ArticlePDF AvailableLiterature Review

Mesenchymal stromal cells (MSCs) and their exosome in acute liver failure (ALF): a comprehensive review

Authors:

Abstract and Figures

Recently, mesenchymal stromal cells (MSCs) and their derivative exosome have become a promising approach in the context of liver diseases therapy, in particular, acute liver failure (ALF). In addition to their differentiation into hepatocytes in vivo, which is partially involved in liver regeneration, MSCs support liver regeneration as a result of their appreciated competencies, such as antiapoptotic, immunomodulatory, antifibrotic, and also antioxidant attributes. Further, MSCs-secreted molecules inspire hepatocyte proliferation in vivo, facilitating damaged tissue recovery in ALF. Given these properties, various MSCs-based approaches have evolved and resulted in encouraging outcomes in ALF animal models and also displayed safety and also modest efficacy in human studies, providing a new avenue for ALF therapy. Irrespective of MSCs-derived exosome, MSCs-based strategies in ALF include administration of native MSCs, genetically modified MSCs, pretreated MSCs, MSCs delivery using biomaterials, and also MSCs in combination with and other therapeutic molecules or modalities. Herein, we will deliver an overview regarding the therapeutic effects of the MSCs and their exosomes in ALF. As well, we will discuss recent progress in preclinical and clinical studies and current challenges in MSCs-based therapies in ALF, with a special focus on in vivo reports.
This content is subject to copyright. Terms and conditions apply.
Shokravietal. Stem Cell Research & Therapy (2022) 13:192
https://doi.org/10.1186/s13287-022-02825-z
REVIEW
Mesenchymal stromal cells (MSCs)
andtheir exosome inacute liver failure (ALF):
acomprehensive review
Samin Shokravi1, Vitaliy Borisov2, Burhan Abdullah Zaman3, Firoozeh Niazvand4, Raheleh Hazrati5,
Meysam Mohammadi Khah6, Lakshmi Thangavelu7, Sima Marzban1*, Armin Sohrabi8,9 and Amir Zamani10*
Abstract
Recently, mesenchymal stromal cells (MSCs) and their derivative exosome have become a promising approach in the
context of liver diseases therapy, in particular, acute liver failure (ALF). In addition to their differentiation into hepato-
cytes in vivo, which is partially involved in liver regeneration, MSCs support liver regeneration as a result of their
appreciated competencies, such as antiapoptotic, immunomodulatory, antifibrotic, and also antioxidant attributes.
Further, MSCs-secreted molecules inspire hepatocyte proliferation in vivo, facilitating damaged tissue recovery in ALF.
Given these properties, various MSCs-based approaches have evolved and resulted in encouraging outcomes in ALF
animal models and also displayed safety and also modest efficacy in human studies, providing a new avenue for ALF
therapy. Irrespective of MSCs-derived exosome, MSCs-based strategies in ALF include administration of native MSCs,
genetically modified MSCs, pretreated MSCs, MSCs delivery using biomaterials, and also MSCs in combination with
and other therapeutic molecules or modalities. Herein, we will deliver an overview regarding the therapeutic effects
of the MSCs and their exosomes in ALF. As well, we will discuss recent progress in preclinical and clinical studies and
current challenges in MSCs-based therapies in ALF, with a special focus on in vivo reports.
Keywords: Mesenchymal stromal cell (MSCs), Exosome, Acute liver failure (ALF), Immunomodulation, Hepatocyte
© The Author(s) 2022. Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which
permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the
original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or
other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line
to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory
regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this
licence, visit http:// creat iveco mmons. org/ licen ses/ by/4. 0/. The Creative Commons Public Domain Dedication waiver (http:// creat iveco
mmons. org/ publi cdoma in/ zero/1. 0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.
Introduction
Acute liver failure (ALF) is characterized by the occur-
rence of coagulopathy (international normalized ratio
[INR] > 1.5) and any level of encephalopathy emerging
24weeks following the occurrence of the first symptoms
in patients who have no history of previous liver disease
[1]. e timing and the level of clinical presentation can
be classified into three types: hyperacute, acute, and sub-
acute [2]. Hyperacute and acute types involve fulminant
hepatic failure, while the subacute type is also named
subfulminant [3]. Interestingly, the mortality rate among
the patients whose hepatic encephalopathy appears
8weeks after the onset of symptoms (fulminant hepatic
failure) is lower than the patients with a more gradually
evolving course [4]. Multiorgan failure (MOF) has proved
to be the main cause of death (> 50%) from ALF, while
intracranial hypertension (ICH) and infection are the
other main causes of mortality in this patient population
[5].
During last two decades, a diversity of stem cells, such
as pluripotent stem cells (PSCs), mesenchymal stro-
mal cells (MSCs), hepatic progenitor cells (HPCs), and
hematopoietic stem cells (HSCs), has been used to treat
liver diseases [68]. However, MSCs are the most com-
mon type used in research, since they pose no ethical
Open Access
*Correspondence: Smarzban@larkinhospital.com; amir.zamani19800@gmail.
com
1 Department of Research and Academic Affairs, Larkin Community
Hospital, Miami, FL, USA
10 Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz,
Iran
Full list of author information is available at the end of the article
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 2 of 21
Shokravietal. Stem Cell Research & Therapy (2022) 13:192
challenges and can be obtained easily [9, 10]. Results
show that MSCs have the capability of differentiating
more than once; moreover, they can self-renew. ey
can differentiate into an array of cell lineages, including
hepatocyte-like cells (HLCs) [11]. MSCs are also charac-
terized by other properties, such as anti-inflammatory,
antiapoptosis, antifibrotic, antioxidant, blood vessel for-
mation, improvement of tissue repair, and growth factor
secretion [12, 13]. Despite many preclinical and clinical
investigations on MSCs used in treating ALF, it is still
unknown what mechanism contributes to the therapeutic
effect of MSCs. Besides, MSCs-exosomes have caught the
attention of many researchers as a new cell-free method
regarding the regeneration of the liver [14, 15]. ey have
dissipated the worries concerning the direct application
of MSC (e.g., immunogenicity and tumor formation [16].
Such exosomes encompass high frequencies of cytoplas-
mic and membrane proteins, including enzymes, tran-
scription factors, lipids, ECM proteins. ey also include
nucleic acids, such as mitochondrial DNA (mtDNA),
single-stranded DNA (ssDNA), double-stranded DNA
(dsDNA), messenger RNA (mRNA), and microRNA
(miRNA) [17]. e size of exosomes varies from 30 to
150nm, and they can be transferred to other cells to do
their functions. As a highly controlled process, the gen-
eration of exosome from the other organisms similar to
themselves is comprised of three main steps: (1) forma-
tion of endocytic vesicles by the folding of the exterior
area of the plasma membrane, (2) formation of multive-
sicular bodies (MVBs) by inward budding of the endo-
somal membrane, and (3) incorporation of established
MVBs with the plasma membrane and secretion of the
vesicular contents, called exosomes [14, 18]. Exosomes
elicit antioxidant effects and motivate target cells to trig-
ger downstream signals. Moreover, they convey genetic
material to target cells, leading to the suppression of
inflammation and apoptosis. [19, 20].
is review aims to give an overview of the present
knowledge to elucidate mechanisms used by MSCs to
underlie liver restoration in ALF. Another aim is to pre-
sent a discussion of new developments in preclinical and
clinical investigations on MSCs therapy in liver-associ-
ated diseases, with a particular focus on ALF.
Pathophysiology ofALF
Acetaminophen (APAP) has proved to be the main
cause of ALF [21]. e following people are highly likely
to experience ALF stimulated by APAP: alcoholic peo-
ple who use APAP; people who suffer from malnutri-
tion; or people who take medications that are believed
to induce CYP450 enzymes (e.g., phenytoin, carbamaz-
epine, or rifampin). Results of a study on 308 patients
with severe liver disorder in the USA revealed APAP as
the main cause of ALF in 40% of patients [22]. e other
causes detected were as follows in the increasing order of
prevalence:
Malignancy (1%)
Budd-Chiari Syndrome (2%)
Pregnancy (2%)
Wilson disease (3%)
Hepatitis A virus infection (4%)
Autoimmune hepatitis (4%)
Ischemic hepatitis (6%),
Hepatitis B virus infection (6%)
Idiosyncratic drug-induced liver injury (13%)
e causes of 17 percent of cases were not known [4].
Based on results, it is possible to categorize the ALF
pathophysiology into two groups: pathophysiologies
of liver problems involving a specific cause and patho-
physiology concerning the appearance of second-
ary multiorgan failure (MOF) [23]. With regard to the
pathophysiology of liver disorders, the results show that
APAP toxicity is the main cause [24]. Secondary MOF
often derives from the primary extensive pro-inflam-
matory effect, which leads to a pervasive inflammatory
effect syndrome. en, a deregulated anti-inflammatory
response ensues [25, 26].
It is not clear what mechanism causes the ongoing
death of tissue when there is no ongoing injury. Oxida-
tive stress results in the formation of reactive oxygen
species (ROS). This, in turn, activates c-Jun N-termi-
nal kinase (JNK) through a series of events [27]. Such
activation may support disruption of normal mito-
chondrial function, which inspires more hepatocyte
necrosis and damage associated molecular patterns
(DAMPs) [28, 29]. DAMPs bring about the activation
of hepatic macrophages, resulting in the formation of
the inflammasome [30, 31]. Concisely, as complexes
characterized by multiple proteins, inflammasomes
receive the intracellular danger signals through NOD-
like receptors (NLRs) [32]. The inflammasome effec-
tively regulates the inflammatory response by eliciting
a response to low-threshold signals. Toll-like receptors
(TLRs) induction by DAMPs leads to the inflamma-
some activation, supporting the subsequent activation
of caspase-1 and IL-1β secretion [33, 34]. Researchers
have identified the characteristics of the NLR family
pyrin domain containing 3 (NLRP3) inflammasome
belonging to the inflammasome family. NLRP3 inflam-
masome has three potential activation pathways: (1)
ATP signal which occurs outside a cell, leading to
potassium efflux and pannexin recruitment; (2) incor-
poration of crystalized cholesterol, uric acid or amy-
loid with lysosomal dysfunction after the ingestion and
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 3 of 21
Shokravietal. Stem Cell Research & Therapy (2022) 13:192
elimination of these particles; and (3) activation by
reactive oxygen species (ROS) [33, 35, 36]. Investiga-
tions have examined the activation of inflammasome
in APAP-induced ALF by a special focus on the con-
tribution of the inflammasome to acute liver disorder
[37]. It appears that DAMPs are released from necrotic
hepatocytes and sinusoidal endothelial cells, leading
to the activation of the inflammasome in the manner
mentioned above.
