,10, 633–642. [CrossRef] [PubMed]
121.
Br ˛azert, M.; Kranc, W.; Celichowski, P.; Jankowski, M.; Piotrowska-Kempisty, H.; Pawelczyk, L.; Bruska, M.; Zabel, M.; Nowicki,
M.; Kempisty, B. Expression of genes involved in neurogenesis, and neuronal precursor cell proliferation and development:
Novel pathways of human ovarian granulosa cell differentiation and transdifferentiation capability
in vitro
.Mol. Med. Rep.
2020
,
21, 1749–1760. [CrossRef] [PubMed]
122.
Virant-Klun, I.; Omejec, S.; Stimpfel, M.; Skerl, P.; Novakovic, S.; Jancar, N.; Vrtacnik-Bokal, E. Female Age Affects the Mesenchy-
mal Stem Cell Characteristics of Aspirated Follicular Cells in the In Vitro Fertilization Programme. Stem Cell Rev. Rep.
2019
,15,
543–557. [CrossRef] [PubMed]
123.
Guillot, P.V.; Gotherstrom, C.; Chan, J.; Kurata, H.; Fisk, N.M. Human First-Trimester Fetal MSC Express Pluripotency Markers
and Grow Faster and Have Longer Telomeres Than Adult MSC. Stem Cells 2007,25, 646–654. [CrossRef] [PubMed]
124.
Blum, B.; Benvenisty, N. The Tumorigenicity of Human Embryonic Stem Cells. Adv. Cancer Res.
2008
,100, 133–158. [CrossRef]
[PubMed]
125.
Fong, C.-Y.; Gauthaman, K.; Bongso, A. Teratomas from pluripotent stem cells: A clinical hurdle. J. Cell. Biochem.
2010
,111,
769–781. [CrossRef] [PubMed]
126.
Götherström, C.; Ringdén, O.; Tammik, C.; Zetterberg, E.; Westgren, M.; Le Blanc, K. Immunologic properties of human fetal
mesenchymal stem cells. Am. J. Obstet. Gynecol. 2004,190, 239–245. [CrossRef]
127.
Götherström, C.; West, A.; Liden, J.; Uzunel, M.; Lahesmaa, R.; Le Blanc, K. Difference in gene expression between human fetal
liver and adult bone marrow mesenchymal stem cells. Haematologica 2005,90, 1017–1026. [PubMed]
128.
Mirmalek-Sani, S.-H.; Tare, R.S.; Morgan, S.M.; Roach, H.I.; Wilson, D.I.; Hanley, N.A.; Oreffo, R.O. Characterization and
Multipotentiality of Human Fetal Femur-Derived Cells: Implications for Skeletal Tissue Regeneration. Stem Cells
2006
,24,
1042–1053. [CrossRef]
129.
Guillot, P.V.; De Bari, C.; Dell’Accio, F.; Kurata, H.; Polak, J.; Fisk, N.M. Comparative osteogenic transcription profiling of various
fetal and adult mesenchymal stem cell sources. Differ. 2008,76, 946–957. [CrossRef] [PubMed]
130. O’Donoghue, K.; Fisk, N.M. Fetal stem cells. Best Pract. Res. Clin. Obstet. Gynaecol. 2004,18, 853–875.
131.
Brumbaugh, J.; Rose, C.; Phanstiel, D.H.; Thomson, J.A.; Coon, J.J. Proteomics and pluripotency. Crit. Rev. Biochem. Mol. Biol.
2011,46, 493–506. [CrossRef]
132.
Radzisheuskaya, A.; Le Bin Chia, G.; dos Santos, R.L.; Theunissen, T.; Castro, L.F.; Nichols, J.; Silva, J.C.R. A defined Oct4 level
governs cell state transitions of pluripotency entry and differentiation into all embryonic lineages. Nat. Cell Biol.
2013
,15, 579–590.
[CrossRef]
133.
Pan, G.J.; Chang, Z.Y.; Schöler, H.R.; Pei, D. Stem cell pluripotency and transcription factor Oct4. Cell Res.
2002
,12, 321–329.
[CrossRef] [PubMed]
134.
Seruya, M.; Shah, A.; Pedrotty, D.; Du Laney, T.; Melgiri, R.; McKee, J.A.; Young, H.E.; Niklason, L.E. Clonal Population of Adult
Stem Cells: Life Span and Differentiation Potential. Cell Transplant. 2004,13, 93–101. [CrossRef]
135.
Xue, G.; Han, X.; Ma, X.; Wu, H.; Qin, Y.; Liu, J.; Hu, Y.; Hong, Y.; Hou, Y. Effect of Microenvironment on Differentiation of Human
Umbilical Cord Mesenchymal Stem Cells into Hepatocytes In Vitro and In Vivo. Biomed Res. Int. 2016,2016, 1–13.
136.
Kusuma, G.D.; Carthew, J.R.; Lim, R.; Frith, J.E. Effect of the Microenvironment on Mesenchymal Stem Cell Paracrine Signaling:
Opportunities to Engineer the Therapeutic Effect. Stem Cells Dev. 2017,26, 617–631. [CrossRef] [PubMed]
Cells 2021,10, 3278 31 of 39
137.
Robert, A.W.; Marcon, B.H.; Dallagiovanna, B.; Shigunov, P. Adipogenesis, Osteogenesis, and Chondrogenesis of Human
Mesenchymal Stem/Stromal Cells: A Comparative Transcriptome Approach. Front. Cell Dev. Biol.
2020
,8, 561. [CrossRef]
[PubMed]
138.
Munir, H.; Ward, L.; Sheriff, L.; Kemble, S.; Nayar, S.; Barone, F.; Nash, G.B.; McGettrick, H.M. Adipogenic Differentiation of
Mesenchymal Stem Cells Alters Their Immunomodulatory Properties in a Tissue-Specific Manner. Stem Cells
2017
,35, 1636–1646.
[CrossRef]
139.
José, B.; Andrades, J.A.; Ertl, D.C.; Sorgente, N.; Nimni, M.E. Demineralized bone matrix mediates differentiation of bone marrow
stromal cells in vitro: Effect of age of cell donor. J. Bone Miner. Res. 2009,11, 1703–1714. [CrossRef] [PubMed]
140.
Zhao, M.; Li, P.; Xu, H.; Pan, Q.; Zeng, R.; Ma, X.; Li, Z.; Lin, H. Dexamethasone-Activated MSCs Release MVs for Stimulating
Osteogenic Response. Stem Cells Int. 2018,2018, 1–12. [CrossRef]
141.
Huang, C.-Y.C.; Hagar, K.L.; Frost, L.E.; Sun, Y.; Cheung, H.S. Effects of Cyclic Compressive Loading on Chondrogenesis of
Rabbit Bone-Marrow Derived Mesenchymal Stem Cells. Stem Cells 2004,22, 313–323. [CrossRef] [PubMed]
142.
Tontonoz, P.; Hu, E.; Spiegelman, B.M. Stimulation of adipogenesis in fibroblasts by PPAR
γ
2, a lipid-activated transcription factor.
Cell 1994,79, 1147–1156. [CrossRef]
143. Rangwala, S.M.; Lazar, M.A. Transcriptional Control of Adipogenesis. Annu. Rev. Nutr. 2000,20, 535–559. [CrossRef]
144.
Furumatsu, T.; Tsuda, M.; Taniguchi, N.; Tajima, Y.; Asahara, H. Smad3 Induces Chondrogenesis through the Activation of SOX9
via CREB-binding Protein/p300 Recruitment. J. Biol. Chem. 2005,280, 8343–8350. [CrossRef]
145.
Chu, M.-S.; Chang, C.-F.; Yang, C.-C.; Bau, Y.-C.; Ho, L.L.-T.; Hung, S.-C. Signalling pathway in the induction of neurite outgrowth
in human mesenchymal stem cells. Cell. Signal. 2006,18, 519–530. [CrossRef] [PubMed]
146.
Wang, T.; Tio, M.; Lee, W.; Beerheide, W.; Udolph, G. Neural differentiation of mesenchymal-like stem cells from cord blood is
mediated by PKA. Biochem. Biophys. Res. Commun. 2007,357, 1021–1027. [CrossRef]
147.
Yanjie, J.; Jiping, S.; Yan, Z.; Xiaofeng, Z.; Boai, Z.; Yajun, L. Effects of Notch-1 signalling pathway on differentiation of marrow
mesenchymal stem cells into neurons in vitro. Neuro Report 2007,18, 1443–1447. [CrossRef]
148.
Qian, L.; Saltzman, W. Improving the expansion and neuronal differentiation of mesenchymal stem cells through culture surface
modification. Biomater. 2004,25, 1331–1337. [CrossRef] [PubMed]
149.
D’Ippolito, G.; Diabira, S.; Howard, G.A.; Menei, P.; Roos, B.A.; Schiller, P.C. Marrow-isolated adult multilineage inducible
(MIAMI) cells, a unique population of postnatal young and old human cells with extensive expansion and differentiation
potential. J. Cell Sci. 2004,117, 2971–2981. [CrossRef] [PubMed]
150.
Ding, D.-C.; Shyu, W.-C.; Chiang, M.-F.; Lin, S.-Z.; Chang, Y.-C.; Wang, H.-J.; Su, C.-Y.; Li, H. Enhancement of neuroplasticity
through upregulation of
β
1-integrin in human umbilical cord-derived stromal cell implanted stroke model. Neurobiol. Dis.
2007
,
27, 339–353. [CrossRef] [PubMed]
151.
Inada, M.; Follenzi, A.; Cheng, K.; Surana, M.; Joseph, B.; Benten, D.; Bandi, S.; Qian, H.; Gupta, S. Phenotype reversion in fetal
human liver epithelial cells identifies the role of an intermediate meso-endodermal stage before hepatic maturation. J. Cell Sci.
2008,121, 1002–1013. [CrossRef]
152.
Yoshida, C.A.; Komori, H.; Maruyama, Z.; Miyazaki, T.; Kawasaki, K.; Furuichi, T.; Fukuyama, R.; Mori, M.; Yamana, K.;
Nakamura, K.; et al. SP7 Inhibits Osteoblast Differentiation at a Late Stage in Mice. PLoS ONE 2012,7, e32364. [CrossRef]
153.
Hagh, M.F.; Noruzinia, M.; Mortazavi, Y.; Soleimani, M.; Kaviani, S.; Abroun, S.; Fard, A.D.; Maymand, M.M. Different
Methylation Patterns of RUNX2, OSX, DLX5 and BSP in Osteoblastic Differentiation of Mesenchymal Stem Cells. Cell J
2017
,17,
71–82.
154.
Leucht, P.; Lee, S.; Yim, N. Wnt signaling and bone regeneration: Can’t have one without the other. Biomaterials
2019
,196, 46–50.
[CrossRef]
155.
Celil, A.B.; Campbell, P.G. BMP-2 and Insulin-like Growth Factor-I Mediate Osterix (Osx) Expression in Human Mesenchymal
Stem Cells via the MAPK and Protein Kinase D Signaling Pathways. J. Biol. Chem.
2005
,280, 31353–31359. [CrossRef] [PubMed]
156.
Zhang, W.; Shen, X.; Wan, C.; Zhao, Q.; Zhang, L.; Zhou, Q.; Deng, L. Effects of insulin and insulin-like growth factor 1 on
osteoblast proliferation and differentiation: Differential signalling via Akt and ERK. Cell Biochem. Funct.
2012
,30, 297–302.
[CrossRef] [PubMed]
157.
Liang, H.; Xu, X.; Feng, X.; Ma, L.; Deng, X.; Wu, S.; Liu, X.; Yang, C. Gold nanoparticles-loaded hydroxyapatite composites guide
osteogenic differentiation of human mesenchymal stem cells through Wnt/
β
-catenin signaling pathway. Int. J. Nanomed.
2019
,
2019, 6151–6163. [CrossRef] [PubMed]
158.
Brady, K.; Dickinson, S.C.; Guillot, P.V.; Polak, J.; Blom, A.W.; Kafienah, W.; Hollander, A.P. Human Fetal and Adult Bone
Marrow-Derived Mesenchymal Stem Cells Use Different Signaling Pathways for the Initiation of Chondrogenesis. Stem Cells Dev.
2014,23, 541–554. [CrossRef] [PubMed]
159.
Sibiak, R.; Jaworski, M.; Dorna, Z.; Pie ´nkowski, W.; Stefa´nska, K.; Bryl, R.; Žáková, J.; Crha, I.; Ventruba, P.; Ješeta, M.; et al.
Human placenta-derived stem cells-recent findings based on the molecular science. Med J. Cell Biol.
2020
,8, 164–169. [CrossRef]
160.
Zhao, Q.; Han, Z.; Wang, J.; Han, Z. Development and investigational new drug application of mesenchymal stem/stromal cells
products in China. Stem Cells Transl. Med. 2021,10, S18–S30. [CrossRef]
161.
Cellular & Gene Therapy Products. Available online: https://www.fda.gov/vaccines-blood-biologics/cellular-gene-therapy-
products (accessed on 11 November 2021).
Cells 2021,10, 3278 32 of 39
162.
Blasimme, A.; Rial-Sebbag, E. Regulation of Cell-Based Therapies in Europe: Current Challenges and Emerging Issues. Stem Cells
Dev. 2013,22, 14–19. [CrossRef] [PubMed]
163.
Regulation of Stem Cell Research in Europe. Available online: https://www.eurostemcell.org/pl/regulation-stem-cell-research-
europe (accessed on 11 November 2021).
164.
Silini, A.R.; Masserdotti, A.; Papait, A.; Parolini, O. Shaping the Future of Perinatal Cells: Lessons From the Past and Interpreta-
tions of the Present. Front. Bioeng. Biotechnol. 2019,7, 75. [CrossRef] [PubMed]
165.
Müller, M.; Czarnecka, J.; Brzezi´nski, M.; Prus, J.; Kulak, B.; Hołubowski, A.; Stasiak, M.; Borowiec, B.; Bryl, R.; Moncrieff, L.; et al.
Current stem cells technologies used in medicine. Med J. Cell Biol. 2021,8, 124–138. [CrossRef]
166.
Erices, A.; Conget, P.; Minguell, J.J. Mesenchymal progenitor cells in human umbilical cord blood. Br. J. Haematol.
2000
,109,
235–242. [CrossRef] [PubMed]
167.
Moschidou, D.; Drews, K.; Eddaoudi, A.; Adjaye, J.; De Coppi, P.; Guillot, P.V. Molecular Signature of Human amniotic Fluid
Stem Cells During Fetal Development. Curr. Stem Cell Res. Ther. 2013,8, 73–81. [CrossRef] [PubMed]
168.
Park, S.; Koh, S.-E.; Hur, C.Y.; Lee, W.-D.; Lim, J.; Lee, Y.-J. Comparison of human first and third trimester placental mesenchymal
stem cell. Cell Biol. Int. 2013,37, 242–249. [CrossRef]
169.
Izumi, M.; Pazin, B.J.; Minervini, C.F.; Gerlach, J.; Ross, M.A.; Stolz, D.B.; Turner, M.E.; Thompson, R.L.; Miki, T. Quantitative
comparison of stem cell marker-positive cells in fetal and term human amnion. J. Reprod. Immunol. 2009,81, 39–43. [CrossRef]
170.
Savickiene, J.; Treigyte, G.; Baronaite, S.; Valiuliene, G.; Kaupinis, A.; Valius, M.; Arlauskiene, A.; Navakauskien
˙
e, R. Human
Amniotic Fluid Mesenchymal Stem Cells from Second- and Third-Trimester Amniocentesis: Differentiation Potential, Molecular
Signature, and Proteome Analysis. Stem Cells Int. 2015,2015, 319238. [CrossRef] [PubMed]
171.
Alessio, N.; Pipino, C.; Mandatori, D.; Di Tomo, P.; Ferone, A.; Marchiso, M.; Melone, M.A.; Peluso, G.; Pandolfi, A.; Galderisi, U.
Mesenchymal stromal cells from amniotic fluid are less prone to senescence compared to those obtained from bone marrow: An
in vitro study. J. Cell. Physiol. 2018,233, 8996–9006. [CrossRef]
172.
Galderisi, U.; Peluso, G.; Di Bernardo, G. Clinical Trials Based on Mesenchymal Stromal Cells are Exponentially Increasing:
Where are We in Recent Years? Stem Cell Rev. Rep. 2021, 1–14. [CrossRef] [PubMed]
173.
Squillaro, T.; Peluso, G.; Galderisi, U. Clinical Trials with Mesenchymal Stem Cells: An Update. Cell Transplant.
2016
,25, 829–848.
[CrossRef]
174.
Silini, A.R.; Di Pietro, R.; Lang-Olip, I.; Alviano, F.; Banerjee, A.; Basile, M.; Borutinskaite, V.; Eissner, G.; Gellhaus, A.; Giebel,
B.; et al. Perinatal Derivatives: Where Do We Stand? A Roadmap of the Human Placenta and Consensus for Tissue and Cell
Nomenclature. Front. Bioeng. Biotechnol. 2020,8, 174. [CrossRef] [PubMed]
175.
Bailo, M.; Soncini, M.; Vertua, E.; Signoroni, P.B.; Sanzone, S.; Lombardi, G.; Arienti, D.; Calamani, F.; Zatti, D.; Paul, P.; et al.
Engraftment Potential of Human Amnion and Chorion Cells Derived from Term Placenta. Transplantation
2004
,78, 1439–1448.
[CrossRef] [PubMed]
176.
Bartholomew, S.; Owens, S.D.; Ferraro, G.L.; Carrade, D.D.; Lara, D.J.; Librach, F.A.; Borjesson, D.L.; Galuppo, L.D. Collection of
equine cord blood and placental tissues in 40 thoroughbred mares. Equine Veter-J. 2009,41, 724–728. [CrossRef]
177.
Carrade, D.D.; Owens, S.D.; Galuppo, L.D.; Vidal, M.A.; Ferraro, G.L.; Librach, F.; Buerchler, S.; Friedman, M.S.; Walker, N.J.;
Borjesson, D.L. Clinicopathologic findings following intra-articular injection of autologous and allogeneic placentally derived
equine mesenchymal stem cells in horses. Cytotherapy 2011,13, 419–430. [CrossRef] [PubMed]
178.
Toupadakis, C.A.; Wong, A.; Genetos, D.C.; Cheung, W.K.; Borjesson, D.L.; Ferraro, G.L.; Galuppo, L.D.; Leach, J.K.; Owens, S.D.;
Yellowley, C.E. Comparison of the osteogenic potential of equine mesenchymal stem cells from bone marrow, adipose tissue,
umbilical cord blood, and umbilical cord tissue. Am. J. Veter-Res. 2010,71, 1237–1245. [CrossRef] [PubMed]
179.
Schuh, E.M.; Friedman, M.S.; Carrade, D.D.; Li, J.; Heeke, D.; Oyserman, S.M.; Galuppo, L.D.; Lara, D.J.; Walker, N.J.; Ferraro,
G.L.; et al. Identification of variables that optimize isolation and culture of multipotent mesenchymal stem cells from equine
umbilical-cord blood. Am. J. Veter-Res. 2009,70, 1526–1535. [CrossRef] [PubMed]
180.
Steven Shaw, S.W.; Bollini, S.; Nader, K.A.; Gastadello, A.; Mehta, V.; Filppi, E.; Cananzi, M.; Gaspar, H.B.; Qasim, W.; de Coppi,
P.; et al. Autologous transplantation of amniotic fluid-derived mesenchymal stem cells into sheep fetuses. Cell Transplant.
2011
,
20, 1015–1031. [CrossRef] [PubMed]
181.
Colosimo, A.; Curini, V.; Russo, V.; Mauro, A.; Bernabò, N.; Marchisio, M.; Alfonsi, M.; Muttini, A.; Mattioli, M.; Barboni, B.
Characterization, GFP Gene Nucleofection, and Allotransplantation in Injured Tendons of Ovine Amniotic Fluid-Derived Stem
Cells. Cell Transplant. 2013,22, 99–117. [CrossRef]
182.
Ribitsch, I.; Chang-Rodriguez, S.; Egerbacher, M.; Gabner, S.; Gueltekin, S.; Huber, J.; Schuster, T.; Jenner, F. Sheep Placenta
Cotyledons: A Noninvasive Source of Ovine Mesenchymal Stem Cells. Tissue Eng. Part C Methods 2017,23, 298–310. [CrossRef]
183.
Zhan, X.-S.; El-Ashram, S.; Luo, D.-Z.; Luo, H.-N.; Wang, B.-Y.; Chen, S.-F.; Bai, Y.-S.; Chen, Z.-S.; Liu, C.-Y.; Ji, H.-Q. A
Comparative Study of Biological Characteristics and Transcriptome Profiles of Mesenchymal Stem Cells from Different Canine
Tissues. Int. J. Mol. Sci. 2019,20, 1485. [CrossRef]
184.
Borghesi, J.; Lima, M.F.; Mario, L.C.; Anunciação, A.R.D.A.D.; Rabelo, A.C.S.; da Silva, M.G.K.C.; Fernandes, F.A.; Miglino, M.A.;
Carreira, A.C.O.; Favaron, P.O. Canine amniotic membrane mesenchymal stromal/stem cells: Isolation, characterization and
differentiation. Tissue Cell 2019,58, 99–106. [CrossRef] [PubMed]
Cells 2021,10, 3278 33 of 39
185.
Saulnier, N.; Loriau, J.; Febre, M.; Robert, C.; Rakic, R.; Bonte, T.; Buff, S.; Maddens, S. Canine placenta: A promising potential
source of highly proliferative and immunomodulatory mesenchymal stromal cells? Veter-Immunol. Immunopathol.
2016
,171,
47–55. [CrossRef] [PubMed]
186.
Long, C.; Lankford, L.; Kumar, P.; Grahn, R.; Borjesson, D.L.; Farmer, D.; Wang, A. Isolation and characterization of canine
placenta-derived mesenchymal stromal cells for the treatment of neurological disorders in dogs. Cytom. Part A
2018
,93, 82–92.
[CrossRef] [PubMed]
187.
Parolini, O.; Alviano, F.; Bagnara, G.P.; Bilic, G.; Bühring, H.-J.; Evangelista, M.; Hennerbichler, S.; Liu, B.; Magatti, M.; Mao,
N.; et al. Concise Review: Isolation and Characterization of Cells from Human Term Placenta: Outcome of the First International
Workshop on Placenta Derived Stem Cells. Stem Cells 2008,26, 300–311. [CrossRef] [PubMed]
188.
Roselli, E.A.; Lazzati, S.; Iseppon, F.; Manganini, M.; Marcato, L.; Gariboldi, M.B.; Maggi, F.; Grati, F.R.; Simoni, G. Fetal
Mesenchymal Stromal Cells From Cryopreserved Human Chorionicvilli: Cytogenetic and Molecular Analysis of Genome Stability
in Long-Term Cultures. Cytotherapy 2013,15, 1340–1351. [CrossRef]
189.
Abumaree, M.H.; Al Jumah, M.A.; Kalionis, B.; Jawdat, D.; Al Khaldi, A.; Altalabani, A.A.; Knawy, B.A. Phenotypic and
Functional Characterization of Mesenchymal Stem Cells from Chorionic Villi of Human Term Placenta. Stem Cell Rev. Rep.
2013
,
9, 16–31. [CrossRef]
190.
Igura, K.; Zhang, X.; Takahashi, K.; Mitsuru, A.; Yamaguchi, S.; Takahashi, T.A. Isolation and characterization of mesenchymal
progenitor cells from chorionic villi of human placenta. Cytotherapy 2004,6, 543–553. [CrossRef] [PubMed]
191.
Castrechini, N.M.; Murthi, P.; Gude, N.M.; Erwich, J.; Gronthos, S.; Zannettino, A.; Brennecke, S.; Kalionis, B. Mesenchymal stem
cells in human placental chorionic villi reside in a vascular Niche. Placenta 2010,31, 203–212. [CrossRef]
192.
Macias, M.I.; Grande, J.; Moreno, A.; Domínguez, I.; Bornstein, R.; Flores, A.I. Isolation and characterization of true mesenchymal
stem cells derived from human term decidua capable of multilineage differentiation into all 3 embryonic layers. Am. J. Obstet.
Gynecol. 2010,203, 495.e9–495.e23. [CrossRef]
193.
Abomaray, F.M.; Al Jumah, M.A.; Alsaad, K.O.; Jawdat, D.; Al Khaldi, A.; Alaskar, A.S.; Al Harthy, S.; Al Subayyil, A.M.; Khatlani,
T.; Alawad, A.O.; et al. Phenotypic and Functional Characterization of Mesenchymal Stem/Multipotent Stromal Cells from
Decidua Basalis of Human Term Placenta. Stem Cells Int. 2016,23, 1193–1207. [CrossRef]
194.
Portmann-Lanz, C.B.; Schoeberlein, A.; Huber, A.; Sager, R.; Malek, A.; Holzgreve, W.; Surbek, D.V. Placental mesenchymal stem
cells as potential autologous graft for pre- and perinatal neuroregeneration. Am. J. Obstet. Gynecol.
2006
,194, 664–673. [CrossRef]
195.
Sudo, K.; Kanno, M.; Miharada, K.; Ogawa, S.; Hiroyama, T.; Saijo, K.; Nakamura, Y. Mesenchymal Progenitors Able to
Differentiate into Osteogenic, Chondrogenic, and/or Adipogenic Cells In Vitro Are Present in Most Primary Fibroblast-Like Cell
Populations. Stem Cells 2007,25, 1610–1617. [CrossRef] [PubMed]
196.
Wolbank, S.; Peterbauer, A.; Fahrner, M.; Hennerbichler, S.; van Griensven, M.; Stadler, G.; Redl, H.; Gabriel, C. Dose-Dependent
Immunomodulatory Effect of Human Stem Cells from Amniotic Membrane: A Comparison with Human Mesenchymal Stem
Cells from Adipose Tissue. Tissue Eng. 2007,13, 1173–1183. [CrossRef]
197.
Lindenmair, A.; Hatlapatka, T.; Kollwig, G.; Hennerbichler, S.; Gabriel, C.; Wolbank, S.; Redl, H.; Kasper, C. Mesenchymal Stem
or Stromal Cells from Amnion and Umbilical Cord Tissue and Their Potential for Clinical Applications. Cells
2012
,1, 1061–1088.