The rationality ofMSCs therapy inALF
MSCs migration to damage tissue by interaction with
several receptors and molecules, and thereby inducing
liver recovery by various mechanisms has been proved
(Fig.1). Although the mechanisms of MSCs transplanta-
tion are still not entirely understood, a growing body of
proof has indicated that their immunomodulation, differ-
entiation, and antifibrotic capabilities play central roles in
liver repair. Among them, anti-inflammatory potential of
MSCs play most critical role. Although there is no clear
Fig. 1 Underlying mechanism complicated in mesenchymal stromal cells (MSCs) migration to damaged liver tissue. The connections between
CXCR4 and SDF-1ɑ, c-Met and HGF, and finally VLA-4 and VCAM-1 underlie cell to cell interaction between endothelial cells (ECs) and MSCs, which,
in turn, facilitate MSCs migration to damaged liver tissue. Then MSCs secrete anti-inflammatory molecules, such as PGE2, IDO, TGF-β, IL-10, and NO
to down-regulate inflammation. These molecules prompt the change of inflammatory to proliferating phase largely defined by the secretion of
PDGF and VEGF, sustaining hepatocyte formation and proliferation
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 4 of 21
Shokravietal. Stem Cell Research & Therapy (2022) 13:192
evidence indicating the MSCs invivo differentiation into
hepatoid cells post-transplantation, MSCs can be differ-
entiated into hepatocyte-like cells (HLCs) in vitro and
then be infused. Of course, this process is time-consum-
ing process with insufficient established HLCs, thereby
hindering its therapeutic utility in clinic. However, there
is some evidence indicating that replacing fetal bovine
serum (FBS) with polyvinyl alcohol (PVA) might lead
to improved differentiation ability [38]. Invivo, as only
a small number of intravenously injected cells reach the
liver, MSCs-mediated favored effects mainly depend on
the secreted molecules rather than their direct effects
or differentiation into hepatocytes post-transplantation
[39].
MSC anti‑inammatory properties
e hepatocyte loss is the first symptom and mechanism
contributing to acute liver damage. It is still unclear what
causes ongoing necrosis when there is no injury. ROS are
produced in response to oxidative stress. is, in turn,
activates c-Jun N-terminal kinase (JNK) through a series
of events, resulting in mitochondrial dysfunctions. ese
events lead to a higher level of hepatocyte necrosis, as
well as the expansion of DAMPs [40]. DAMPs stimulate
the activation of hepatic macrophages and the formation
of the inflammasome [41]. In the next stage, the release
of pro-inflammatory cytokines eases the recruitment of a
larger number of immune cells to the inflammation area,
and so advances hepatocyte cell necrosis.
e majority of past investigations have indicated that
MSCs play a therapeutic role in the treatment of liver
dysfunction by releasing trophic factors and the factors
modulating the immune system [42]. Although the role
of MSCs in modulating the immune system is unclear,
they might control the immune cells through the secre-
tion of soluble factors and the contacts between cells. e
regulation of adaptive and innate immune responses by
MSCs is exerted by inhibiting T cells and dendritic cells
(DCs), which leads to a reduction in the activation and
growth of B cells [43, 44]. is, in turn, enhances the
formation of regulatory T (Treg) cells and prevents the
growth and toxicity of natural killer (NK) cells induced by
the chemotherapeutic molecules [45]. Also, transforming
growth factor-beta (TGF-β) and interleukin 10 (IL-10)
as crucial factors in the regulation of a large number of
inflammatory cells [46, 47]. Studies revealed a significant
increase in the amounts of TGF-β and IL-10 in serum
following the injection of UC-MSCs, but a significant
decrease in the amounts of IL-6, tumor necrosis factor-
alpha (TNF-α), and cytotoxic T lymphocytes (CTLs) was
seen in peripheral blood [48]. is led to the restoration
of liver function, as well as a reduction in the develop-
ment of disease and the level of mortality. Furthermore,
transient T cell apoptosis can be induced by BM-MSCs
through the Fas ligand (FasL)-dependent pathway [49].
en, macrophages are stimulated by apoptotic T cells
to form high amounts of TGF-β, resulting in the up-reg-
ulation of Treg cells to trigger immune tolerance. MSCs
can prevent cytotoxic CTLs and NK cells through the
contact between cells and paracrine factors, including
indoleamine 2,3-dioxygenase (IDO), TGF-β, and prosta-
glandin E2 (PGE2) [50, 51]. Of course, TGF-β acts as a
two-edged sword. It can weaken the immune system and
thereby suppress liver inflammation [49]; on the other
hand, it can increase liver fibrosis [52]. MSCs, in fact,
can act as an immunomodulatory agent in reducing the
inflammation of the body through up-regulating anti-
inflammatory Treg cells and decreasing T helper 1 (1)
and 17 cells in ALF [53]. Moreover, the inflammation
after MSC transplantation can be indirectly stimulated
by up-regulating M2-type macrophages, leading to the
secretion of a variety of anti-inflammatory factors, such
as chemokine ligand 1 (CCL-1) and IL-10, up-regulation
2, and Treg cells [54]. Also, MSCs play an important
role in the reduction of B-cell growth through contact
between cells and the secretion of soluble factors [55].
Finally, MSC transplantation can play an effective role in
mitigating liver damage in ALF by decreasing the number
and activity of neutrophils in both peripheral blood and
the liver.
MSCs dierentiate intoHLCs
MSCs are characterized by their ability to proliferate and
differentiate invitro. For the first time, Friedenstein etal.
procured MSCs in 1968 from the bone marrow (BM) [56,
57]. After that, MSCs obtained from multiple sources,
making them an excellent supply of multipotent cells for
treatment of liver dysfunctions. A variety of methods are
used to differentiate MSCs into HLCs [58, 59]. Studies
show that multiple signals contribute to the regulation
of the cells’ behavior in a cooperative manner. Such sig-
nals are usually triggered by extracellular matrix (ECM),
growth factors, and even juxtacrine signals [60]. Each one
of the organs, as well as the developmental stage, is char-
acterized by a specific regulated timing and distribution
pattern of signals. As a result, achieving better results
in the case of in vitro cultures requires establishing a
type of environment that resembles the local environ-
ment. Based on the research previously done, it is pos-
sible to differentiate MSCs obtained from various sources
into hepatocytes in the case of both mice and humans
through the implementation of a variety of protocols and
methods invitro [61, 62].
Hepatic differentiation protocol is known to be the
most frequently used method for hepatic differentia-
tion, which benefits from Iscove’s Modified Dulbecco’s
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 5 of 21
Shokravietal. Stem Cell Research & Therapy (2022) 13:192
Medium (IMDM), as well as cytokine cocktail. Epider-
mal growth factor (EGF) or fibroblast growth factors
(FGF) trigger the MSCs to differentiate into endoder-
mal cells during the early induction stage. EGF prompts
the MSCs to proliferate by interfaces with EGF receptor
(EGFR) [63]. Besides, FGF is a member of a bigger fam-
ily that is comprised of seven polypeptides with similar
characteristics [64]. is family plays an essential role
in the primary stage of endodermal patterning [65].
In particular, FGF-4 and basic FGF (bFGF) are com-
monly used. Like EGF, FGF influences the growth rate
of MSCs [66].
Generally, the differentiation of cells is stimulated
by adding FGF, HGF, nicotinamide (NTA), and also
insulin-transferrin-selenium (ITS) into cultures [67].
As a mesenchymal origin pleiotropic cytokine, hepato-
cyte growth factor (HGF) contributes to adjustment of
growth, differentiation, and chemotactic migration of
MSCs [68]. MSCs’ exposure to HGF for a short time
causes the activation of c-Met receptors along with
its downstream agents such as extracellular signal-
regulated protein kinase (ERK)1/2, p38, mitogen-acti-
vated protein kinases (MAPKs), and phosphoinositide
3-kinase (PI3K) /Akt [69, 70]. MSCs’ exposure to
HGF for a long time will make changes in cytoskeletal
arrangement; moreover, it results in the migration of
cells and a notable decrease in proliferation. In addi-
tion, ITS and NTA promote the growth and survival of
primary hepatocytes [71].
Despite the fact that MSCs can differentiate in culture
through induction, the organ-specific microenvironment
is the best technique, enabling MSCs differentiation into
a certain cell type. e ability to express hepatocyte-
specific genes is one of the specific characteristics of
hepatic-differentiated cells, which can be affected by
microenvironmental features [72]. Reports display that
in the case of humans, the differentiation of the MSCs
obtained from umbilical cord (UC) into HLCs occurs
more quickly in the fibrotic liver microenvironment [73].
Other studies also show that the differentiation of
MSCs into functional hepatocytes does not occur
directly; rather, these cells initially differentiate into bil-
iary epithelial cells (BEC)-like cells, followed by differen-
tiation into HLCs [74]. However, according to the results
of other investigations, MSCs transdifferentiation infre-
quently occurs after MSC infusion in animal models
[75]. MSCs obtained from menstrual blood, for instance,
turned out to prevent hepatic satellite cells (HSCs) acti-
vation and resultant liver fibrosis, leading to the improve-
ment of liver function. Yet, very few transplanted MSCs
differentiated into functional HLCs in vivo [76]. ese
results demonstrate that the therapeutic impact of MSCs
is mediated mainly by indirect paracrine signaling.