[CrossRef] [PubMed]
198.
Kang, J.W.; Koo, H.C.; Hwang, S.Y.; Kang, S.K.; Ra, J.C.; Lee, M.H.; Park, Y.H. Immunomodulatory effects of human amniotic
membrane-derived mesenchymal stem cells. J. Veter-Sci. 2012,13, 23–31. [CrossRef] [PubMed]
199.
Bilic, G.; Zeisberger, S.M.; Mallik, A.S.; Zimmermann, R.; Zisch, A.H. Comparative Characterization of Cultured Human Term
Amnion Epithelial and Mesenchymal Stromal Cells for Application in Cell Therapy. Cell Transplant.
2008
,17, 955–968. [CrossRef]
[PubMed]
200.
Kim, J.; Kang, H.M.; Kim, H.; Kim, M.R.; Kwon, H.C.; Gye, M.C.; Kang, S.G.; Yang, H.S.; You, J. Ex Vivo Characteristics of Human
Amniotic Membrane-Derived Stem Cells. Cloning Stem Cells 2007,9, 581–594. [CrossRef] [PubMed]
201.
Alviano, F.; Fossati, V.; Marchionni, C.; Arpinati, M.; Bonsi, L.; Franchina, M.; Lanzoni, G.; Cantoni, S.; Cavallini, C.; Bianchi,
F.; et al. Term amniotic membrane is a high throughput source for multipotent mesenchymal stem cells with the ability to
differentiate into endothelial cells in vitro. BMC Dev. Biol. 2007,7, 11. [CrossRef] [PubMed]
202.
Soncini, M.; Vertua, E.; Gibelli, L.; Zorzi, F.; Denegri, M.; Albertini, A.; Wengler, G.S.; Parolini, O. Isolation and characterization of
mesenchymal cells from human fetal membranes. J. Tissue Eng. Regen. Med. 2007,1, 296–305. [CrossRef] [PubMed]
203.
Huang, Q.; Yang, Y.; Luo, C.; Wen, Y.; Liu, R.; Li, S.; Chen, T.; Sun, H.; Tang, L. An efficient protocol to generate placental chorionic
plate-derived mesenchymal stem cells with superior proliferative and immunomodulatory properties. Stem Cell Res. Ther.
2019
,
10, 301. [CrossRef] [PubMed]
204.
Ma, J.; Wu, J.; Han, L.; Jiang, X.; Yan, L.; Hao, J.; Wang, H. Comparative analysis of mesenchymal stem cells derived from amniotic
membrane, umbilical cord, and chorionic plate under serum-free condition. Stem Cell Res. Ther. 2019,10, 19. [CrossRef]
205.
Kim, M.J.; Shin, K.S.; Jeon, J.H.; Lee, D.R.; Shim, S.H.; Kim, J.K.; Cha, D.-H.; Yoon, T.K.; Kim, G.J. Human chorionic-plate-derived
mesenchymal stem cells and Wharton’s jelly-derived mesenchymal stem cells: A comparative analysis of their potential as
placenta-derived stem cells. Cell Tissue Res. 2011,346, 53–64. [CrossRef] [PubMed]
206.
Majore, I.; Moretti, P.; Hass, R.; Kasper, C. Identification of subpopulations in mesenchymal stem cell-like cultures from human
umbilical cord. Cell Commun. Signal. 2009,7, 6. [CrossRef] [PubMed]
Cells 2021,10, 3278 34 of 39
207.
Karahuseyinoglu, S.; Cinar, O.; Kilic, E.; Kara, F.; Akay, G.G.; Demiralp, D.Ö.; Tukun, D.; Uckan, D.; Can, A. Biology of Stem Cells
in Human Umbilical Cord Stroma: In Situ and In Vitro Surveys. Stem Cells 2007,25, 319–331. [CrossRef]
208.
Moretti, P.; Hatlapatka, T.; Marten, D.; Lavrentieva, A.; Majore, I.; Hass, R.; Kasper, C. Mesenchymal Stromal Cells Derived from
Human Umbilical Cord Tissues: Primitive Cells with Potential for Clinical and Tissue Engineering Applications. Bioreact. Syst.
Tissue Eng. II 2009,123, 29–54. [CrossRef]
209.
He, H.; Nagamura-Inoue, T.; Tsunoda, H.; Yuzawa, M.; Yamamoto, Y.; Yorozu, P.; Agata, H.; Tojo, A. Stage-Specific Embryonic
Antigen 4 in Wharton’s Jelly–Derived Mesenchymal Stem Cells Is Not a Marker for Proliferation and Multipotency. Tissue Eng.
Part A 2014,20, 1314–1324. [CrossRef] [PubMed]
210.
Batsali, A.K.; Kastrinaki, M.-C.; Papadaki, H.A.; Pontikoglou, C. Mesenchymal Stem Cells Derived from Wharton’s Jelly of the
Umbilical Cord: Biological Properties and Emerging Clinical Applications. Curr. Stem Cell Res. Ther.
2013
,8, 144–155. [CrossRef]
211.
Stefa´nska, K.; Bryl, R.; Hutchings, G.; Shibli, J.A.; Dyszkiewicz-Konwi ´nska, M. Human umbilical cord stem cells–the discovery,
history and possible application. Med J. Cell Biol. 2020,8, 78–82. [CrossRef]
212.
Moraghebi, R.; Kirkeby, A.; Chaves, P.; Rönn, R.E.; Sitnicka, E.; Parmar, M.; Larsson, M.; Herbst, A.; Woods, N.-B. Term amniotic
fluid: An unexploited reserve of mesenchymal stromal cells for reprogramming and potential cell therapy applications. Stem Cell
Res. Ther. 2017,8, 190. [CrossRef]
213.
Moorefield, E.C.; McKee, E.E.; Solchaga, L.; Orlando, G.; Yoo, J.J.; Walker, S.; Furth, M.E.; Bishop, C.E. Cloned, CD117 Selected
Human Amniotic Fluid Stem Cells Are Capable of Modulating the Immune Response. PLoS ONE 2011,6, e26535. [CrossRef]
214.
Araújo, A.B.; Salton, G.D.; Furlan, J.M.; Schneider, N.; Angeli, M.H.; Laureano, Á.M.; Silla, L.; Passos, E.P.; Paz, A.H. Comparison
of human mesenchymal stromal cells from four neonatal tissues: Amniotic membrane, chorionic membrane, placental decidua
and umbilical cord. Cytotherapy 2017,19, 577–585. [CrossRef] [PubMed]
215.
Ferreira, M.S.V.; Bienert, M.; Müller, K.; Rath, B.; Goecke, T.; Opländer, C.; Braunschweig, T.; Mela, P.; Brümmendorf, T.H.; Beier,
F.; et al. Comprehensive characterization of chorionic villi-derived mesenchymal stromal cells from human placenta. Stem Cell
Res. Ther. 2018,9, 28. [CrossRef] [PubMed]
216.
Tai, C.; Wang, L.; Xie, Y.; Gao, T.; Huang, F.; Wang, B. Analysis of Key Distinct Biological Characteristics of Human Placenta-
Derived Mesenchymal Stromal Cells and Individual Heterogeneity Attributing to Donors. Cells Tissues Organs
2021
,210, 45–57.
[CrossRef]
217.
Seok, J.; Jun, S.; Lee, J.O.; Kim, G.J. Mitochondrial Dynamics in Placenta-Derived Mesenchymal Stem Cells Regulate the Invasion
Activity of Trophoblast. Int. J. Mol. Sci. 2020,21, 8599. [CrossRef] [PubMed]
218.
Seok, J.; Jun, S.; Cho, J.; Park, S.; Lee, J.O.; Kim, G.J. Human placenta-derived mesenchymal stem cells induce trophoblast invasion
via dynamic effects on mitochondrial function. J. Cell. Physiol. 2021,236, 6678–6690. [CrossRef] [PubMed]
219.
Yang, S.-E.; Ha, C.-W.; Jung, M.; Jin, H.-J.; Lee, M.; Song, H.; Choi, S.; Oh, W.; Yang, Y.-S. Mesenchymal stem/progenitor cells
developed in cultures from UC blood. Cytotherapy 2004,6, 476–486. [CrossRef] [PubMed]
220. de la Torre, P.; Flores, A.I. Current status and future prospects of perinatal stem cells. Genes 2021,12, 6. [CrossRef] [PubMed]
221.
Amati, E.; Sella, S.; Perbellini, O.; Alghisi, A.; Bernardi, M.; Chieregato, K.; Lievore, C.; Peserico, D.; Rigno, M.; Zilio, A.; et al.
Generation of mesenchymal stromal cells from cord blood: Evaluation of
in vitro
quality parameters prior to clinical use. Stem
Cell Res. Ther. 2017,8, 14. [CrossRef] [PubMed]
222.
Yoshioka, S.; Miura, Y.; Iwasa, M.; Fujishiro, A.; Yao, H.; Miura, M.; Fukuoka, M.; Nakagawa, Y.; Yokota, A.; Hirai, H.; et al.
Isolation of mesenchymal stromal/stem cells from small-volume umbilical cord blood units that do not qualify for the banking
system. Int. J. Hematol. 2015,102, 218–229. [CrossRef] [PubMed]
223.
Seifabadi, Z.S.; Rezaei-Tazangi, F.; Azarbarz, N.; Nejad, D.B.; Mohammadiasl, J.; Darabi, H.; Pezhmanlarki-Tork, S. Assessment of
viability of wharton’s jelly mesenchymal stem cells encapsulated in alginate scaffold by WST-8 assay kit. Med J. Cell Biol.
2021
,9,
42–47. [CrossRef]
224.
Borys-Wójcik, S.; Moncrieff, L.; Kempisty, B.; Dyszkiewicz-Konwi´nska, M. Current state of umbilical cord stem cells in humans.
Med J. Cell Biol. 2019,7, 86–89. [CrossRef]
225.
Borys-Wójcik, S.; Józkowiak, M.; Stefa´nska, K.; Knap, S.; Pie ´nkowski, W.; Gutaj, P.; Bruska, M.; Kempisty, B. Historical background
of umbilical stem cell culture. Med J. Cell Biol. 2019,7, 11–14. [CrossRef]
226.
Lim, I.J.; Phan, T.T. Epithelial and Mesenchymal Stem Cells from the Umbilical Cord Lining Membrane. Cell Transplant.
2014
,23,
497–503. [CrossRef]
227.
Wang, H.; Yan, X.; Jiang, Y.; Wang, Z.; Li, Y.; Shao, Q. The human umbilical cord stem cells improve the viability of OA degenerated
chondrocytes. Mol. Med. Rep. 2018,17, 4474–4482. [CrossRef] [PubMed]
228.
Fu, Y.-S.; Shih, Y.-T.; Cheng, Y.-C.; Min, M.-Y. Transformation of human umbilical mesenchymal cells into neurons
in vitro
.
J. Biomed. Sci. 2004,11, 652–660. [CrossRef] [PubMed]
229.
Campard, D.; Lysy, P.A.; Najimi, M.; Sokal, E. Native Umbilical Cord Matrix Stem Cells Express Hepatic Markers and Differentiate
Into Hepatocyte-like Cells. Gastroenterology 2008,134, 833–848. [CrossRef] [PubMed]
230.
Wu, K.H.; Zhou, B.; Lu, S.H.; Feng, B.; Yang, S.G.; Du, W.T.; Gu, D.S.; Han, Z.C.; Liu, Y.L.
In vitro
and
in vivo
differentiation of
human umbilical cord derived stem cells into endothelial cells. J. Cell. Biochem. 2007,100, 608–616. [CrossRef] [PubMed]
231.
Stefa´nska, K.; Bryl, R.; Moncrieff, L.; Pinto, N.; Shibli, J.A.; Dyszkiewicz-Konwi ´nska, M. Mesenchymal stem cells–a historical
overview. Med J. Cell Biol. 2020,8, 83–87. [CrossRef]
Cells 2021,10, 3278 35 of 39
232.
Loukogeorgakis, S.P.; De Coppi, P. Concise Review: Amniotic Fluid Stem Cells: The Known, the Unknown, and Potential
Regenerative Medicine Applications. STEM CELLS 2017,35, 1663–1673. [CrossRef] [PubMed]
233.
Eslaminejad, M.B.; Jahangir, S. Amniotic Fluid Stem Cells and Their Application in Cell-Based Tissue Regeneration. Int. J. Fertil.
Steril. 2012,6, 147–156.
234.
Loukogeorgakis, S.P.; De Coppi, P. Stem cells from amniotic fluid–Potential for regenerative medicine. Best Pract. Res. Clin. Obstet.
Gynaecol. 2016,31, 45–57. [CrossRef]
235.
Bajek, A.; Olkowska, J.; Walentowicz-Sadłecka, M.; Walentowicz, P.; Sadłecki, P.; Grabiec, M.; Bodnar, M.; Marszałek, A.; D˛ebski,
R.; Porowi´nska, D.; et al. High Quality Independent From a Donor: Human Amniotic Fluid Derived Stem Cells-A Practical
Analysis Based on 165 Clinical Cases. J. Cell. Biochem. 2016,118, 116–126. [CrossRef]
236.