MSCs antibrotic properties
anks to their antifibrotic and immunosuppressive
properties, MSCs play an important role in the treatment
of fibrosis [77]. Also, fibrosis in not a common pathologi-
cal signs of ALF; long-term liver damage mainly results
in fibrosis. MSC transplantation could attenuate liver
fibrosis by down-regulation of TGF-β1, Smad2, collagen
type I, and smooth muscle alpha-actin (αSMA), reduc-
ing liver fibrosis regions invivo [78]. Besides, BM-MSCs
decreased hepatic collagen distribution by impairing the
TGF-β/Smad signaling pathway in a cirrhosis rat models
[79]. MSCs also ameliorated hepatic microvascular dys-
function and portal hypertension, which contribute to
complications defining clinical decompensating [80]. Fur-
ther, the expression of matrix metalloproteinase (MMP)-
2, -9, -13, and -14 can be up-regulated by MSCs [81],
which, in turn, attenuates liver fibrosis through degrad-
ing extracellular matrix (ECM) [82]. MSCs reinforce this
effect by the down-regulation of the tissue inhibitors of
matrix metalloproteinases (TIMPs). Importantly, there is
an association between the balanced levels of MMPs and
TIMP and fibrosis resolution [83]. Moreover, MSCs have
both direct and indirect roles in inhibiting the activation
and growth of hepatic satellite cells (HSCs) and thereby
could inhibit collagen synthesis [84]. e direct inter-
actional relationship between MSCs and HSCs helps to
inhibit HSC proliferation by stimulating G0/G1 cell-cycle
arrest. is is done by inhibiting the phosphorylation of
ERK1/2 [85]. On the other hand, MSCs contain substan-
tial levels of milk fat globule-EGF factor 8 (MFGE8). e
MFGE8 reduces expression levels of TGF-β1 receptor on
HSCs, thus strikingly fences primary human HSCs acti-
vation [86]. In co-culture conditions, MSCs also mainly
impair α-smooth muscle actin (α-SMA) expression of
HSCs, which is mediated, in part, by the activation of the
Notch pathway [87]. e indirect secretion of some piv-
otal factors (IL-10, HGF, TGF-β, and TNF-α) by MSCs
averts the growth of HSCs and reduces the formation of
collagen. In contrast, HGF and NGF enhance the apopto-
sis of HSCs [88, 89].
MSCs antioxidant properties
One of the events deriving from ROS is oxidative stress,
which is known as a common driver in creating dam-
age to the liver. Some of these damages include the
liver failure, liver fibrosis, liver cirrhosis, viral hepatitis,
and also hepatocellular carcinoma (HCC) [90, 91]. e
results of some investigations have revealed that MSCs
play a strong mediatory antioxidant role in different
animal models [92, 93]. Oxidative liver injury is mostly
caused by thioacetamide (TAA) or carbon tetrachloride
(CCl4) as the most commonly used toxins. ese types
of toxins give rise to hepatocyte dysfunction through
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 6 of 21
Shokravietal. Stem Cell Research & Therapy (2022) 13:192
the peroxidation of lipid and proteins alkylation, nucleic
acids, and lipids [94, 95], resulting in the inflammatory
response, hepatocellular injury, and liver fibrosis. Cell
signaling and homeostasis require a low level of physi-
ologic ROS formed by the mitochondrial respiration.
MSCs have proved to have the capability of mitigating
oxidative stress simulated by CCl4 and TAA invivo [96,
97]. rough enhancing the superoxide dismutase (SOD)
expression and antioxidant response elements (AREs)
stimulation, MSCs boost antioxidant and cytoprotective
performance, leading to a reduction in hepatocyte apop-
tosis [98, 99]. Due to their antioxidant role along with
their role in modulating the immune systems, MSCs can
be very useful in developing therapies for liver injuries.
The importance ofMSCs‑exosome ascell‑free
approach inALF
Exosome is a main subtype of extracellular vesicles (EVs)
with a diameter in the range of 40–150 nm. Exosome
are mainly produced by a diversity of human cells, such
as stem/stromal cells, immune cells, and tumor cells
[100]. ey include several biological components, more
importantly, miRNAs, proteins, lipids and mRNAs, as
cargo [101, 102]. e production and secretion proce-
dure of exosome consists of three chief steps: (1) crea-
tion of endocytic vesicles through invagination of the
plasma membrane, (2) creation of multivesicular bodies
(MVBs) upon endosomal membranes’ inward budding,
and (3) incorporation of created MVBs with the plasma
membrane and releases of the vesicular contents termed
exosomes [14, 18]. en, the contents of exosomes are
transferred to the recipient cells, and thereby modify
physiological cells [15]. As a result of their great capa-
bilities to elicit hepatoprotective effect, exosomes are
recently been considered as a rational therapeutic option
for liver failure, thereby circumventing comprehensions
concerning stromal cells’ direct transplantation [103,
104]. ey are smaller and less complex compared with
parent cells, and thereby easier to produce and store.
Also, they exhibit no risk of tumor formation. Impor-
tantly, exosomes are less immunogenic than their par-
ent cells due to their lower membrane-bound proteins.
Recently, UC-MSCs-derived glutathione peroxidase1
(GPX1) enriched exosome showed the capacity to com-
promise oxidative stress as well as apoptosis in the hepat-
ocyte, stimulating hepatoprotective effect in ALF rodent
models [105]. Also, MSCs-derived exosome potently
reduced inflammatory response in ALF animal models
by impairment of IL-6-mediated signaling axis [106] and
also down-regulation of NLRP3 pathway [107]. However,
further studies are strongly needed to entirely elucidate
how MSCs-derived exosomes exert their hepatoprotec-
tive influences invivo.
MSCs inALF (animal studies)
Native MSCs
MSCs-based treatments have shown huge potential for
regenerating the liver and repairing its injury, which
resulted from several liver disorders (Tables1, 2). Invivo,
MSCs can migrate to damaged tissues and constrain the
production of pro-inflammatory molecules (e.g., IL-1,
IL-6, and TNF-ɑ) and ultimately potentiate liver cells
growth. As described, the chief mechanism behind the
MSCs-mediated positive effects is their immunoregula-
tory potential rather than direct differentiation into hap-
toid cells. ese cells efficiently hinder the activation of
both innate and adaptive immune system cells, such as
neutrophils, macrophages, NK cells, DCs, monocytes,
and also lymphocytes. Studies in liver failure animal
models revealed that MSCs could transdifferentiate into
albumin-expressing HLCs, and also may support nor-
mal hepatocytes proliferation in vivo upon fusion with
them [108]. Findings have outlined the important roles of
SDF-1/CXCR4 axis to ease MSCs migration to damaged
tissue, sustaining liver rescue in ALF [108]. As well, injec-
tion of MSCs-derived hepatocyte into mice with liver fail-
ure ameliorated liver function, as evidenced by analysis of
serum profile as well as biochemical factors rates [109].
Notably, the serum levels of TGF-β1 and IL-10 in trans-
planted animals were more prominent than in control
animals [109]. Other studies displayed that pyroptosis, a
unique shape of programmed cell death induced by pen-
etrating inflammatory reaction, was suppressed by MSCs
therapy in ALF preclinical model [110]. Accordingly,
MSCs administration caused liver repair in C57BL/6 mice
by up-regulation of IL-10 and concomitantly suppression
of NLRP3 [110]. Given that NLRP3 inflammasome elicits
liver failure through induction of procaspase-1 and pro-
IL-1 β accompanied with the adjustment of downstream
CD40-CD40L signaling, its inhibition as elicited by MSCs
can enable liver recovery in ALF [111]. Besides, the study
of the soluble factor produced by MSCs and their potent
desired impacts in a rat model of ALF revealed that
IL-10, which mainly is secreted by MSCs, has a central
role in ALF recovery post-transplantation [112]. It was
found that phosphorylated STAT3 diminished upon
IL-10 injection and conversely STAT3 suppression abro-
gated IL-10-induced effects in vivo, reflecting the emi-
nent role of STAT3 signaling in exerting IL-10-induced
anti-inflammatory influences [112]. In addition to the
IL-10, MSC-produced PGE2 could constrain apoptosis
and simultaneously augment hepatocyte proliferation,
thereby decreasing ALF [113]. In fact, PGE2 stimulated
YAP activation and then activated YAP suppressed phos-
phatase and tensin homolog (PTEN) and consequently
up-regulated mammalian target of rapamycin (mTOR),
a foremost controller of cell growth. is axis in turn
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 7 of 21
Shokravietal. Stem Cell Research & Therapy (2022) 13:192
protected versus ALF through increasing hepatocyte
proliferation [113]. Furthermore, there is clear evidence
signifying that MSCs could modify phenotype and func-
tion of macrophages, adjust DCs either differentiation or
maturation, and impede the T cell activities by the pro-
duction of tumor necrosis factor-alpha-stimulated gene 6
(TSG-6) in ALF models [114]. TSG-6 mainly averts the
inflammatory response as a result of suppressing P38 and
JNK signaling axes, providing a suitable milieu for ALF
rescue upon MSCs transplantation [115]. MSCs also can
induce their favored influences by heme oxygenase (HO)
1, a rate-limiting enzyme in heme metabolism, which is
noted as an effective antioxidative and cytoprotective
molecule. Recently, it was proven that MSCs administra-
tion gave rise to HO-1 up-regulation, whereas suppress-
ing HO-1 impaired MSCs-induced desired effects and
also hepatocyte autophagy [116]. ese favored effects
upon MSCs therapy were most probably caused by PI3K/
Akt signaling pathway-induced HO-1 up-regulation
[116]. Also, Zhang etal. found that systemic administra-
tion of BM-MSCs into the ALF rat model attenuated ALF
by up-regulation of the HO-1 expression and subsequent
attenuation in neutrophil infiltration and activation [117].
is event finally reduced hepatocyte apoptosis and also
improve their proliferation, culminating liver recovery.