Tsai, M.S.; Lee, J.L.; Chang, Y.J.; Hwang, S.M. Isolation of human multipotent mesenchymal stem cells from second-trimester
amniotic fluid using a novel two-stage culture protocol. Hum. Reprod. 2004,19, 1450–1456. [CrossRef] [PubMed]
237.
Laowanitwattana, T.; Aungsuchawan, S.; Narakornsak, S.; Markmee, R.; Tancharoen, W.; Keawdee, J.; Boonma, N.; Tasuya, W.;
Peerapapong, L.; Pangjaidee, N.; et al. Osteoblastic differentiation potential of human amniotic fluid-derived mesenchymal stem
cells in different culture conditions. Acta Histochem. 2018,120, 701–712. [CrossRef]
238.
Di Trapani, M.; Bassi, G.; Fontana, E.; Giacomello, L.; Pozzobon, M.; Guillot, P.V.; De Coppi, P.; Krampera, M. Immune Regulatory
Properties of CD117pos Amniotic Fluid Stem Cells Vary According to Gestational Age. Stem Cells Dev.
2015
,24, 132–143.
[CrossRef] [PubMed]
239.
Wolfrum, K.; Wang, Y.; Prigione, A.; Sperling, K.; Lehrach, H.; Adjaye, J. The LARGE Principle of Cellular Reprogramming: Lost,
Acquired and Retained Gene Expression in Foreskin and Amniotic Fluid-Derived Human iPS Cells. PLoS ONE
2010
,5, e13703.
[CrossRef]
240.
Balbi, C.; Bollini, S. Fetal and perinatal stem cells in cardiac regeneration: Moving forward to the paracrine era. Placenta
2017
,59,
96–106. [CrossRef]
241.
Sibiak, R.; Jaworski, M.; Barrett, S.; Bryl, R.; Gutaj, P.; Kulus, J.; Bukowska, D.; Petitte, J.; Crha, I.; Ventruba, P.; et al. Trophoblast
stem cells-methods of isolation, histological and cellular characteristic, and their possible applications in human and animal
models. Med J. Cell Biol. 2020,8, 95–100. [CrossRef]
242.
Bischof, P.; Irminger-Finger, I. The human cytotrophoblastic cell, a mononuclear chameleon. Int. J. Biochem. Cell Biol.
2005
,37,
1–16. [CrossRef] [PubMed]
243.
Gerami-Naini, B.; Dovzhenko, O.V.; Durning, M.; Wegner, F.H.; Thomson, J.A.; Golos, T.G. Trophoblast Differentiation in
Embryoid Bodies Derived from Human Embryonic Stem Cells. Endocrinol. 2004,145, 1517–1524. [CrossRef]
244.
Xu, R.-H.; Chen, X.; Li, D.S.; Li, R.; Addicks, G.C.; Glennon, C.; Zwaka, T.P.; Thomson, J.A. BMP4 initiates human embryonic stem
cell differentiation to trophoblast. Nat. Biotechnol. 2002,20, 1261–1264. [CrossRef]
245.
Genbacev, O.; Miller, R. Post-implantation Differentiation and Proliferation of Cytotrophoblast Cells: In Vitro Models—A Review.
Placenta 2000,21, S45–S49. [CrossRef] [PubMed]
246.
Caniggia, I.; Mostachfi, H.; Winter, J.; Gassmann, M.; Lye, S.J.; Kuliszewski, M.; Post, M. Hypoxia-inducible factor-1 mediates the
biological effects of oxygen on human trophoblast differentiation through TGF
β
3. J. Clin. Investig.
2000
,105, 577–587. [CrossRef]
[PubMed]
247.
Frendo, J.-L.; Olivier, D.; Cheynet, V.; Blond, J.-L.; Bouton, O.; Vidaud, M.; Rabreau, M.; Evain-Brion, D.; Mallet, F. Direct
Involvement of HERV-W Env Glycoprotein in Human Trophoblast Cell Fusion and Differentiation. Mol. Cell. Biol.
2003
,23,
3566–3574. [CrossRef] [PubMed]
248.
Frendo, J.-L.; Cronier, L.; Bertin, G.; Guibourdenche, J.; Vidaud, M.; Evain-Brion, D.; Malassiné, A. Involvement of connexin 43 in
human trophoblast cell fusion and differentiation. J. Cell Sci. 2003,116, 3413–3421. [CrossRef] [PubMed]
249.
Okae, H.; Toh, H.; Sato, T.; Hiura, H.; Takahashi, S.; Shirane, K.; Kabayama, Y.; Suyama, M.; Sasaki, H.; Arima, T. Derivation of
Human Trophoblast Stem Cells. Cell Stem Cell 2018,22, 50–63.e6. [CrossRef] [PubMed]
250.
Latos, P.A.; Hemberger, M. From the stem of the placental tree: Trophoblast stem cells and their progeny. Development
2016
,143,
3650–3660. [CrossRef] [PubMed]
251.
Gauthier-Fisher, A.; Szaraz, P.; Librach, C.L. Pericytes in the Umbilical Cord. In Advances in Experimental Medicine and Biology;
Springer: Berlin/Heidelberg, Germany, 2019; Volume 1122, pp. 211–233.
252.
Subramanian, A.; Fong, C.-Y.; Biswas, A.; Bongso, A. Comparative Characterization of Cells from the Various Compartments
of the Human Umbilical Cord Shows that the Wharton’s Jelly Compartment Provides the Best Source of Clinically Utilizable
Mesenchymal Stem Cells. PLoS ONE 2015,10, e0127992. [CrossRef]
253.
Takechi, K.; Kuwabara, Y.; Mizuno, M. Ultrastructural and immunohistochemical studies of Wharton’s jelly umbilical cord cells.
Placenta 1993,14, 235–245. [CrossRef]
254.
Schugar, R.C.; Chirieleison, S.M.; Wescoe, K.E.; Schmidt, B.T.; Askew, Y.; Nance, J.J.; Evron, J.M.; Péault, B.; Deasy, B.M. High
Harvest Yield, High Expansion, and Phenotype Stability of CD146 Mesenchymal Stromal Cells from Whole Primitive Human
Umbilical Cord Tissue. J. Biomed. Biotechnol. 2009,2009, 1–11. [CrossRef] [PubMed]
255.
Crisan, M.; Yap, S.; Casteilla, L.; Chen, C.-W.; Corselli, M.; Park, T.S.; Andriolo, G.; Sun, B.; Zheng, B.; Zhang, L.; et al. A
Perivascular Origin for Mesenchymal Stem Cells in Multiple Human Organs. Cell Stem Cell
2008
,3, 301–313. [CrossRef] [PubMed]
256.
Sarugaser, R.; Lickorish, D.; Baksh, D.; Hosseini, M.M.; Davies, J.E. Human Umbilical Cord Perivascular (HUCPV) Cells: A
Source of Mesenchymal Progenitors. Stem Cells 2005,23, 220–229. [CrossRef]
Cells 2021,10, 3278 36 of 39
257.
Stratman, A.N.; Schwindt, A.E.; Malotte, K.M.; Davis, G.E. Endothelial-derived PDGF-BB and HB-EGF coordinately regulate
pericyte recruitment during vasculogenic tube assembly and stabilization. Blood 2010,116, 4720–4730. [CrossRef] [PubMed]
258.
Grzywocz, Z.; Pius-Sadowska, E.; Klos, P.; Gryzik, M.; Wasilewska, D.; Aleksandrowicz, B.; Dworczynska, M.; Sabalinska, S.;
Hoser, G.; Machalinski, B.; et al. Growth factors and their receptors derived from human amniotic cells
in vitro
.Folia Histochem.
Et Cytobiol. 2014,52, 163–170. [CrossRef]
259.
Takashima, Y.; Era, T.; Nakao, K.; Kondo, S.; Kasuga, M.; Smith, A.; Nishikawa, S.-I. Neuroepithelial Cells Supply an Initial
Transient Wave of MSC Differentiation. Cell 2007,129, 1377–1388. [CrossRef] [PubMed]
260.
Gonzalez, R.; Griparic, L.; Umana, M.; Burgee, K.; Vargas, V.; Nasrallah, R.; Silva, F.; Patel, A. An Efficient Approach to Isolation
and Characterization of Pre- and Postnatal Umbilical Cord Lining Stem Cells for Clinical Applications. Cell Transplant.
2010
,19,
1439–1449. [CrossRef] [PubMed]
261.
Dobreva, M.; Pereira, P.N.G.; Deprest, J.; Zwijsen, A. On the origin of amniotic stem cells: Of mice and men. Int. J. Dev. Biol.
2010
,
54, 761–777. [CrossRef]
262. Fauza, D. Amniotic fluid and placental stem cells. Best Pract. Res. Clin. Obstet. Gynaecol. 2004,18, 877–891. [CrossRef]
263.
Tong, X. Amniotic fluid may act as a transporting pathway for signaling molecules and stem cells during the embryonic
development of amniotes. J. Chin. Med Assoc. 2013,76, 606–610. [CrossRef] [PubMed]
264.
Torricelli, F.; Brizzi, L.; A Bernabei, P.; Gheri, G.; Di Lollo, S.; Nutini, L.; Lisi, E.; Di Tommaso, M.; Cariati, E. Identification of
hematopoietic progenitor cells in human amniotic fluid before the 12th week of gestation. Ital. J. Anat. Embryol. Arch. Ital. Di
Anat. Ed Embriol. 1993,98, 264.
265.
In ’t Anker, P.S.; Scherjon, S.A.; Kleijburg-van der Keur, C.; Noort, W.A.; Claas, F.H.J.; Willemze, R.; Fibbe, W.E.; Kanhai, H.H.H.
Amniotic fluid as a novel source of mesenchymal stem cells for therapeutic transplantation. Blood
2003
,102, 1548–1549. [CrossRef]
266.
Kolambkar, Y.M.; Peister, A.; Soker, S.; Atala, A.; Guldberg, R.E. Chondrogenic differentiation of amniotic fluid-derived stem cells.
J. Mol. Histol. 2007,38, 405–413. [CrossRef]
267.
Bajek, A.; Olkowska, J.; Walentowicz-Sadłecka, M.; Sadłecki, P.; Grabiec, M.; Porowinska, D.; Drewa, T.; Roszkowski, K. Human
Adipose-Derived and Amniotic Fluid-Derived Stem Cells: A Preliminary In Vitro Study Comparing Myogenic Differentiation
Capability. Med Sci. Monit. 2018,24, 1733–1741. [CrossRef]
268.
Rahman, S.; Spitzhorn, L.-S.; Wruck, W.; Hagenbeck, C.; Balan, P.; Graffmann, N.; Bohndorf, M.; Ncube, A.; Guillot, P.V.; Fehm,
T.; et al. The presence of human mesenchymal stem cells of renal origin in amniotic fluid increases with gestational time. Stem
Cell Res. Ther. 2018,9, 1–17. [CrossRef]
269.
Hengstschläger, M.; Siegel, N.; Valli, A.; Fuchs, C.; Rosner, M. Expression of mTOR pathway proteins in human amniotic fluid
stem cells. Int. J. Mol. Med. 2009,23, 779–784. [CrossRef] [PubMed]
270.
Valli, A.; Rosner, M.; Fuchs, C.; Siegel, N.; E Bishop, C.; Dolznig, H.; Mädel, U.; Feichtinger, W.; Atala, A.; Hengstschläger,
M. Embryoid body formation of human amniotic fluid stem cells depends on mTOR. Oncogene
2010
,29, 966–977. [CrossRef]
[PubMed]
271.
Barboni, B.; Russo, V.; Berardinelli, P.; Mauro, A.; Valbonetti, L.; Sanyal, H.; Canciello, A.; Greco, L.; Muttini, A.; Gatta, V.; et al.
Placental Stem Cells from Domestic Animals: Translational Potential and Clinical Relevance. Cell Transplant.
2018
,27, 93–116.
[CrossRef]
272.
Kusuma, G.D.; Menicanin, D.; Gronthos, S.; Manuelpillai, U.; Abumaree, M.H.; Pertile, M.D.; Brennecke, S.P.; Kalionis, B. Ectopic
Bone Formation by Mesenchymal Stem Cells Derived from Human Term Placenta and the Decidua. PLoS ONE
2015
,10, e0141246.
[CrossRef] [PubMed]
273.
Reddy, S.; Wasnik, S.; Guha, A.; Kumar, J.M.; Sinha, A.; Singh, S. Evaluation of nano-biphasic calcium phosphate ceramics for
bone tissue engineering applications: In vitro and preliminary in vivo studies. J. Biomater. Appl. 2013,27, 565–575. [CrossRef]
274.
Wei, J.P.; Nawata, M.; Wakitani, S.; Kametani, K.; Ota, M.; Toda, A.; Konishi, I.; Ebara, S.; Nikaido, T. Human Amniotic
Mesenchymal Cells Differentiate into Chondrocytes. Cloning Stem Cells 2009,11, 19–26. [CrossRef]
275.
Li, F.; Chen, Y.-Z.; Miao, Z.-N.; Zheng, S.-Y.; Jin, J. Human Placenta-Derived Mesenchymal Stem Cells with Silk Fibroin Biomaterial
in the Repair of Articular Cartilage Defects. Cell. Reprogramming 2012,14, 334–341. [CrossRef] [PubMed]
276.