Similarly, the pivotal role of neutrophils in ALF patho-
genesis has been clarified by other reports [118]. In the
D-galactosamine-induced ALF animal model, the great
number of neutrophils aggregated in the liver tissue along
with promoted myeloperoxidase (MPO) activity and
enhanced alanine aminotransferase (ALT) and aspartate
aminotransferase (AST) serum levels are mainly detected
[118]. Nonetheless, injection of BM-MSCs brought
about functional recovery, which was documented by
reduced ALT and AST levels and also improved sur-
vival rate in the treatment group compared with the
Table 1 Direct administration of native mesenchymal stromal cells (MSCs) in liver failure preclinical models, especially acute liver
failure (ALF)
Aspartate aminotransferase (AST), Alanine aminotransferase (ALT), Glycogen synthase kinase-3β (GSK-3β), Mammalian target of rapamycin (mTOR), Phosphoinositide
3-kinases (PI3Ks), CXC chemokine receptor 4 (CXCR4), Stromal derived factor-1α (SDF-1α or CXCL12), Prostaglandin E2 (PGE2), Nuclear factor-erythroid factor 2-related
factor 2 (Nrf2), Nuclear factor-kappa B (NF-κB), Natural killer T (NKT ) cells, T helper 17 (Th17), Interleukin-10 (IL-10)
Sources Model Result (ref)
Placenta Rat Migration to damaged site and induction of immunomodulatory effects by secreting paracrine factors in ALF [193]
Bone marrow Rat Systemic administration of MSCs reduced ALT, AST, and bilirubin levels [124]
Bone marrow Rat Reducing ALF, improving glucose metabolism and survival, and also stimulation of the hepatocyte proliferation by activating
AKT/GSK-3β/β-catenin pathway [122]
Adipose tissue Rat Normalization of amino acids, sphingolipids, and glycerophospholipids in the liver and blood along with attenuation hepato-
cyte apoptosis and conversely promoting their proliferation rate [194]
Placenta Rat Stimulation of liver repair through the antifibrotic and autophagic mechanisms [149]
Umbilical cord Monkey Inhibition of the activity of IL-6 producing monocyte, amelioration of the liver histology, and also animal survival [119]
Adipose tissue Rat Suppression of the secondary complications of liver failure [195]
Bone marrow Porcine Improving the liver function homeostasis, attenuation of reactive oxygen species (ROS) following efficient homing, and also
differentiation into hepatocytes [196]
Bone marrow Rat Amelioration of mitochondrial activities and normalization of lipid metabolism upon modifying the mTOR pathway [197]
Umbilical cord Rat Provoking the endogenous liver regeneration, hindrance of hepatocyte apoptosis by up-regulated c-Met in hepatocyte [120]
Bone marrow Rat Potentiating of MSCs-elicited liver regeneration following the abrogation of autophagy in MSCs [198]
Bone marrow Rat Amelioration of ALF by up-regulation of the heme oxygenase 1 (HO-1) expression, which resulted in inspiring the autophagy
process through PI3K/AKT signaling axis [116]
Bone marrow Mice Enhancing MSCs competencies to stimulate liver recovery following transdifferentiation as well as fusion with hepatocytes
by SDF-1/CXCR4 axis [199]
Bone marrow Mice Reducing ALF by IL-10 produced by MSCs, which ultimately inhibits pyroptosis [110]
Bone marrow Mice MSCs derived from adipose tissue showed superiority over MSCs isolated from bone marrow in ALF [125]
Bone marrow Mice Improvement of hepatocyte mediated by PGE2 released by MSCs, ameliorating ALF [113]
Wharton’s jelly Mice Restoration of hepatotoxicity by WJ-MSC [200]
Bone marrow Swine Averting ALF upon stimulation of hepatocyte proliferation and suppressing their apoptosis by intraportal MSCs transplanta-
tion [123]
Bone marrow Rat Attenuated aggregation and function of neutrophils [118]
Adipose tissue Mice Protection against ALF by affecting the Nrf2 and cytochrome P450 expression [201]
Umbilical cord Mice Inducing the endogenous liver regeneration but not notable hepatogenic differentiation [202]
Umbilical cord Mice Attenuation of ALF by down-regulation of MyD88/NF-κB pathway involved in inflammation [203]
Bone marrow Mice Attenuation of ALF by modifying ratio between Th17 and regulatory NKT cells [204]
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 8 of 21
Shokravietal. Stem Cell Research & Therapy (2022) 13:192
control group (50% vs 12.5%). Notably, the intervention
led to a robust decrease in the population of neutro-
phils in the liver, MPO function, and also the expression
of pro-inflammatory factors, including TNF-α, IL-1β,
interferon gamma (IFNγ) and CXC chemokine ligands
1/2 (CXCL1/2) [118]. In addition, in a monkey model of
ALF, systemic administration of the MSCs derived from
another source, unbiblical cord (UC), reduced hepatic
aggregation and maturation of circulating monocytes and
their IL-6 releases, resulted in prolonged survival [119].
UC-MSCs also could induce a reduction in ALF by pro-
voking the endogenous liver regeneration in association
with suppression of liver cell apoptosis by up-regulating
HGF/c-Met signaling axis [120] or down-regulation of
Notch and STAT1/STAT3 signaling [121]. e positive
influences of MSC therapy on hepatocyte proliferation
also may arise from activation of AKT/ glycogen synthase
kinase 3 beta (GSK-3β)/β-catenin pathway and enhance-
ment in glucose metabolism leading to improved survival
rate in ALF animal model [122]. Interestingly, intraportal
injection of MSCs showed superiority over hepatic intra-
arterial, intravenous, and intrahepatic injection in terms
of liver recovery rate in swine with ALF. Notably, the liver
recovery might be attributable to down-regulation of
caspase-3, up-regulation of apoptosis inhibitor survivin
as well as activation of AKT and ERK axes [123]. On the
other hand, another study revealed that systemic infusion
of MSCs was more effective than the intraperitoneal (IP)
injection to support liver recovery because of the more
significant increase in expression levels of growth factor
vascular endothelium growth factor (VEGF) [124]. Also,
compared with BM-MSC, adipose tissue (AT)-derived
MSCs displayed higher therapeutic capacities, as defined
by estimation of ALT and AST levels post-transplanta-
tion in ALF murine model [125].
MSCs delivery using biomaterials
Present cell transplantation approaches are hindered
via poor post-delivery survival, liver ECM and vascula-
ture deterioration, and also difficulties in fusion into the
host tissue [126]. As a result, scientists are persuaded
to deliver MSCs within biomaterial structure to sustain
the transplants’ viability and also potentiate MSCs long-
standing activation invivo [126].
Recent reports noted that BM-MSCs are valued options
to co-culture with hepatocytes in poly (lactic acid-gly-
colic acid) (PLGA) scaffolds, enhancing the hepatocel-
lular activities [127]. Administration of BM-MSCs and
hepatocyte seeded PLGA scaffolds led to the considerably
advanced cellular proliferation and conversely supported
Table 2 Administration of modified mesenchymal stromal cells (MSCs) or/and native MSCs in combination with other modalities in
liver failure preclinical models, especially acute liver failure (ALF)
CXC chemokine receptor 4 (CXCR4), Interleukin-1 (IL-1), Hepatocyte nuclear factor 4 alpha (HNF4α), Transforming growth factor (TGF-β), Vascular endothelial growth
factor 165 (VEGF165), Granulocyte colony-stimulating factor (G-CSF), Hepatocyte growth factor (HGF)
Sources Model Intervention Result (ref)
Adipose tissue Rat MSCs plus Eugenol Enhancing antifibrotic competencies of MSCs by eugenol
through down-regulation of TGF-β/Smad axis [205]
Bone marrow Rat MSC plus
Neutrophil depletion Amelioration of ALF in rats [206]
Umbilical cord Rat MSC plus Icaritin Enhancing the antiapoptotic capability of MSCs by promot-
ing the HGF/c-Met pathway [131]
Umbilical cord Mice HNF4α-overexpressing MSCs plus Hepatocyte Improving the EGF release by HNF4α-UMSCs [207]
Umbilical cord blood Rat VEGF165 -overexpressing MSCs Induction of marked therapeutic influences on ALF [143]
Bone marrow Mice CXCR4-overexpressing MSCs Improved migration and reduced damaged tissue by
stimulating hepatoprotective impacts [142]
Amniotic fluid Rat IL-1-overexpressing MSCs Improved liver function along with prolonged survival [145]
NA Swine MSCs plus IL-lRa-loaded chitosan nanoparticles Eliciting a synergistic impact by abrogating liver inflamma-
tion [136]
Bone marrow Rat Dexmedetomidine and Midazolam primed MSCs Enhancing the therapeutic merits of MSCs [208]
Umbilical cord Rat MSCs plus G-CSF Attenuation of liver damage by suppressing the genera-
tion of pro-inflammatory cytokines, alleviation of oxidative
stress, and reducing liver cell loss [132]
Bone marrow Swine MSCs plus IL-1R antagonism Exerting synergistic influences by prohibiting the inflamma-
tion and apoptotic signaling [135]
Bone marrow Mice MSCs seeded on human amniotic membranes (HAM) Improving survival rate [209]
Bone marrow Mice Poly lactic-co-glycolic acid (PLGA) scaffold loaded with
MSCs Stimulation of hepatoprotective impacts by paracrine fac-
tors [127, 128]
Bone marrow Mice Regenerated silk fibroin (RSF) scaffold loaded with MSCs Potentiating liver function by provoking angiogenesis [130]
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 9 of 21
Shokravietal. Stem Cell Research & Therapy (2022) 13:192
a striking reduction in ALT, AST, and total bilirubin in
ALF preclinical models post-transplantation, ultimately
leading to the prolonged survival [127]. Another study
demonstrated that MSCs seeded PLGA scaffolds were
survived for 3weeks, and displayed more evident activi-
ties than MSCs injected by intravenous route, which was
verified by lower mortality invivo [128]. However, there
was no significant alteration in hepatic inflammation and
necrosis zones between the two applied interventions
[128]. Also, poly L-lactic acid (PLLA) nanofiber scaffold
could improve the hepatic differentiation of BM-MSCs
[129]. Invitro, analysis exhibited that expression levels
of liver-specific markers, more importantly, albumin and
α-fetoprotein, were greater in differentiated cells on the
nanofibers compared with differentiated cells in plates.
ese results deliver the proof of the theory that engi-
neered PLLA scaffold could be an efficient alternative
to augment MSCs differentiation into functional HLCs
[129]. Besides, BM- and AT-MSC seeded regenerated silk
fibroin (RSF) matrices potently differentiated into HLCs
in vitro and also stimulated angiogenesis and restored
liver functions in the ALF mice model invivo [130].
Combination therapy withMSCs
A diversity of studies recently has focused on combina-
tion therapy with MSCs and other molecules or modali-
ties to diminish ALF. Meanwhile, co-administration of
MSCs with Icaritin, a well-known ingredient isolated
from traditional Chinese medicine, resulted in promis-
ing outcomes in vivo [131]. Indeed, MSCs co-cultured
with Icaritin improved survival, reduced serum levels of
AST and ALT, and elicited histological variations in liver
tissue more potently than MSCs alone. Importantly, the
up-regulation of HGF/c-Met by Icaritin was found to be
involved in MSCs-triggered antiapoptotic influences on
hepatocytes, reflecting the potential of herbal extracts
to promote MSC-mediated therapeutic impacts [131].
e addition of the granulocyte colony-stimulating fac-
tor (G-CSF) to UCB-MSCs also improved survival and
reduced ROS and pro-inflammatory cytokines expres-
sions in ALF murine model [132]. Also, intervention
engendered a significant reduction in cell apoptosis in
liver tissues more evidently than UCB-MSCs alone [132].
ese findings were similar to previous reports display-
ing that G-CSF therapy alone could significantly attenu-
ate short-term mortality in patients suffering from liver
failure mainly by reducing inflammation concomitant
with activating PI3K/Akt axis in hepatocytes [133, 134].
In another study, thanks to the crucial role of IL-1 in
the progress of ALF, Sang etal. evaluated possible syn-
ergetic effects of combined use of MSCs with 2 mg/kg
interleukin-1 receptor antagonist (IL-1Ra) invivo [135].
ey found that treatment significantly attenuated liver
cell apoptosis, improved their proliferation, and even-
tually enhanced animal survival. It is supposed that the
observed effects were dependent on enhancement in
AKT and also a reduction in nuclear factor (NF)-κB
expression, potentiating liver cell proliferation [135].
Similarly, co-administration of MSCs plus IL-1Ra chi-
tosan nanoparticles (NPs) was more effective than MSC
transplantation alone for treating ALF [136]. IL-1Ra-
loaded NPs administration by ear veins exhibited syner-
gistic impacts with portal vein injection of MSC in a mini
swine model of ALF by the hindrance of liver inflamma-
tion [136].
Pretreated MSCs
Current studies have verified that pretreatment
with chemical agents, hypoxia, and also cytokine or
chemokine invitro can improve the therapeutic merits of
MSCs upon transplantation invivo [137, 138]. Compared
to native MSCs, pretreated MSCs largely demonstrate
developed hepatogenic differentiation, homing capability,
and survival and paracrine impacts.
In 2019, Nie etal. suggested that IL-1β pretreatment
could circumvent the MSC’s poor migration toward the
injured region in ALF murine model [139]. Correspond-
ingly, IL-1β-MSCs showed superiority over native MSCs
respecting the survival time and liver function in vivo.
Remarkably, IL-1β-MSCs suppressed liver cell apoptosis
and necrosis and also provoked their proliferation. Pre-
ferred effects were most probably enticed by improved
CXCR4 expression resulting from IL-1β pretreatment
and thereby increased migration toward CXCL12 (SDF-1
α) in damage tissue [139]. Interestingly, pretreatment
with injured liver tissue might improve MSCs homing
and also their hepatogenic differentiation [140]. Invivo,
transplantation of pretreated MSCs led to an enhance-
ment in albumin, cytokeratin 8, 18, and antiapoptotic
protein Bcl-xl levels, whereas supported a reduction in
pro-apoptotic protein Bax and caspase-3 levels [140].
Likewise, short-term, but not long-term, sodium butyrate
(NaB) treatment augmented hepatogenic differentiation
of BM-MSCs and consequently alleviated liver injury
invivo, according to Li etal. reports [141].
Genetically modied MSCs
Genetically modified MSCs mainly are used to enhance
their colonization rate post-transplantation, leading to
ameliorated liver recovery in ALF. Meanwhile, geneti-
cally modified MSC to overexpress the CXCR4 gene
demonstrated more appropriate migration capability
toward SDF-1α and also induce better hepatoprotective
impacts invitro [142]. In ALF murine model, CXCR4-
MSCs efficiently migrated to damaged tissue, and in
turn, brought about prolonged survival and restored
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 10 of 21
Shokravietal. Stem Cell Research & Therapy (2022) 13:192
liver activity more prominent than native MSCs trans-
plantation [142]. Besides, UCB-MSCs modified to
overexpress vascular endothelial growth factor 165
(VEGF165), a strong pro-angiogenic factor, potenti-
ated the UCB-MSCs multipotency and also resulted in
better homing and colonization in liver tissues post-
transplantation [143]. While both native UCB-MSCs
and VEGF165-encoding UCB-MSC restored liver activ-
ity in the ALF rat model, modified stromal cells exhib-
ited more desired therapeutic influences on ALF [143].
Given that IL-35 plays a pivotal role in Treg-induced
immunoregulation, Wang etal. evaluated the therapeu-
tic merits of IL-35 overexpressing MSCs in ALF [144].
ey showed that modified stromal cells migrated
to the damaged tissues and considerably reduced
the necrosis zones of damaged livers. Moreover, IL-
35-MSCs averted hepatocyte apoptosis through down-
regulation of the FASL expression by immune cells.
ey also attenuated IFN-γ levels secreted by immune
cells potently via targeting JAK1-STAT1/STAT4 signal
pathway [144]. As described in previous sections, IL-
1Ra elicits strong anti-inflammatory and antiapoptotic
impacts on immune response in liver failure. Accord-
ingly, Zheng and coworkers showed that transplanta-
tion of IL-1Ra-encoding amniotic fluid (AF)-MSCs by
the portal vein in the ALF rat model led to reduced
mortality as well as ameliorated liver activity [145].
MSCs‑exosome inALF
Exosomes are small membrane-bound EVs that are pro-
duced and then released by numerous types of cells,
such as stem/stromal cells, immune cells, or tumor cells.
Exosomes are comprised of a myriad of biological com-
ponents, including proteins, lipids, mRNAs as well as
miRNAs as cargo, which can be conveyed to the recipi-
ent cells [103]. Such cargo can adjust physiological cell
functions and thereby adapt tissue microenvironment,
and also inspire hepatocyte proliferation, reflecting their
competencies to be described as a rational therapeutic
option in liver diseases, such as ALF (Table3). Reduced
levels of miR-20a-5p accompanied with the enhanced
level of CXCL8, most eminent neutrophil chemoattract-
ants, are mainly observed in hepatocytes during ALF.
But, BM-MSCs-exosome could improve miR-20a-5p
expression and conversely attenuate CXCL8 levels in
hepatocytes [146]. Also, systemic injection of UC-MSC-
exosome (16 mg/kg) induced liver restoration in the
ALF mice model [105]. It was found that glutathione
peroxidase1 (GPX1) enriched exosome-mitigated oxi-
dative stress and apoptosis in the hepatocyte, while the
elimination of GPX1 led to the abrogated UC-MSCs-
exosome-elicited hepatoprotective impacts in mice [105].
In addition, UC-MSC-exosomes potently modified the
membranous expression of CD154 (or CD40 ligand) in
intrahepatic CD4+ T cells, largely contributing to the
inflammatory response in the liver [147]. e suppressive
Table 3 Mesenchymal stromal cells (MSCs) derived molecules (e.g., exosome) in liver failure preclinical models, especially acute liver
failure (ALF)
Silica magnetic graphene oxide (SMGO), NLR family pyrin domain containing 3 (NLRP3), Tumor necrosis factor-ɑ (TNF-ɑ), T helper 1/2 (Th1/2), Vascular endothelial
growth factor (VEGF), Interleukin 8 (IL-8 or CXCL8), Conditioned medium (CM), Embryonic stem cells (ESCs), Glutathione peroxidase1 (GPX1), C-reactive protein (CRP)
Sources Model Intervention Result (ref)
Umbilical cord Mice MSCs-exosome GPX1 enriched exosomes diminished oxidative stress and also apoptosis [105]
Placenta Rat MSCs-exosome CRP enriched exosome provoked angiogenesis by up-regulation of Wnt signaling axis [149]
Bone marrow Rat MSCs-exosome Stimulation of hepatoprotective impacts by exosome-rich fractionated secretome [150]
Bone marrow Mice MSCs-exosome Suppression of NLRP3 in macrophage and thereby reducing ALF by TNF-ɑ pretreated exo-
some [107]
Menstrual blood Mice MSCs-exosome Liver function recovery, improved survival rates, and suppressed hepatocellular apoptosis
[151]
Umbilical cord Mice MSCs-extracellular vesicles Inhibition of T cell activation in liver tissue following reserve of CD154 expression [147]
Bone marrow Mice MSCs-conditioned medium Promoting hepatocyte proliferation, inhibition of their apoptosis, hindrance of the infiltration
of macrophages, improving Th2/Th1 ratio, and enabling hepatic stellate cell (HSC) loss [157]
Bone marrow Rat MSCs-conditioned medium Marked attenuation of panlobular immune cells infiltrates and also hepatocellular apoptosis
[210]
ESCs-MSCs Mice MSCs-conditioned medium Supporting hepatocytes growth by VEGF enriched conditioned medium [156]
Bone marrow Mice MSCs-exosome Attenuation of liver inflammation by exosomal miR-20a-5p/intracellular CXCL8 axis [146]
Bone marrow Rat MSCs-conditioned medium Reduced hepatocyte apoptosis [154]
Bone marrow Rat MSCs-conditioned medium Improving the hepatoprotective impacts of the conditioned medium by SMGO potently
elicited through inhibition of inflammation and loss of hepatocytes [155]
Amniotic fluid Mice MSCs-conditioned medium Hepatic progenitor-like (HPL)-CM showed superiority over amniotic fluid-MSCs in terms of
liver recovery [158]
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 11 of 21
Shokravietal. Stem Cell Research & Therapy (2022) 13:192
effect on CD154 expression and resultant inflamma-
tion was due to the existence of chaperonin containing
TCP1 subunit 2 (CCT2) in these exosomes, which tar-
gets Ca2 + influx and down-regulates CD154 genera-
tion in CD4 + T cells [147]. In another study, Shao etal.
screened the miRNAs in the MSCs-exosomes compli-
cated in inhibition of IL-6-mediated signaling axis in ALF
mice model. ey showed that miR-455-3p was released
by exosomes and efficiently instigated PI3K signaling,
and in turn, sustained hepatocyte proliferation [106].