Nogami, M.; Tsuno, H.; Koike, C.; Okabe, M.; Yoshida, T.; Seki, S.; Matsui, Y.; Kimura, T.; Nikaido, T. Isolation and Characterization
of Human Amniotic Mesenchymal Stem Cells and Their Chondrogenic Differentiation. Transplantation
2012
,93, 1221–1228.
[CrossRef] [PubMed]
277.
Zhao, P.; Ise, H.; Hongo, M.; Ota, M.; Konishi, I.; Nikaido, T. Human Amniotic Mesenchymal Cells Have Some Characteristics of
Cardiomyocytes. Transplant. 2005,79, 528–535. [CrossRef]
278.
Zhang, W.; Liu, X.-C.; Yang, L.; Zhu, D.-L.; Zhang, Y.-D.; Chen, Y.; Zhang, H.-Y. Wharton’s jelly-derived mesenchymal stem cells
promote myocardial regeneration and cardiac repair after miniswine acute myocardial infarction. Coron. Artery Dis.
2013
,24,
549–558. [CrossRef] [PubMed]
279.
Nascimento, D.S.; Mosqueira, D.; Sousa, L.M.; Teixeira, M.; Filipe, M.; Resende, T.P.; Araújo, A.F.; Valente, M.; Almeida, J.;
Martins, J.P.; et al. Human umbilical cord tissue-derived mesenchymal stromal cells attenuate remodeling after myocardial
infarction by proangiogenic, antiapoptotic, and endogenous cell-activation mechanisms. Stem Cell Res. Ther.
2014
,5, 5. [CrossRef]
280.
Simioniuc, A.; Campan, M.; Lionetti, V.; Marinelli, M.; Aquaro, G.D.; Cavallini, C.; Valente, S.; Di Silvestre, D.; Cantoni, S.; Bernini,
F.; et al. Placental stem cells pre-treated with a hyaluronan mixed ester of butyric and retinoic acid to cure infarcted pig hearts: A
multimodal study. Cardiovasc. Res. 2011,90, 546–556. [CrossRef]
Cells 2021,10, 3278 37 of 39
281.
Weiss, M.L.; Medicetty, S.; Bledsoe, A.R.; Rachakatla, R.S.; Choi, M.; Merchav, S.; Luo, Y.; Rao, M.S.; Velagaleti, G.; Troyer, D.
Human Umbilical Cord Matrix Stem Cells: Preliminary Characterization and Effect of Transplantation in a Rodent Model of
Parkinson’s Disease. Stem Cells 2006,24, 781–792. [CrossRef]
282.
Fu, Y.-S.; Cheng, Y.-C.; Lin, M.-Y.A.; Cheng, H.; Chu, P.-M.; Chou, S.-C.; Shih, Y.-H.; Ko, M.-H.; Sung, M.-S. Conversion of Human
Umbilical Cord Mesenchymal Stem Cells in Wharton’s Jelly to Dopaminergic Neurons In Vitro: Potential Therapeutic Application
for Parkinsonism. Stem Cells 2006,24, 115–124. [CrossRef] [PubMed]
283.
Park, S.; Kim, E.; Koh, S.-E.; Maeng, S.; Lee, W.-D.; Lim, J.; Shim, I.; Lee, Y.-J. Dopaminergic differentiation of neural progenitors
derived from placental mesenchymal stem cells in the brains of Parkinson’s disease model rats and alleviation of asymmetric
rotational behavior. Brain Res. 2012,1466, 158–166. [CrossRef] [PubMed]
284.
Kim, K.-S.; Kim, H.S.; Park, J.-M.; Kim, H.W.; Park, M.-K.; Lee, H.-S.; Lim, D.S.; Lee, T.H.; Chopp, M.; Moon, J. Long-term
immunomodulatory effect of amniotic stem cells in an Alzheimer’s disease model. Neurobiol. Aging
2013
,34, 2408–2420.
[CrossRef]
285.
Mohamadi, Y.; Moghahi, S.M.H.N.; Mousavi, M.; Borhani-Haghighi, M.; Abolhassani, F.; Kashani, I.R.; Hassanzadeh, G.
Intrathecal transplantation of Wharton’s jelly mesenchymal stem cells suppresses the NLRP1 inflammasome in the rat model of
spinal cord injury. J. Chem. Neuroanat. 2019,97, 1–8. [CrossRef] [PubMed]
286.
Badner, A.; Hacker, J.; Hong, J.; Mikhail, M.; Vawda, R.; Fehlings, M.G. Splenic involvement in umbilical cord matrix-derived
mesenchymal stromal cell-mediated effects following traumatic spinal cord injury. J. Neuroinflammation
2018
,15, 219. [CrossRef]
287.
Chudickova, M.; Vackova, I.; Urdzikova, L.M.; Jancova, P.; Kekulova, K.; Rehorova, M.; Turnovcova, K.; Jendelova, P.; Kubinova,
S. The Effect of Wharton Jelly-Derived Mesenchymal Stromal Cells and Their Conditioned Media in the Treatment of a Rat Spinal
Cord Injury. Int. J. Mol. Sci. 2019,20, 4516. [CrossRef]
288.
Krupa, P.; Vackova, I.; Ruzicka, J.; Zaviskova, K.; Dubisova, J.; Koci, Z.; Turnovcova, K.; Urdzikova, L.M.; Kubinova, S.; Rehak,
S.; et al. The Effect of Human Mesenchymal Stem Cells Derived from Wharton’s Jelly in Spinal Cord Injury Treatment Is
Dose-Dependent and Can Be Facilitated by Repeated Application. Int. J. Mol. Sci. 2018,19, 1503. [CrossRef] [PubMed]
289.
Clark, K.; Zhang, S.; Barthe, S.; Kumar, P.; Pivetti, C.; Kreutzberg, N.; Reed, C.; Wang, Y.; Paxton, Z.; Farmer, D.; et al. Placental
Mesenchymal Stem Cell-Derived Extracellular Vesicles Promote Myelin Regeneration in an Animal Model of Multiple Sclerosis.
Cells 2019,8, 1497. [CrossRef]
290.
Fisher-Shoval, Y.; Barhum, Y.; Sadan, O.; Yust-Katz, S.; Ben-Zur, T.; Lev, N.; Benkler, C.; Hod, M.; Melamed, E.; Offen, D.
Transplantation of Placenta-Derived Mesenchymal Stem Cells in the EAE Mouse Model of MS. J. Mol. Neurosci.
2012
,48, 176–184.
[CrossRef] [PubMed]
291.
Tsai, P.-C.; Fu, T.-W.; Chen, Y.-M.A.; Ko, T.-L.; Chen, T.-H.; Shih, Y.-H.; Hung, S.-C.; Fu, Y.-S. The therapeutic potential of human
umbilical mesenchymal stem cells from Wharton’s jelly in the treatment of rat liver fibrosis. Liver Transplant.
2009
,15, 484–495.
[CrossRef] [PubMed]
292.
Moodley, Y.; Atienza, D.; Manuelpillai, U.; Samuel, C.S.; Tchongue, J.; Ilancheran, S.; Boyd, R.; Trounson, A. Human Umbilical
Cord Mesenchymal Stem Cells Reduce Fibrosis of Bleomycin-Induced Lung Injury. Am. J. Pathol.
2009
,175, 303–313. [CrossRef]
293.
Yu, J.; Hao, G.; Wang, D.; Liu, J.; Dong, X.; Sun, Y.; Pan, Q.; Li, Y.; Shi, X.; Li, L.; et al. Therapeutic Effect and Location of
GFP-Labeled Placental Mesenchymal Stem Cells on Hepatic Fibrosis in Rats. Stem Cells Int. 2017,2017, 1–11. [CrossRef]
294.
Jung, J.; Choi, J.H.; Lee, E.; Park, J.-W.; Oh, I.-H.; Hwang, S.-G.; Kim, K.-S.; Kim, G.J. Human Placenta-Derived Mesenchymal
Stem Cells Promote Hepatic Regeneration in CCl4-Injured Rat Liver Model via Increased Autophagic Mechanism. Stem Cells
2013,31, 1584–1596. [CrossRef]
295.
Zhang, D.; Jiang, M.; Miao, D. Transplanted Human Amniotic Membrane-Derived Mesenchymal Stem Cells Ameliorate Carbon
Tetrachloride-Induced Liver Cirrhosis in Mouse. PLoS ONE 2011,6, e16789. [CrossRef]
296.
Cao, H.; Yang, J.; Yu, J.; Pan, Q.; Li, J.; Zhou, P.; Li, Y.; Pan, X.; Li, J.; Wang, Y.; et al. Therapeutic potential of transplanted placental
mesenchymal stem cells in treating Chinese miniature pigs with acute liver failure. BMC Med. 2012,10, 56. [CrossRef]
297.
Kim, H.; Shin, T.; Lee, B.; Yu, K.; Seo, Y.; Lee, S.; Seo, M.; Hong, I.; Choi, S.W.; Seo, K.; et al. Human Umbilical Cord Blood
Mesenchymal Stem Cells Reduce Colitis in Mice by Activating NOD2 Signaling to COX2. Gastroenterol.
2013
,145, 1392–1403.e8.
[CrossRef]
298.
Song, J.-Y.; Kang, H.J.; Hong, J.S.; Kim, C.J.; Shim, J.-Y.; Lee, C.W.; Choi, J. Umbilical cord-derived mesenchymal stem cell extracts
reduce colitis in mice by re-polarizing intestinal macrophages. Sci. Rep. 2017,7, 9412. [CrossRef] [PubMed]
299.
Kim, B.S.; Chun, S.Y.; Kil Lee, J.; Lim, H.J.; Bae, J.-S.; Chung, H.-Y.; Atala, A.; Soker, S.; Yoo, J.J.; Kwon, T.G. Human amniotic fluid
stem cell injection therapy for urethral sphincter regeneration in an animal model. BMC Med.
2012
,10, 94. [CrossRef] [PubMed]
300.
Sabapathy, V.; Sundaram, B.; VM, S.; Mankuzhy, P.; Kumar, S. Human Wharton’s Jelly Mesenchymal Stem Cells plasticity
augments scar-free skin wound healing with hair growth. PLoS ONE 2014,9, e93726. [CrossRef]
301.
Khalifeh Soltani, S.; Forogh, B.; Ahmadbeigi, N.; Hadizadeh Kharazi, H.; Fallahzadeh, K.; Kashani, L.; Karami, M.; Kheyrollah,
Y.; Vasei, M. Safety and efficacy of allogenic placental mesenchymal stem cells for treating knee osteoarthritis: A pilot study.
Cytotherapy 2019,21. [CrossRef]
302.
Winkler, T.; Perka, C.; von Roth, P.; Agres, A.N.; Plage, H.; Preininger, B.; Pumberger, M.; Geissler, S.; Hagai, E.L.; Ofir, R.; et al.
Immunomodulatory placental-expanded, mesenchymal stromal cells improve muscle function following hip arthroplasty.
J. Cachexia Sarcopenia Muscle 2018,9, 880–897. [CrossRef] [PubMed]
Cells 2021,10, 3278 38 of 39
303.
Norgren, L.; Weiss, N.; Nikol, S.; Hinchliffe, R.J.; Lantis, J.C.; Patel, M.R.; Reinecke, H.; Ofir, R.; Rosen, Y.; Peres, D.; et al. PLX-PAD
Cell Treatment of Critical Limb Ischaemia: Rationale and Design of the PACE Trial. Eur. J. Vasc. Endovasc. Surg.
2019
,57.
[CrossRef] [PubMed]
304.
Levy, J.A.; Marchand, M.; Iorio, L.; Cassini, W.; Zahalsky, M.P. Determining the Feasibility of Managing Erectile Dysfunction in
Humans With Placental-Derived Stem Cells. J. Osteopat. Med. 2016,116, e1–e5. [CrossRef] [PubMed]
305.
Zeng, X.; Tang, Y.; Hu, K.; Jiao, W.; Ying, L.; Zhu, L.; Liu, J.; Xu, J. Three-week topical treatment with placenta-derived
mesenchymal stem cells hydrogel in a patient with diabetic foot ulcer: A case report. Medcine 2017,96, e9212. [CrossRef]
306.
Harrell, C.R.; Miloradovic, D.; Sadikot, R.; Fellabaum, C.; Markovic, B.S.; Miloradovic, D.; Acovic, A.; Djonov, V.; Arsenijevic, N.;
Volarevic, V. Molecular and Cellular Mechanisms Responsible for Beneficial Effects of Mesenchymal Stem Cell-Derived Product
“Exo-d-MAPPS” in Attenuation of Chronic Airway Inflammation. Anal. Cell. Pathol. 2020,2020, 1–15. [CrossRef] [PubMed]
307.
Hashemian, S.-M.R.; Aliannejad, R.; Zarrabi, M.; Soleimani, M.; Vosough, M.; Hosseini, S.-E.; Hossieni, H.; Keshel, S.H.;
Naderpour, Z.; Hajizadeh-Saffar, E.; et al. Mesenchymal stem cells derived from perinatal tissues for treatment of critically ill
COVID-19-induced ARDS patients: A case series. Stem Cell Res. Ther. 2021,12, 1–12. [CrossRef] [PubMed]
308.
Ringden, O.; Baygan, A.; Remberger, M.; Gustafsson, B.; Winiarski, J.; Khoein, B.; Moll, G.; Klingspor, L.; Westgren, M.; Sadeghi,
B. Placenta-Derived Decidua Stromal Cells for Treatment of Severe Acute Graft-Versus-Host Disease. Stem Cells Transl. Med.
2018
,
7, 325–331. [CrossRef] [PubMed]
309.
Dilogo, I.H.; Canintika, A.F.; Hanitya, A.L.; Pawitan, J.A.; Liem, I.K.; Pandelaki, J. Umbilical cord-derived mesenchymal stem
cells for treating osteoarthritis of the knee: A single-arm, open-label study. Eur. J. Orthop. Surg. Traumatol.
2020
,30, 799–807.
[CrossRef] [PubMed]
310.
Matas, J.; Orrego, M.; Amenabar, D.; Infante, C.; Tapia-Limonchi, R.; Cadiz, M.I.; Alcayaga-Miranda, F.; González, P.L.; Muse, E.;
Khoury, M.; et al. Umbilical Cord-Derived Mesenchymal Stromal Cells (MSCs) for Knee Osteoarthritis: Repeated MSC Dosing Is
Superior to a Single MSC Dose and to Hyaluronic Acid in a Controlled Randomized Phase I/II Trial. Stem Cells Transl. Med.
2019
,
8, 215–224. [CrossRef] [PubMed]
311.
Park, Y.-B.; Ha, C.-W.; Lee, C.-H.; Yoon, Y.C. Cartilage Regeneration in Osteoarthritic Patients by a Composite of Allogeneic
Umbilical Cord Blood-Derived Mesenchymal Stem Cells and Hyaluronate Hydrogel: Results from a Clinical Trial for Safety and
Proof-of-Concept with 7 Years of Extended Follow-Up. Stem Cells Transl. Med. 2016,6, 613–621. [CrossRef]
312.
Wang, Y.; Jin, W.; Liu, H.; Cui, Y.; Mao, Q.; Fei, Z.; Xiang, C. Curative effect of human umbilical cord mesenchymal stem cells
by intra-articular injection for degenerative knee osteoarthritis. Zhongguo Xiu Fu Chong Jian Wai Ke Za Zhi = Zhongguo Xiufu
Chongjian Waike Zazhi = Chin. J. Reparative Reconstr. Surg. 2016,30, 1472–1477. [CrossRef]
313.
He, X.; Wang, Q.; Zhao, Y.; Zhang, H.; Wang, B.; Pan, J.; Li, J.; Yu, H.; Wang, L.; Dai, J.; et al. Effect of Intramyocardial Grafting
Collagen Scaffold With Mesenchymal Stromal Cells in Patients with Chronic Ischemic Heart Disease: A Randomized Clinical
Trial. JAMA Netw. Open 2020,3, e2016236. [CrossRef] [PubMed]
314.
Bartolucci, J.; Verdugo, F.J.; González, P.L.; Larrea, R.E.; Abarzua, E.; Goset, C.; Rojo, P.; Palma, I.; Lamich, R.; Pedreros, P.A.; et al.
Safety and efficacy of the intravenous infusion of umbilical cord mesenchymal stem cells in patients with heart failure: A phase
1/2 randomized controlled trial (RIMECARD trial [Randomized clinical trial of intravenous infusion umbilical cord mesenchymal.
Circ. Res. 2017,121, 1192–1204. [CrossRef] [PubMed]
315.
Hashemi, S.; Mohammadi, A.A.; Kabiri, H.; Hashempoor, M.R.; Mahmoodi, M.; Amini, M.; Mehrabani, D.; Mahmudi, M. The
healing effect of Wharton’s jelly stem cells seeded on biological scaffold in chronic skin ulcers: A randomized clinical trial.
J. Cosmet. Dermatol. 2019,18, 1961–1967. [CrossRef]
316.
Kim, Y.J.; Ahn, H.J.; Lee, S.H.; Lee, M.H.; Kang, K.S. Effects of conditioned media from human umbilical cord blood-derived
mesenchymal stem cells in the skin immune response. Biomed. Pharmacother. 2020,131, 110789. [CrossRef] [PubMed]
317.
Kim, J.; Kim, B.; Kim, S.; Lee, Y.I.; Kim, J.; Lee, J.H. The effect of human umbilical cord blood–derived mesenchymal stem cell
media containing serum on recovery after laser treatment: A double-blinded, randomized, split-face controlled study. J. Cosmet.
Dermatol. 2020,19, 651–656. [CrossRef]
318.
He, X.; Yang, Y.; Yao, M.; Yang, L.; Ao, L.; Hu, X.; Li, Z.; Wu, X.; Tan, Y.; Xing, W.; et al. Combination of human umbilical cord
mesenchymal stem (stromal) cell transplantation with IFN-
γ
treatment synergistically improves the clinical outcomes of patients
with rheumatoid arthritis. Ann. Rheum. Dis. 2020,79, 1298–1304. [CrossRef] [PubMed]
319.
Qi, T.; Gao, H.; Dang, Y.; Huang, S.; Peng, M. Cervus and cucumis peptides combined umbilical cord mesenchymal stem cells
therapy for rheumatoid arthritis. Medicine 2020,99, e21222. [CrossRef] [PubMed]
320.
Wang, L.; Huang, S.; Li, S.; Li, M.; Shi, J.; Bai, W.; Wang, Q.; Zheng, L.; Liu, Y. Efficacy and Safety of Umbilical Cord Mesenchymal
Stem Cell Therapy for Rheumatoid Arthritis Patients: A Prospective Phase I/II Study. Drug Des. Dev. Ther.
2019
,13, 4331–4340.
[CrossRef] [PubMed]
321.
Park, E.H.; Lim, H.; Lee, S.; Roh, K.; Seo, K.; Kang, K.; Shin, K. Intravenous Infusion of Umbilical Cord Blood-Derived
Mesenchymal Stem Cells in Rheumatoid Arthritis: A Phase Ia Clinical Trial. Stem Cells Transl. Med. 2018,7, 636–642. [CrossRef]
[PubMed]
322.
Deng, D.; Zhang, P.; Guo, Y.; Lim, T.O. A randomised double-blind, placebo-controlled trial of allogeneic umbilical cord-derived
mesenchymal stem cell for lupus nephritis. Ann. Rheum. Dis. 2017,76, 1436–1439. [CrossRef]
Cells 2021,10, 3278 39 of 39
323.
Riordan, N.H.; Morales, I.; Fernández, G.; Allen, N.; Fearnot, N.E.; Leckrone, M.E.; Markovich, D.J.; Mansfield, D.; Avila, D.; Patel,
A.N.; et al. Clinical feasibility of umbilical cord tissue-derived mesenchymal stem cells in the treatment of multiple sclerosis. J.
Transl. Med. 2018,16, 57. [CrossRef]
324.
Gu, J.; Huang, L.; Zhang, C.; Wang, Y.; Zhang, R.; Tu, Z.; Wang, H.; Zhou, X.; Xiao, Z.; Liu, Z.; et al. Therapeutic evidence of
umbilical cord-derived mesenchymal stem cell transplantation for cerebral palsy: A randomized, controlled trial. Stem Cell Res.
Ther. 2020,11, 43. [CrossRef]
325.
Riordan, N.H.; Hincapié, M.L.; Morales, I.; Fernández, G.; Allen, N.; Leu, C.; Madrigal, M.; Paz Rodríguez, J.; Novarro, N.
Allogeneic Human Umbilical Cord Mesenchymal Stem Cells for the Treatment of Autism Spectrum Disorder in Children: Safety
Profile and Effect on Cytokine Levels. Stem Cells Transl. Med. 2019,8, 1008–1016. [CrossRef]
326.
Zhang, J.; Lv, S.; Liu, X.; Song, B.; Shi, L. Umbilical Cord Mesenchymal Stem Cell Treatment for Crohn’s Disease: A Randomized
Controlled Clinical Trial. Gut Liver 2018,12, 73–78. [CrossRef] [PubMed]
327.
Sun, Q.; Huang, Z.; Han, F.; Zhao, M.; Cao, R.; Zhao, D.; Hong, L.; Na, N.; Li, H.; Miao, B.; et al. Allogeneic mesenchymal stem
cells as induction therapy are safe and feasible in renal allografts: Pilot results of a multicenter randomized controlled trial. J.
Transl. Med. 2018,16, 1–10. [CrossRef] [PubMed]
328.
Shi, M.; Liu, Z.; Wang, Y.; Xu, R.; Sun, Y.; Zhang, M.; Yu, X.; Wang, H.; Meng, L.; Su, H.; et al. A Pilot Study of Mesenchymal Stem
Cell Therapy for Acute Liver Allograft Rejection. Stem Cells Transl. Med. 2017,6, 2053–2061. [CrossRef]
329.
Shi, L.; Huang, H.; Lu, X.; Yan, X.; Jiang, X.; Xu, R.; Wang, S.; Zhang, C.; Yuan, X.; Xu, Z.; et al. Effect of human umbilical cord-
derived mesenchymal stem cells on lung damage in severe COVID-19 patients: A randomized, double-blind, placebo-controlled
phase 2 trial. Signal Transduct. Target. Ther. 2021,6, 1–9. [CrossRef]
330.
Meng, F.; Xu, R.; Wang, S.; Xu, Z.; Zhang, C.; Li, Y.; Yang, T.; Shi, L.; Fu, J.; Jiang, T.; et al. Human umbilical cord-derived
mesenchymal stem cell therapy in patients with COVID-19: A phase 1 clinical trial. Signal Transduct. Target. Ther.
2020
,5, 172.
[CrossRef] [PubMed]
331.
Shu, L.; Niu, C.; Li, R.; Huang, T.; Wang, Y.; Huang, M.; Ji, N.; Zheng, Y.; Chen, X.; Shi, L.; et al. Treatment of severe COVID-19
with human umbilical cord mesenchymal stem cells. Stem Cell Res. Ther. 2020,11, 361. [CrossRef] [PubMed]
332.
He, X.; Ai, S.; Guo, W.; Yang, Y.; Wang, Z.; Jiang, D.; Xu, X. Umbilical cord-derived mesenchymal stem (stromal) cells for treatment
of severe sepsis: Aphase 1 clinical trial. Transl. Res. 2018,199, 52–61. [CrossRef] [PubMed]
333.
Squillaro, T.; Antonucci, I.; Alessio, N.; Esposito, A.; Cipollaro, M.; Melone, M.A.B.; Peluso, G.; Stuppia, L.; Galderisi, U. Impact
of lysosomal storage disorders on biology of mesenchymal stem cells: Evidences from
in vitro
silencing of glucocerebrosidase
(GBA) and alpha-galactosidase A (GLA) enzymes. J. Cell. Physiol. 2017,232, 3454–3467. [CrossRef] [PubMed]
... Among different types of stem cells, MSCs can be named the most investigated type of stem cells to date. MSCs are mature non-hematopoietic stem cells that play important roles in the proliferation, implantation, differentiation, and self-renewal of cells through the production and secretion of certain growth factors and MVs (Kulus et al. 2021). MSCs can be isolated from the tissues of mesodermal origin, and they contribute to the regeneration of several tissues, including bone and cartilage. ...
Article
Full-text available
Background Microvesicles are membraned particles produced by different types of cells recently investigated for anticancer purposes. The current study aimed to investigate the effects of human bone marrow mesenchymal stem cell-derived microvesicles (BMSC-MVs) on the multiple myeloma cell line U266. BMSC-MVs were isolated from BMSCs via ultracentrifugation and characterized using transmission electron microscopy (TEM) and dynamic light scattering (DLS). U266 cells were treated with 15, 30, 60, and 120 µg/mL BMSC-MVs for three and seven days and the effects of treatment in terms of viability, cytotoxicity, and DNA damage were investigated via the MTT assay, lactate dehydrogenase (LDH) assay, and 8‑hydroxy-2’-deoxyguanosine (8‑OHdG) measurement, respectively. Moreover, the apoptosis rate of the U266 cells treated with 60 µg/mL BMSC-MVs was also assessed seven days following treatment via flow cytometry. Ultimately, the expression level of BCL2, BAX, and CCND1 by the U266 cells was examined seven days following treatment with 60 µg/mL BMSC-MVs using qRT-PCR. Results BMSC-MVs had an average size of ~ 410 nm. According to the MTT and LDH assays, BMSC-MV treatment reduced the U266 cell viability and mediated cytotoxic effects against them, respectively. Moreover, elevated 8‑OHdG levels following BMSC-MV treatment demonstrated a dose-dependent increase of DNA damage in the treated cells. BMSC-MV-treated U266 cells also exhibited an increased apoptosis rate after seven days of treatment. The expression level of BCL2 and CCND1 decreased in the treated cells whereas the BAX expression demonstrated an incremental pattern. Conclusions Our findings accentuate the therapeutic benefit of BMSC-MVs against the multiple myeloma cell line U266 and demonstrate how microvesicles could be of therapeutic advantage. Future in vivo studies could further corroborate these findings.