Also, IL-6 pretreated MSCs or exosomes exhibited higher
levels of miR-455-3p compared with native MSCs or
their derivative exosome. In fact, miR-455-3p-enriched
exosomes suppressed macrophages activation, reduced
local liver injury, and also diminish the expression of pro-
inflammatory cytokines in vivo [106]. e miR-455-3p
also could constrain activation of HSCs and liver fibrosis
upon down-regulation of the heat shock protein (HSP)
47/TGF-β/Smad4 signaling pathway [148]. Importantly,
C-reactive protein (CRP) enriched placenta-derived mes-
enchymal stromal cells (PD-MSCs)- exosome could up-
regulate Wnt signaling pathway as well as angiogenesis
in animal hepatocytes by interacting with endothelial
cells [149]. Another study also revealed that rat BM-
MSCs-exosome-rich fractionated secretome could bring
about a hepatoprotective impact in ALF models mainly
caused by diminished oxidative stress [150]. Similarly,
transplantation of exosomes derived from menstrual
blood-mesenchymal stromal cells (Men-SCs) that con-
tained a diversity of cytokines, such as intercellular adhe-
sion molecule-1 (ICAM-1 or CD54), angiopoietin-2, Axl,
angiogenin, insulin-like growth factor-binding protein
6 (IGFBP-6), osteoprotegerin, IL-6, and IL-8, improved
liver function in the ALF animal model [151]. Treatment
resulted in improved survival rates as well as reserved
hepatocyte apoptosis. Notably, attenuated numbers of
neutrophils and also diminished levels of caspase-3 were
evidenced post-transplantation, assuming that Men-SC-
exosome can be a substitute treatment to support liver
failure [151]. Pretreatment of UC-MSCs-exosome with
TNF-α also enhanced exosome-induced hepatoprotec-
tive influence in the ALF mice model [107]. Pretreated
exosomes led to the attenuated serum ALT, AST, and
pro-inflammatory cytokines levels and concomitantly
down-regulated stimulation of NLRP3 inflammasome.
Molecular analysis revealed that miRNA-299-3p up-reg-
ulated in TNF-α-primed MSCs-exosome played an emi-
nent role in the amelioration of liver damage in ALF by
blocking the NLRP3 pathway [107]. Apart from its role
in liver failure recovery, a miR-299-3p activity as a potent
tumor suppressor has been documented in hepatocellu-
lar carcinoma by alleviating tumor size and venous infil-
tration [152].
MSCs-conditioned medium (CM) could also mod-
ify morphological characteristics of hepatocytes in the
ALF model. Meanwhile, secretome achieved by culti-
vating MSCs with low oxygen content (10%) provoked
more prominent hepatoprotective influence, and sig-
nificantly reduced ALT and AST and also pro-inflamma-
tory cytokines serum levels following injection in vivo
[153]. In another study, Li and coworkers evaluated the
therapeutic merits of CM from MSCs co-cultured with
hepatocytes in the ALF rat model [154]. e apoptotic
cells frequency was lower in CM derived from co-cul-
tured cells than MSCs-CM or hepatocyte-CM. Also, CM
derived from co-cultured cells strikingly alleviated liver
injury and facilitated liver recovery, indicating the advan-
tages of this strategy for liver failure therapy [154]. Also,
silica magnetic graphene oxide (SMGO) could enhance
the hypo protective influences of MSC-CM in ALF
in vivo [155]. Meanwhile, administration of 300 μg/kg
SMGO boosted MSC-CM competencies to avert necro-
sis, apoptosis, and inflammation of hepatocytes. Besides,
SMGO therapy up-regulated the expression of VEGF
and matrix metalloproteinase-9 (MMP-9) invitro [155].
Another report also demonstrated that administration
of CM from embryonic stem cell (ESC)-derived MSCs
potentiated the proliferation of primary hepatocyte and
improved IL-10 secretion from immune cells in vivo
[156]. It appeared that such events might arouse because
of the existence of VEGF in ESC-MSC-CM, which affect
hepatocytes proliferation and migration, generating new
avenues to cure ALF [156]. Likewise, MSC-CM sus-
tained hepatocytes proliferation, reduced their apop-
tosis, compromised macrophages infiltration, elevated
2 and Treg cells population, decreased levels of 17
cells population, and eventually enabled HSCs death in
ALF preclinical model [157]. e MSC-CM injection
caused glycogen synthesis and storage recovery and also
ameliorated ALF with no effect on 1 cells [157]. Also,
CM achieved from either amniotic fluid (AF)-MSCs or
hepatic progenitor-like (HPL) cells derived from AF-
MSCs thanks to the presence of IL-10, IL-1Ra, IL-13, and
IL-27 stimulated liver recovery in the mice model with
ALF [158].
MSCs inliver‑associated conditions (clinical trials)
Several clinical trials have been accomplished or are
ongoing to address the safety, feasibility and efficacy of
MSCs therapy in liver-associated conditions, most fre-
quently in liver failure and liver cirrhosis (Table4, Fig.2).
BM-MSCs and UC-MSCs are most commonly used types
of cells. Of course, there is still no definite standard for
which source of MSCs should be applied for clinical use.
It seems that UC-MSCs are preferred for liver failure
treatment as a result of higher differentiation capability.
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 12 of 21
Shokravietal. Stem Cell Research & Therapy (2022) 13:192
Also, the immunogenicity of UC-MSCs is lower than that
of BM-MSCs [159, 160]. Hence, autologous BM-MSCs
and allogeneic UC-MSCs are highly preferred. On the
other hand, poor proliferation, anti-inflammatory and
self-renewal ability impedes AT-MSCs application in
clinic [161]. Although intravenous injection is most used
route, intraportal administration is evidently the optimal
route for MSC therapy in liver-associated conditions due
to the faster engraftment and the prohibited off-target
accumulation. However, we must assess patients’ condi-
tions and the potential risk of applying a particular route
before choosing the administration route.
Liver failure
A study of the safety and preliminary efficacy of UC-MSC
transplantation (3 times at 4-week intervals) was car-
ried out by Ming and colleagues [162]. ey showed that
the intervention had no unwanted effects, while attenu-
ated total bilirubin and ALT levels, prolonged survival
rate, and finally ameliorated liver functions, as evidenced
by improved serum albumin, and prothrombin activity
[162]. As well, intrasplenic and intrahepatic administra-
tion of autologous BM-MSCs derived hepatocyte inspired
short-term amelioration in patient’s ascites, lower limb
edema, and serum albumin [163]. Of course, defining the
life span of the transplanted cells, and also determining
the presence of long-term side effects is urgently required
[163]. Moreover, another trial, which was accomplished
from 2010 to 2013, indicated that systemic administra-
tion of allogeneic BM-MSCs could exert therapeutic
benefits in patients suffering from HBV-related LF [164].
Meanwhile, stromal cell therapy augmented serum total
bilirubin and ultimately promoted the 24-week survival
rate by stimulating liver rescue concomitant with lessen-
ing the occurrences of stern infections compared with the
control group (16.1% versus 33.3%) [164]. Likewise, other
trials also exhibited that autologous BM-MSC transplan-
tation was safe for chronic HBV-induced LF patients, as
Table 4 Clinical trials based on MSCs-based therapies in liver diseases (e.g., ALF)
Gamma-glutamyl transferase (GGT), Alkaline phosphatase (ALP), Bone marrow (BM), Umbilical cord (UC), Umbilical cord blood (UCB), Adipose tissue (AT), Hepatitis C
virus (HCV), Hepatitis B virus (HBV )
Condition Cell Source Participant no Main results (ref )
Primary biliary cirrhosis Allogeneic UC 7 Robust attenuation in serum ALP and GGT levels [168]
Liver failure Allogeneic UC 43 Enchantment in the survival rates without side effects [162]
HBV-induced liver cirrhosis Autologous BM 56 Improving the Treg/Th17 cell ration [171]
Liver cirrhosis Autologous BM 25 Removing the HCV RNA caused by transplanted MSCs-medi-
ated paracrine effect [211]
Decompensated liver cirrhosis Autologous BM 4 Improved the quality of life without serious side effects [167]
Alcoholic liver cirrhosis Autologous BM 12 No side effects in concomitant with histological and quantita-
tive amelioration [212]
HCV-induced liver cirrhosis Autologous BM 40 Normalization of liver enzymes levels in association with resto-
ration in liver function [213]
Liver cirrhosis Autologous BM 8 Improved liver function evidenced by enhanced serum albumin
and reduced total bilirubin[172]
Liver failure Autologous BM-derived hepatocyte 40 Improvement in ascites, lower limb edema as well as serum
albumin levels [163]
Decompensated liver cirrhosis Allogeneic UC 45 Improved level function documented with enhanced serum
albumin levels and reduced total bilirubin levels [180]
HCV-induced liver cirrhosis Autologous BM 20 Amelioration of liver function in Egyptian patients [170]
HCV-induced liver cirrhosis Autologous BM 25 Partial rescue in liver function [169]
Decompensated liver cirrhosis Autologous BM 27 No significant beneficial effect [174]
Liver failure Allogeneic BM 110 Improved overall survival and also reduced incidence of severe
infections [164]
Liver cirrhosis Allogeneic (UC, UCB, BM) 26 Stromal cell injection by peripheral vein was safe and partially
effective [173]
HBV-induced liver cirrhosis Allogeneic UC 40 Enhanced IL-10 levels and also reduced IL-6 and TNF-ɑ levels
[214]
Ischemic-type biliary lesions
following liver transplantation Allogeneic UC 12 Stem cell injection was safe and elicited favorable short-term
outcomes [215]
Alcoholic liver cirrhosis Autologous BM 72 Ameliorated histologic fibrosis and liver normal activity [216]
Liver allograft rejection Allogeneic UC 27 Improved Treg/Th17 cell ratio [217]
Liver allograft rejection Allogeneic BM 10 No side effect [218]
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 13 of 21
Shokravietal. Stem Cell Research & Therapy (2022) 13:192
shown by the incidence of no serious intervention-related
events and carcinoma during 192weeks follow-up [165].
Also, the short-term outcome was promising; however,
long-term efficacy was not clearly amended [165].
Liver cirrhosis
Cirrhosis is a late-stage liver disease in which healthy
liver tissue is substituted with scar tissue and the liver is
perpetually damaged. Liver transplantation is a standard
therapeutic plan aiming to treat liver cirrhosis patients
[166]. Meanwhile, MSCs have recently been noted as a
possible therapeutic option to partially ameliorate liver
function in this condition as a result of their appreciated
antifibrotic and immunoregulatory attributes [55].