... However, they are found in almost all postnatal tissue types. Isolated cells consist of mixed populations of progenitor cells, multipotent stem cells and stem cells with varying degrees of differentiating capacity and differentiated cells [1]. According to the International Society for Cellular Therapy ISTC, MSCs must fulfil minimum criteria: (i) MSCs must be able to adhere to plastic, (ii) they must express the surface markers CD105, CD73 and CD90 but not CD45, CD34, CD14, CD19 and HLA-DR, (iii) MSCs must differentiate in vitro into adipocytes, chondrocytes and osteocytes [2]. ...
Article
Full-text available
Mesenchymal stem/stromal cells (MSCs) are not only capable of self-renewal, trans-differentiation, homing to damaged tissue sites and immunomodulation by secretion of trophic factors but are also easy to isolate and expand. Because of these characteristics, they are used in numerous clinical trials for cell therapy including immune and neurological disorders, diabetes, bone and cartilage diseases and myocardial infarction. However, not all trials have successful outcomes, due to unfavourable microenvironmental factors and the heterogenous nature of MSCs. Therefore, genetic manipulation of MSCs can increase their prospect. Currently, most studies focus on single transfection with one gene. Even though the introduction of more than one gene increases the complexity, it also increases the effectivity as different mechanism are triggered, leading to a synergistic effect. In this review we focus on the methodology and efficiency of co-transfection, as well as the opportunities and pitfalls of these genetically engineered cells for therapy. Graphical abstract
... MSCs are isolated primarily from bone marrow, adipose tissue, and umbilical cord, which have the ability to differentiate into bone, cartilage, or fat cells [1]. Current criteria for MSCs isolation yield heterogeneous, non-clonal cultures of stromal cells, including stem cells with diverse multipotential properties, committed progenitors, and differentiated cells [2,3]. As promising new therapeutic strategy, MSCs-based therapy is under investigation for treatment of many conditions, including inflammatory, ischemic and neurodegenerative diseases [4]. ...
Article
Full-text available
Background The clinical application of human bone-marrow derived mesenchymal stem cells (MSCs) for the treatment of refractory diseases has achieved remarkable results. However, there is a need for a systematic evaluation of the quality and safety of MSCs sourced from donors. In this study, we sought to assess one potential factor that might impact quality, namely the age of the donor. Methods We downloaded two data sets from each of two Gene Expression Omnibus (GEO), GSE39035 and GSE97311 databases, namely samples form young (< 65 years of age) and old (> 65) donor groups. Through, bioinformatics analysis and experimental validation to these retrieved data, we found that MSCs derived from aged donors can lead to differential expression of gene profiles compared with those from young donors, and potentially affect the function of MSCs, and may even induce malignant tumors. Results We identified a total of 337 differentially expressed genes (DEGs), including two upregulated and eight downregulated genes from the databases of both GSE39035 and GSE97311. We further identified 13 hub genes. Six of them, TBX15, IGF1, GATA2, PITX2, SNAI1 and VCAN, were highly expressed in many human malignancies in Human Protein Atlas database. In the MSCs in vitro senescent cell model, qPCR analysis validated that all six hub genes were highly expressed in senescent MSCs. Our findings confirm that aged donors of MSCs have a significant effect on gene expression profiles. The MSCs from old donors have the potential to cause a variety of malignancies. These TBX15, IGF1, GATA2, PITX2, SNAI1, VCAN genes could be used as potential biomarkers to diagnosis aging state of donor MSCs, and evaluate whether MSCs derived from an aged donor could be used for therapy in the clinic. Our findings provide a diagnostic basis for the clinical use of MSCs to treat a variety of diseases. Conclusions Therefore, our findings not only provide guidance for the safe and standardized use of MSCs in the clinic for the treatment of various diseases, but also provide insights into the use of cell regeneration approaches to reverse aging and support rejuvenation.
Article
Full-text available
Mesenchymal stem cells (MSCs) are known for their immunosuppressive properties. Based on the demonstrated anti-inflammatory effect of mouse MSCs from hair follicles (moMSCORS) in a murine wound closure model, this study evaluates their potential for preventing type 1 diabetes (T1D) in C57BL/6 mice. T1D was induced in C57BL/6 mice by repeated low doses of streptozotocin. moMSCORS were injected intravenously on weekly basis. moMSCORS reduced T1D incidence, the insulitis stage, and preserved insulin production in treated animals. moMSCORS primarily exerted immunomodulatory effects by inhibiting CD4+ T cell proliferation and activation. Ex vivo analysis indicated that moMSCORS modified the cellular immune profile within pancreatic lymph nodes and pancreatic infiltrates by reducing the numbers of M1 pro-inflammatory macrophages and T helper 17 cells and upscaling the immunosuppressive T regulatory cells. The proportion of pathogenic insulin-specific CD4+ T cells was down-scaled in the lymph nodes, likely via soluble factors. The moMSCORS detected in the pancreatic infiltrates of treated mice presumably exerted the observed suppressive effect on CD4+ through direct contact. moMSCORS alleviated T1D symptoms in the mouse, qualifying as a candidate for therapeutic products by multiple advantages: non-invasive sampling by epilation, easy access, permanent availability, scalability, and benefits of auto-transplantation.
Article
A BSTRACT Background Pregnancy-Induced hypertension (PIH) is a common health problem that occurs during pregnancy and may lead to limited fetal growth and other major health problems. Being a serious public health issue, PIH is responsible for a significant number of deaths across the globe. Hyrtl’s anastomosis of the umbilical arteries and placenta is a blood vessel system that can affect the growth and development of the fetus. The studies conducted earlier have suggested that the abnormalities in Hyrtl’s anastomosis may be associated with the development of PIH and other adverse pregnancy outcomes. Aim The aim of the current systematic literature review is to investigate the potential advantages and restrictions of Hyrtl’s anastomosis as a medical diagnosis and treatment tool for PIH by reviewing the available evidence. Materials and Methods The current study is a systematic literature review that involves the selection of 10 articles published in or after 2019 from reputed journals that evaluated the effectiveness of Hyrtl’s anastomosis in PIH. The inclusion criteria for this review are as follows: studies that involved human subjects, were published in English, were randomized controlled trials, case-control studies, observational studies, or systematic reviews, and evaluated the effectiveness of Hyrtl’s anastomosis. The exclusion criteria are as follows: studies published in other languages did not involve human subjects and did not measure the effectiveness of Hyrtl’s anastomosis in PIH. The search strategy includes a comprehensive search of the relevant databases using keywords related to Hyrtl’s anastomosis, umbilical artery, placenta, and PIH. Both data collection and analysis were conducted when the authenticity and validity of the articles used in this study were maintained. Data extraction was carried out by identifying the key findings of the articles selected for review in this particular study. The quality of the studies included in this review was assessed based on the JADAD scale. Findings and Conclusion The aim of the systematic literature review is to evaluate the effectiveness of Hyrtl’s anastomosis in the detection and management of PIH. The current study design involved the selection of 10 articles published in or after 2019, with inclusion and exclusion criteria defined for the selection process. The search strategy included a comprehensive search of relevant databases. Both data collection and analysis were conducted when the authenticity and validity of the chosen articles were maintained. The quality of the studies included in the review was assessed based on the JADAD scale.
Article
Full-text available
Chronic kidney disease (CKD) is a slowly progressive condition characterized by decreased kidney function, tubular injury, oxidative stress, and inflammation. CKD is a leading global health burden that is asymptomatic in early stages but can ultimately cause kidney failure. Its etiology is complex and involves dysregulated signaling pathways that lead to fibrosis. Transforming growth factor (TGF)-β is a central mediator in promoting transdifferentiation of polarized renal tubular epithelial cells into mesenchymal cells, resulting in irreversible kidney injury. While current therapies are limited, the search for more effective diagnostic and treatment modalities is intensive. Although biopsy with histology is the most accurate method of diagnosis and staging, imaging techniques such as diffusion-weighted magnetic resonance imaging and shear wave elastography ultrasound are less invasive ways to stage fibrosis. Current therapies such as renin-angiotensin blockers, mineralocorticoid receptor antagonists, and sodium/glucose cotransporter 2 inhibitors aim to delay progression. Newer antifibrotic agents that suppress the downstream inflammatory mediators involved in the fibrotic process are in clinical trials, and potential therapeutic targets that interfere with TGF-β signaling are being explored. Small interfering RNAs and stem cell-based therapeutics are also being evaluated. Further research and clinical studies are necessary in order to avoid dialysis and kidney transplantation.
Article
Epidermolysis bullosa (EB) is a group of rare genetic skin fragility disorders, which are hereditary. These disorders are associated with mutations in at least 16 genes that encode components of the epidermal adhesion complex. Currently, there are no effective treatments for this disorder. All current treatment approaches focus on topical treatments to prevent complications and infections. In recent years, significant progress has been achieved in the treatment of the severe genetic skin blistering condition known as EB through preclinical and clinical advancements. Promising developments have emerged in the areas of protein and cell therapies, such as allogeneic stem cell transplantation; in addition, RNA-based therapies and gene therapy approaches have also become a reality. Stem cells obtained from embryonic or adult tissues, including the skin, are undifferentiated cells with the ability to generate, maintain, and replace fully developed cells and tissues. Recent advancements in preclinical and clinical research have significantly enhanced stem cell therapy, presenting a promising treatment option for various diseases that are not effectively addressed by current medical treatments. Different types of stem cells such as primarily hematopoietic and mesenchymal, obtained from the patient or from a donor, have been utilized to treat severe forms of diseases, each with some beneficial effects. In addition, extensive research has shown that gene transfer methods targeting allogeneic and autologous epidermal stem cells to replace or correct the defective gene are promising. These methods can regenerate and restore the adhesion of primary keratinocytes in EB patients. The long-term treatment of skin lesions in a small number of patients has shown promising results through the transplantation of skin grafts produced from gene-corrected autologous epidermal stem cells. This article attempts to summarize the current situation, potential development prospects, and some of the challenges related to the cell therapy approach for EB treatment.
Article
Full-text available
Spontaneous bacterial peritonitis (SBP) is a severe complication in patients with decompensated liver cirrhosis and is commonly treated with broad spectrum antibiotics. However, the rise of antibiotic resistance requires alternative therapeutic strategies. As recently shown, human amnion-derived mesenchymal stem cells (hA-MSCs) are able, in vitro, to promote bacterial clearance and modulate the immune and inflammatory response in SBP. Our results highlight the upregulation of FOXO1, CXCL5, CXCL6, CCL20, and MAPK13 in hA-MSCs as well as the promotion of bacterial clearance, prompting a shift in the immune response toward a Th17 lymphocyte phenotype after 72 h treatment. In this study, we used an in vitro SBP model and employed omics techniques (next-generation sequencing) to investigate the mechanisms by which hA-MSCs modify the crosstalk between immune cells in LPS-stimulated ascitic fluid. We also validated the data obtained via qRT-PCR, cytofluorimetric analysis, and Luminex assay. These findings provide further support to the hope of using hA-MSCs for the prevention and treatment of infective diseases, such as SBP, offering a viable alternative to antibiotic therapy.
Chapter
Stem cells with self-renewal and multi-lineage differentiation potential have potential for developing medicines for a range of refractory and recurrent disease. This book mainly focuses on the landscape of the biological properties and translational research of stem cells types, including hematopoietic stem cells (HSCs), neural stem cells (NSCs) and mesenchymal stem/stromal cells (MSCs). The book also introduces readers to the current updates and development prospects of stem cells in singular or combination therapies with advanced biomaterials and technological innovations towards large-scale standardization and productization.
Article
Full-text available
Ischemic stroke (IS) is a leading cause of death and disability worldwide. Tissue plasminogen activator (tPA) administration and mechanical thrombectomy are the main treatments but have a narrow time window. Mesenchymal stem cells (MSCs), which are easily scalable in vitro and lack ethical concerns, possess the potential to differentiate into various types of cells and secrete a great number of growth factors for neuroprotection and regeneration. Moreover, MSCs have low immunogenicity and tumorigenic properties, showing safety and preliminary efficacy both in preclinical studies and clinical trials of IS. However, it is unlikely that MSC treatment alone will be sufficient to maximize recovery due to the low survival rate of transplanted cells and various mechanisms of ischemic brain damage in the different stages of IS. Preconditioning was used to facilitate the homing, survival, and secretion ability of the grafted MSCs in the ischemic region, while combination therapies are alternatives that can maximize the treatment effects, focusing on multiple therapeutic targets to promote stroke recovery. In this case, the combination therapy can yield a synergistic effect. In this review, we summarize the type of MSCs, preconditioning methods, and combined strategies as well as their therapeutic mechanism in the treatment of IS to accelerate the transformation from basic research to clinical application.
Article
Full-text available
Mesenchymal stem/stromal cells (MSCs) have broad application prospects for regenerative medicine due to their self‐renewal, high plasticity, ability for differentiation, and immune response and modulation. Interest in turning MSCs into clinical applications has never been higher than at present. Many biotech companies have invested great effort from development of clinical grade MSC product to investigational new drug (IND) enabling studies. Therefore, the growing demand for publication of MSC regulation in China necessitates various discussions in accessible professional journals. The National Medical Products Administration has implemented regulations on the clinical application of MSCs therapy. The regulations for MSCs products as drug have been updated in recent years in China. This review will look over the whole procedure in allogeneic MSC development, including regulations, guidance, processes, quality management, pre‐IND meeting, and IND application for obtaining an approval to start clinical trials in China. The review focused on process and regulatory challenges in the development of MSCs products, with the goal of providing strategies to meet regulatory demands. This article describes a path for scientists, biotech companies, and clinical trial investigators toward the successful development of MSC‐based therapeutic product.