A phase 1 trial on 4 patients with decompensated liver
cirrhosis verified the safety and feasibility of MSCs ther-
apy [167]. Moreover, the life quality of all patients was
ameliorated post-transplantation concerning the mean
physical and mental component scales [167]. In primary
biliary cirrhosis patients, UC-MSC injection by periph-
eral vein (3 times at 4-week intervals) exhibited no seri-
ous untoward effects [168]. Also, intervention caused a
Fig. 2 Clinical trials based on mesenchymal stromal cells (MSCs) therapy in liver-associated conditions registered in ClinicalTrials.gov (November
2021). The schematic demonstrates clinical depending on the study phase (A), study status (B), MSCs source (C), study location (D), participant
number (E), and condition (F)
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 14 of 21
Shokravietal. Stem Cell Research & Therapy (2022) 13:192
robust reduction in serum alkaline phosphatase (ALP)
and γ-glutamyltransferase (GGT) levels compared to the
control group during 4years follow-up. Notwithstand-
ing, no alteration was observed in levels of ALT, AST,
total bilirubin, albumin, INR, or the prothrombin time
activity. ereby, comprehensive randomized controlled
cohort trials are justified to authorize the clinical merits
of UC-MSC transplantation [168]. In addition, injection
of autologous BM-MSCs led to a partial amelioration
of liver function in 25 Egyptian patients suffering from
HCV-triggered liver cirrhosis, as evinced by improved
prothrombin activity and serum albumin levels along
with reduced bilirubin level [169]. In a similar condi-
tion, Amin etal. found that intrasplenic administration
of autologous BM-MSCs potentiated liver function with
attenuation in total bilirubin, AST, ALT, prothrombin
time (PT), and also INR levels [170]. Autologous BM-
MSCs therapy also inspired an improvement in liver
function among HBV-related liver cirrhosis patients fol-
lowing transplantation [171]. is trial was conducted in
56 patients with HBV-induced liver cirrhosis, and results
showed an enhancement in Treg/17 ratio post-trans-
plantation during 24-week follow-up [171]. Consistently,
mRNA levels of forkhead box protein P3 (FOXP3), an
eminent Treg-associated transcription factor, strikingly
were diminished, whereas retinoic acid-related orphan
receptor gamma t (RORγt) expression levels which are
tightly in association with 17 cells were reduced. Fur-
ther, the intervention resulted in an enhancement in
TGF-β levels, while IL-17, TNF-α, and IL-6 were signifi-
cantly decreased following transplantation [171]. In con-
trast to several cited trials implying that the autologous
MSC therapy can be a safe and effective alternative for
patients with liver cirrhosis [172, 173], Mohamadnejad
etal. noted that MSC infusion by peripheral vein had no
advantageous result in cirrhotic patients [174]. Overall,
large-scale studies are required to achieve reliable results
concerning MSCs therapy in liver cirrhosis.
Potential risks ofMSC transplantation
e treatment of liver dysfunctions through MSCs has
been the central aim of several clinical and preclini-
cal investigations. In this context, a few issues need to
be dealt with cautiously (e.g., the possible emergence of
carcinogenesis and the transmission of the virus). Dif-
ferent growth factors can be secreted by MSCs, and this
may stimulate the growth of tumor cells and angiogenesis
[175, 176]. e past experimental investigations showed
that the number of passages is a defining factor in ren-
dering a tissue malignant or cancerous. Studies show that
chromosome abnormalities may occur after more than
three passages in the MSCs of mice [177, 178]. Moreover,
MSCs are likely to experience telomeric deletions after
a multitude of passages. Despite the lack of any clini-
cal reports on the malignant transformation of human
MSCs, the follow-up period was not long enough for the
formation of a tumor for most of them [179]. As a result,
there need to be more studies on chromosomal integrity
before MSCs transplantation to make sure that the pro-
cedure is completely safe.
Contrary to autotransplantation, allotransplantation
can pose the danger of the spread of the virus to the
patients [180]. Even though the spread of parvovirus B19
into BM cells was observed invitro, there is no confirmed
case of parvovirus B19-positive MSC-related viremia
in humans. Yet, we do not know the spread of the her-
pes simplex virus (HSV) and cytomegalovirus (CMV)
via MSCs invivo. Owing to these facts, recipients, and
donors of MSC are recommended to be screened for
parvovirus B19, HSV, and CMV, as immunosuppressed
patients are likely to catch infectious [181].
Enhancing thequantity ofMSCs‑secreted
molecules
Now, restricted secretion of soluble mediators, such as
exosome, from parental MSCs fences their wide-ranging
application in clinics. Following some passages, MSCs
mainly demonstrates abrogated competence to produce
and then release soluble factor. Recent studies have indi-
cated that tangential flow filtration (TFF) system-based
tactics support the secretion of greater levels of vesi-
cles from origin stromal cells than vesicle isolation by
ultracentrifuge [182]. Further, ultrasonication of MSC-
derived extracellular vesicles could improve their yields
up to 20-fold [183]. Other proofs are indicating that three
dimensional (3D) culture may facilities the incessant pro-
duction of MSC-derived exosome [184, 185]. Cultivation
of MSCs in 3D cultures together with conventional either
differential ultracentrifugation or TFF also could engen-
der a higher quantity of MSCs-derived secretome [186].
Also, MSCs culture on particular biomaterials, such as
alginate hydrogel [187] and avitene ultrafoam collagen
ease generation of exosome with higher quantity and
also potency [188]. As well, pretreatment of MSCs with
hypoxia or various molecules, in particular cytokines or
chemokines (e.g., IFN-γ, TNFα, IL-1β, IL-6, and TGF-β),
gives largely rise to the secretion of vesicles with greater
regenerative competencies [189191].
Conclusion andfuture direction
Some investigations in preclinical models of liver dis-
eases, such as ALF, have verified the MSC’s unique com-
petence to establish hepatocyte in vivo. Nonetheless, it
seems that the therapeutic merits of MSCs largely rely
on their aptitudes to secrete a myriad of factors, more
importantly, cytokines, growth factors, and miRNAs,
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 15 of 21
Shokravietal. Stem Cell Research & Therapy (2022) 13:192
facilitating liver recovery. During last two decades, vari-
ous clinical trials have been conducted to evaluate the
capability of MSCs therapy in liver-associated condi-
tions, such as ALF (Table4, Fig. 2); however, achieved
outcomes are quite inconsistent. Given that autologous
MSCs derived from elder patients or patients with obe-
sity experienced abrogated proliferation and differen-
tiation capability, using allogeneic cells in some cased is
urgently required. In this circumstance, screening recip-
ients and donors of MSC for parvovirus B19, HSV, and
CMV are of paramount importance. Taken together, the
providing of a universal MSC quality standard evaluation
system is required.
To determine the mechanism contributed to MSCs
therapy, it is urgently required to determine the pro-
tein, DNA and RNA secreted by MSCs. e proteomics
and transcriptomics can play a pivotal role in evaluating
the underlying mechanism. Notably, improving the fre-
quency of cells homing to the damaged liver is the central
point to potentiate the therapeutic impacts of MSCs. In
fact, investigation of the homing attributes of MSCs is of
paramount importance to augment the effective thera-
peutic quantity of such cells. In published clinical results,
MSCs have been administrated into patients by several
available routes, more frequently intravenous routes fol-
lowed by intrahepatic injection (e.g., by the portal vein
and hepatic artery). Also, intrasplenic injection has been
applied in a few studies. Based on findings, a remarkable
number of cells are trapped in the lungs upon systemic
injection and thereby did not move to the liver afterward.
Hence, finding better administration route is recom-
mended to achieve significant outcome in vivo. Mean-
while, a study indicated that intraportal injection was
more effective than hepatic intra-arterial injection and
also intravenous injection to restore liver injury invivo
[123]. As well, it has been shown that portal vein injec-
tion has superiority over intrasplenic injection [192]. On
the other hand, other reports exhibited that injection by
the hepatic artery was not beneficial for the transdiffer-
entiation of MSCs.
Among the recent clinical trials concerning the MSCs
therapy for liver diseases (e.g., liver failure) treatment,
the total number of MSCs employed was from 106 to
109, irrespective of which method was applied to deliver
the dose. e large range of doses applied is difficult to
explicate as there are few reports including comparisons
of several doses in the same clinical trial. Nonetheless, it
seems that as few as 1 × 107 cells can be helpful based on
recent published results.
In sum, although clinical trials have evidenced the
safety and modest efficacy of short-term application of
MSCs, further trials are warranted before MSCs appli-
cation in clinical to treat ALF and other liver-associated
conditions for optimizing administration routes as well
as dosses.
Abbreviations
MSCs: Mesenchymal stromal cells; ALF: Acute live failure; HGF: Hepatocyte
growth factor; TNF α: Tumor necrosis factor α; IFNγ: Interferon gamma; VEGF:
Vascular endothelial growth factor; BM: Bone marrow; UC: Umbilical cord; AT:
Adipose tissue; miRs: MicroRNAs; TGFβ: Transforming growth factor β; AST:
Aspartate aminotransferase; ALT: Alanine aminotransferase; IL: Interleukin.
Acknowledgements
Not applicable.
Author contributions
All authors contributed to the conception and the main idea of the work. SS,
BAZ, VB, FN, RH, MMK, LT, AS, and SM drafted the main text, figures, and tables.
AZ and SM supervised the work and provided the comments and additional
scientific information. SS and BAZ and AS also reviewed and revised the text.
All authors read and approved the final manuscript.
Funding
No Funders.
Availability of data and materials
Not applicable.
Declarations
Ethics approval and consent to participate
Not applicable.
Consent for publication
Not applicable.
Competing interests
The authors declare that they have no competing interests.
Author details
1 Department of Research and Academic Affairs, Larkin Community Hospital,
Miami, FL, USA. 2 I.M. Sechenov First Moscow State Medical University (Sech-
enov University), Moscow, Russian Federation. 3 Basic Sciences Department,
College of Pharmacy, University of Duhok, Duhok, Kurdistan Region, Iraq.