Article
Full-text available
More than 80 diseases are currently classified as autoimmune, with a rising prevalence throughout the world. Systemic lupus erythematosus (SLE) is classified as a systemic autoimmune disorder, but the exact pathogenesis of SLE remains elusive. Currently available treatment strategies offer only the possibility for disease remission making it essential to develop more effective and safer strategies for treatment. Recently MSCs are gaining attention as attractive therapeutic tools for autoimmune disease treatment. Special focus should be given to MSCs originated from perinatal tissues such as Wharton’s jelly, as they present unique immunomodulatory properties and remarkably low immunogenicity. MSCs exert their immunomodula-tory effects via direct cell-to-cell communication as well as in a paracrine manner, creating possibility to apply secretome of MSCs as an individual therapeutic tool. Although the secretome of MSCs has not yet been utilized in SLE treatment, its efficacy has been suggested in other disorders, such as multiple sclerosis or Alzheimer’s disease. Regular administration of paracrine factors derived from MSCs could potentially effect in significant reduction of SLE symptoms and in maintenance of disease remission.
Article
Full-text available
Mesenchymal stromal cells (MSCs), present in the stromal component of several tissues, include multipotent stem cells, progenitors, and differentiated cells. MSCs have quickly attracted considerable attention in the clinical field for their regenerative properties and their ability to promote tissue homeostasis following injury. In recent years, MSCs mainly isolated from bone marrow, adipose tissue, and umbilical cord—have been utilized in hundreds of clinical trials for the treatment of various diseases. However, in addition to some successes, MSC-based therapies have experienced several failures. The number of new trials with MSCs is exponentially growing; still, complete results are only available for a limited number of trials. This dearth does not help prevent potentially inefficacious and unnecessary clinical trials. Results from unsuccessful studies may be useful in planning new therapeutic approaches to improve clinical outcomes. In order to bolster critical analysis of trial results, we reviewed the state of art of MSC clinical trials that have been published in the last six years. Most of the 416 published trials evaluated MSCs’ effectiveness in treating cardiovascular diseases, GvHD, and brain and neurological disorders, although some trials sought to treat immune system diseases and wounds and to restore tissue. We also report some unorthodox clinical trials that include unusual studies. Graphical abstract
Article
Full-text available
The use of stem cells is part of a strategy for the treatment of a large number of diseases. However, the source of the original stem cells for use is extremely important and determines their therapeutic potential. Mesenchymal stromal cells (MSC) have proven their therapeutic effectiveness when used in a number of pathological models. However, it remains an open question whether the chronological age of the donor organism affects the effectiveness of the use of MSC. The asymmetric division of stem cells, the result of which is some residential stem cells acquiring a non-senile phenotype, means that stem cells possess an intrinsic ability to preserve juvenile characteristics, implying an absence or at least remarkable retardation of senescence in stem cells. To test whether residential MSC senesce, we evaluated the physiological changes in the MSC from old rats, with a further comparison of the neuroprotective properties of MSC from young and old animals in a model of traumatic brain injury. We found that, while the effect of administration of MSC on lesion volume was minimal, functional recovery was remarkable, with the highest effect assigned to fetal cells; the lowest effect was recorded for cells isolated from adult rats and postnatal cells, having intermediate potency. MSC from the young rats were characterized by a faster growth than adult MSC, correlating with levels of proliferating cell nuclear antigen (PCNA). However, there were no differences in respiratory activity of MSC from young and old rats, but young cells showed much higher glucose utilization than old ones. Autophagy flux was almost the same in both types of cells, but there were remarkable ultrastructural differences in old and young cells.
Article
Full-text available
Cell encapsulation utilizing biodegradable material has promising outcomes for tissue engineering. From a long time ago, alginate has been generally utilized for drug delivery, cell transplantation and as a scaffold in biomedical applications. The aim of this study was the comparison of cell viability in the presence of two polymerizing ions: Ba ²⁺ and Ca ²⁺ to improvement the quality of alginate scaffold. For this purpose, WJMSCs after three passage were encapsulated in alginate scaffold in the presence of Ba ²⁺ and ca ²⁺ . Cell viability was evaluated by WST-8 assay kit after 24, 48 and 72 hours. The results showed that encapsulated cells in the presence of Ca ²⁺ had more viability than Ba ²⁺ . It was also found that using the WST-8 assay kit is a convenient and fast method for evaluation the viability of cells. It can be claimed that Calcl2 polymerizing solution provides more favorable conditions for cell viability compared to Bacl2 solution. Running title: Assessing the viability of stem cells by WST-8 assay kit
Article
Full-text available
The trophoblast is a critical cell for placental development and embryo implantation in the placenta. We previously reported that placenta‐derived mesenchymal stem cells (PD‐MSCs) increase trophoblast invasion through several signaling pathways. However, the paracrine effects of PD‐MSCs on mitochondrial function in trophoblasts are still unclear. Therefore, the objective of the study was to analyze the mitochondrial function of trophoblasts in response to cocultivation with PD‐MSCs. The results showed that PD‐MSCs regulate the balance between cell survival and death and protect damaged mitochondria in trophoblasts from oxidative stress. Moreover, PD‐MSCs upregulate factors involved in mitochondrial autophagy in trophoblast cells. Finally, PD‐MSCs improve trophoblast invasion. Taken together, the data indicate that PD‐MSCs can regulate trophoblast invasion through dynamic effects on mitochondrial energy metabolism. These results support the fundamental role of mitochondrial energy mechanism in trophoblast invasion and suggest a new therapeutic strategy for infertility. Placenta‐derived mesenchymal stem cells can regulate trophoblast invasion through dynamic effects on mitochondrial energy metabolism. The fundamental role of mitochondrial energy mechanism in trophoblast invasion and suggest a new therapeutic strategy for infertility.
Article
Full-text available
Treatment of severe Coronavirus Disease 2019 (COVID-19) is challenging. We performed a phase 2 trial to assess the efficacy and safety of human umbilical cord-mesenchymal stem cells (UC-MSCs) to treat severe COVID-19 patients with lung damage, based on our phase 1 data. In this randomized, double-blind, and placebo-controlled trial, we recruited 101 severe COVID-19 patients with lung damage. They were randomly assigned at a 2:1 ratio to receive either UC-MSCs (4 × 10 ⁷ cells per infusion) or placebo on day 0, 3, and 6. The primary endpoint was an altered proportion of whole lung lesion volumes from baseline to day 28. Other imaging outcomes, 6-minute walk test (6-MWT), maximum vital capacity, diffusing capacity, and adverse events were recorded and analyzed. In all, 100 COVID-19 patients were finally received either UC-MSCs ( n = 65) or placebo ( n = 35). UC-MSCs administration exerted numerical improvement in whole lung lesion volume from baseline to day 28 compared with the placebo (the median difference was −13.31%, 95% CI −29.14%, 2.13%, P = 0.080). UC-MSCs significantly reduced the proportions of solid component lesion volume compared with the placebo (median difference: −15.45%; 95% CI −30.82%, −0.39%; P = 0.043). The 6-MWT showed an increased distance in patients treated with UC-MSCs (difference: 27.00 m; 95% CI 0.00, 57.00; P = 0.057). The incidence of adverse events was similar in the two groups. These results suggest that UC-MSCs treatment is a safe and potentially effective therapeutic approach for COVID-19 patients with lung damage. A phase 3 trial is required to evaluate effects on reducing mortality and preventing long-term pulmonary disability. (Funded by The National Key R&D Program of China and others. ClinicalTrials.gov number, NCT04288102.
Article
Full-text available
Background: Acute respiratory distress syndrome (ARDS) is a fatal complication of coronavirus disease 2019 (COVID-19). There are a few reports of allogeneic human mesenchymal stem cells (MSCs) as a potential treatment for ARDS. In this phase 1 clinical trial, we present the safety, feasibility, and tolerability of the multiple infusions of high dose MSCs, which originated from the placenta and umbilical cord, in critically ill COVID-19-induced ARDS patients. Methods: A total of 11 patients diagnosed with COVID-19-induced ARDS who were admitted to the intensive care units (ICUs) of two hospitals enrolled in this study. The patients were critically ill with severe hypoxemia and required mechanical ventilation. The patients received three intravenous infusions (200 × 106 cells) every other day for a total of 600 × 106 human umbilical cord MSCs (UC-MSCs; 6 cases) or placental MSCs (PL-MSCs; 5 cases). Findings: There were eight men and three women who were 42 to 66 years of age. Of these, six (55%) patients had comorbidities of diabetes, hypertension, chronic lymphocytic leukemia (CLL), and cardiomyopathy (CMP). There were no serious adverse events reported 24-48 h after the cell infusions. We observed reduced dyspnea and increased SpO2 within 48-96 h after the first infusion in seven patients. Of these seven patients, five were discharged from the ICU within 2-7 days (average: 4 days), one patient who had signs of acute renal and hepatic failure was discharged from the ICU on day 18, and the last patient suddenly developed cardiac arrest on day 7 of the cell infusion. Significant reductions in serum levels of tumor necrosis factor-alpha (TNF-α; P < 0.01), IL-8 (P < 0.05), and C-reactive protein (CRP) (P < 0.01) were seen in all six survivors. IL-6 levels decreased in five (P = 0.06) patients and interferon gamma (IFN-γ) levels decreased in four (P = 0.14) patients. Four patients who had signs of multi-organ failure or sepsis died in 5-19 days (average: 10 days) after the first MSC infusion. A low percentage of lymphocytes (< 10%) and leukocytosis were associated with poor outcome (P = 0.02). All six survivors were well with no complaints of dyspnea on day 60 post-infusion. Radiological parameters of the lung computed tomography (CT) scans showed remarkable signs of recovery. Interpretation: We suggest that multiple infusions of high dose allogeneic prenatal MSCs are safe and can rapidly improve respiratory distress and reduce inflammatory biomarkers in some critically ill COVID-19-induced ARDS cases. Patients that develop sepsis or multi-organ failure may not be good candidates for stem cell therapy. Large randomized multicenter clinical trials are needed to discern the exact therapeutic potentials of MSC in COVID-19-induced ARDS.
Article
The most common clinical manifestations of age-related musculoskeletal degeneration are osteoarthritis and osteoporosis, and these represent an enormous burden on modern society. Mesenchymal stromal cells (MSCs) have pivotal roles in musculoskeletal tissue development. In adult organisms, MSCs retain their ability to regenerate tissues following bone fractures, articular cartilage injuries, and other traumatic injuries of connective tissue. However, their remarkable regenerative ability appears to be impaired through aging, and in particular in age-related diseases of bones and joints. Here, we review age-related alterations of MSCs in musculoskeletal tissues, and address the underlying mechanisms of aging and senescence of MSCs. Furthermore, we focus on the properties of MSCs in osteoarthritis and osteoporosis, and how their changes contribute to onset and progression of these disorders. Finally, we consider current treatments that exploit the enormous potential of MSCs for tissue regeneration, as well as for innovative cell-free extracellular-vesicle-based and anti-aging treatment approaches.
Article
For potential clinical applications in the future, we investigated the distinct biological features of mesenchymal stromal cells (MSCs) derived from different origin areas of human placenta and individual heterogeneity among different donors. Chorionic plate MSCs (CP-MSCs), amniotic membrane MSCs (AM-MSCs), and decidual plate MSCs (DP-MSCs) were isolated from 5 human placentae and were analyzed in terms of main features of MSCs including surface marker profile, growth, differentiation potential, immune regulation capability, and tubulin acetylation (Ac-tubulin). The expression profile of surface markers in the 3 types of MSCs derived from the 5 donors was relatively stable. Heterogeneity was found in growth, differentiation potential, and immune regulation among MSCs according to the different areas of isolation and different donors. CP-MSCs and AM-MSCs derived from the placentae of donors 1–3 had a higher osteogenic differentiation potential than the corresponding DP-MSCs, but those derived from the placentae of donors 4 and 5 had a markedly lower osteogenic differentiation potential than DP-MSCs. All CP-MSCs derived from donors 1–3 had the highest adipogenic differentiation potential, but CP-MSCs derived from donors 4 and 5 did not show strong capability of adipogenic differentiation. CP-MSCs markedly inhibited the proliferation of peripheral blood mononuclear cells (PBMCs) induced by phytohemagglutinin, whereas AM- and DP-MSCs did not. All MSCs decreased the proportion of CD3+/CD8–/IFN-γ+ Th1 and CD3+/CD8–/IL17+ Th17 cells, but increased the proportion of Treg cells in PBMCs, with individual differences among the 5 donors. DP-MSCs from donors 1 and 2 had higher levels of Ac-tubulin compared with CP- and AM-MSCs. However, the levels of Ac-tubulin in AM-MSCs from donors 3 and 5 were higher than those of the other 2 types of MSCs. Our results revealed that there was tissue-specific heterogeneity among the 3 types of MSCs from different origin tissues of placenta and individual heterogeneity among donors. In future, the pre-selected placenta-derived MSCs with specific biological advantages may improve the curative effect of cell therapy in different situations.