4 School of Medicine, Abadan University of Medical Sciences, Abadan, Iran.
5 Department of Medicinal Chemistr y, Pharmacy Faculty, Tabriz University
of Medical Sciences, Tabriz, Iran. 6 Department of Oral and Maxillofacial Sur-
gery, School of Dentistry, Shahid Beheshti University of Medical Sciences, Teh-
ran, Iran. 7 Department of Pharmacology, Saveetha Dental College, Saveetha
Institute of Medical and Technical Science, Saveetha University, Chennai, India.
8 Student Research Committee, Tabriz University of Medical Sciences, Tabriz,
Iran. 9 Immunology Research Center, Tabriz University of Medical Sciences,
Tabriz, Iran. 10 Stem Cell Research Center, Tabriz University of Medical Sciences,
Tabriz, Iran.
Received: 19 November 2021 Accepted: 28 February 2022
References
1. Hadem J, Tacke F, Bruns T, Langgartner J, Strnad P, Denk GU, Fikatas P,
Manns MP, Hofmann WP, Gerken G. Etiologies and outcomes of acute
liver failure in Germany. Clin Gastroenterol Hepatol. 2012;10(6):664-9.
e2.
2. Lee WM, editor. Acute liver failure. Seminars in respiratory and critical
care medicine. New York: Thieme Medical Publishers; 2012.
3. Blackmore L, Bernal W. Acute liver failure. Clin Med. 2015;15(5):468.
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 16 of 21
Shokravietal. Stem Cell Research & Therapy (2022) 13:192
4. Patton H, Misel M, Gish RG. Acute liver failure in adults: an evidence-
based management protocol for clinicians. Gastroenterol Hepatol.
2012;8(3):161–212.
5. Lee WM. Acute liver failure in the United States. Semin Liver Dis.
2003;23(3):217–26.
6. Tao Y-C, Wang M-L, Chen E-Q, Tang H. Stem cells transplantation in
the treatment of patients with liver failure. Curr Stem Cell Res Ther.
2018;13(3):193–201.
7. Schacher FC, Martins Pezzi da Silva A, Silla LMDR, Álvares-da-Silva MR.
Bone marrow mesenchymal stem cells in acute-on-chronic liver failure
grades 2 and 3: a phase I-II randomized clinical trial. Can J Gastroenterol
Hepatol. 2021;2021:1–9.
8. Choi J, Kang S, Kim B, So S, Han J, Kim G-N, Lee M-Y, Roh S, Lee J-Y, Oh
SJ. Efficient hepatic differentiation and regeneration potential under
xeno-free conditions using mass-producible amnion-derived mesen-
chymal stem cells. Stem Cell Res Ther. 2021;12(1):1–20.
9. Wang J-l, Ding H-r, Pan C-y, Shi X-l, Ren H-z. Mesenchymal stem cells
ameliorate lipid metabolism through reducing mitochondrial damage
of hepatocytes in the treatment of post-hepatectomy liver failure. Cell
Death Dis. 2021;12(1):1–15.
10. Marofi F, Vahedi G, Hasanzadeh A, Salarinasab S, Arzhanga P, Khademi B,
Farshdousti HM. Mesenchymal stem cells as the game-changing tools
in the treatment of various organs disorders: Mirage or reality? J Cell
Physiol. 2019;234(2):1268–88.
11. Saito Y, Ikemoto T, Morine Y, Shimada M. Current status of hepatocyte-
like cell therapy from stem cells. Surg Today. 2021;51(3):340–9.
12. Markov A, Thangavelu L, Aravindhan S, Zekiy AO, Jarahian M, Chartrand
MS, Pathak Y, Marofi F, Shamlou S, Hassanzadeh A. Mesenchymal stem/
stromal cells as a valuable source for the treatment of immune-medi-
ated disorders. Stem Cell Res Ther. 2021;12(1):1–30.
13. Tavakoli S, Ghaderi Jafarbeigloo HR, Shariati A, Jahangiryan A, Jadidi F,
Jadidi Kouhbanani MA, Hassanzadeh A, Zamani M, Javidi K, Naimi A.
Mesenchymal stromal cells; a new horizon in regenerative medicine. J
Cell Physiol. 2020;235(12):9185–210.
14. Moghadasi S, Elveny M, Rahman HS, Suksatan W, Jalil AT, Abdelbasset
WK, Yumashev AV, Shariatzadeh S, Motavalli R, Behzad F, et al. A para-
digm shift in cell-free approach: the emerging role of MSCs-derived
exosomes in regenerative medicine. J Transl Med. 2021;19(1):302.
15. Hassanzadeh A, Rahman HS, Markov A, Endjun JJ, Zekiy AO, Chartrand
MS, Beheshtkhoo N, Kouhbanani MAJ, Marofi F, Nikoo M. Mesenchymal
stem/stromal cell-derived exosomes in regenerative medicine and
cancer; overview of development, challenges, and opportunities. Stem
Cell Res Ther. 2021;12(1):1–22.
16. Golchin A, Hosseinzadeh S, Ardeshirylajimi A. The exosomes released
from different cell types and their effects in wound healing. J Cell
Biochem. 2018;119(7):5043–52.
17. Marote A, Teixeira FG, Mendes-Pinheiro B, Salgado AJ. MSCs-derived
exosomes: cell-secreted nanovesicles with regenerative potential. Front
Pharmacol. 2016;7:231.
18. Jadli AS, Ballasy N, Edalat P, Patel VB. Inside (sight) of tiny communica-
tor: exosome biogenesis, secretion, and uptake. Mol Cell Biochem.
2020;467(1):77–94.
19. Nakamura Y, Kita S, Tanaka Y, Fukuda S, Obata Y, Okita T, Nishida H, Taka-
hashi Y, K awachi Y, Tsugawa-Shimizu Y. Adiponectin stimulates exosome
release to enhance mesenchymal stem-cell-driven therapy of heart
failure in mice. Mol Ther. 2020;28(10):2203–19.
20. Psaraki A, Ntari L, Karakostas C, Korrou-Karava D, Roubelakis MG.
Extracellular vesicles derived from mesenchymal stem/stromal cells: the
regenerative impact in liver diseases. Hepatology. 2021;14:261–8.
21. Simmons OL, Meinzer C, Rule J, Lee WM. Liver transplantation for aceta-
minophen-induced acute liver failure: role of psychiatric comorbidity in
listing decisions and outcomes. Dig Dis Sci. 2020;65(6):1861–8.
22. Ostapowicz G, Fontana RJ, Schiødt FV, Larson A, Davern TJ, Han SH,
McCashland TM, Shakil AO, Hay JE, Hynan L, et al. Results of a prospec-
tive study of acute liver failure at 17 tertiary care centers in the United
States. Ann Intern Med. 2002;137(12):947–54.
23. Dong V, Nanchal R, Karvellas CJ. Pathophysiology of acute liver failure.
Nutr Clin Pract. 2020;35(1):24–9.
24. Riordan SM, Williams R. Acute liver failure: targeted artificial and hepat-
ocyte-based support of liver regeneration and reversal of multiorgan
failure. J Hepatol. 2000;32:63–76.
25. Chapel A, Bertho JM, Bensidhoum M, Fouillard L, Young RG, Frick J,
Demarquay C, Cuvelier F, Mathieu E, Trompier F. Mesenchymal stem
cells home to injured tissues when co-infused with hematopoietic cells
to treat a radiation-induced multi-organ failure syndrome. J Gene Med.
2003;5(12):1028–38.
26. Bone RC. Sepsis, the sepsis syndrome, multi-organ failure: a plea for
comparable definitions. Philadelphia: American College of Physicians;
1991.
27. Das J, Ghosh J, Manna P, Sil PC. Acetaminophen induced acute liver
failure via oxidative stress and JNK activation: protective role of
taurine by the suppression of cytochrome P450 2E1. Free Radic Res.
2010;44(3):340–55.
28. Jiang W-P, Deng J-S, Huang S-S, Wu S-H, Chen C-C, Liao J-C, Chen H-Y,
Lin H-Y, Huang G-J. Sanghuangporus sanghuang mycelium prevents
paracetamol-induced hepatotoxicity through regulating the MAPK/
NF-κB, Keap1/Nrf2/HO-1, TLR4/PI3K/Akt, and CaMKKβ/LKB1/AMPK
pathways and suppressing oxidative stress and inflammation. Antioxi-
dants. 2021;10(6):897.
29. Shi S, Wang L, van der Laan LJ, Pan Q, Verstegen MM. Mitochon-
drial dysfunction and oxidative stress in liver transplantation and
underlying diseases: new insights and therapeutics. Transplantation.
2021;105(11):2362–73.
30. Wu X, Dong L, Lin X, Li J. Relevance of the NLRP3 inflammasome in the
pathogenesis of chronic liver disease. Front Immunol. 2017;8:1728.
31. Del Campo JA, Gallego P, Grande L. Role of inflammatory response in
liver diseases: therapeutic strategies. World J Hepatol. 2018;10(1):1.
32. He K, Zhu X, Liu Y, Miao C, Wang T, Li P, Zhao L, Chen Y, Gong J, Cai C.
Inhibition of NLRP3 inflammasome by thioredoxin-interacting protein
in mouse Kupffer cells as a regulatory mechanism for non-alcoholic
fatty liver disease development. Oncotarget. 2017;8(23):37657.
33. Zhan C, Lin G, Huang Y, Wang Z, Zeng F, Wu S. A dopamine-precursor-
based nanoprodrug for in-situ drug release and treatment of acute
liver failure by inhibiting NLRP3 inflammasome and facilitating liver
regeneration. Biomaterials. 2021;268:120573.
34. Woolbright BL, Jaeschke H. Role of the inflammasome in aceta-
minophen-induced liver injury and acute liver failure. J Hepatol.
2017;66(4):836–48.
35. Zhao Q, Wu C-S, Fang Y, Qian Y, Wang H, Fan Y-C, Wang K. Glucocorti-
coid regulates NLRP3 in acute-on-chronic hepatitis B liver failure. Int J
Med Sci. 2019;16(3):461.
36. Tao Y, Wang N, Qiu T, Sun X. The role of autophagy and NLRP3 inflam-
masome in liver fibrosis. BioMed Res Int. 2020;2020:1–8.
37. Li L, Shan S, Kang K, Zhang C, Kou R, Song F. The cross-talk of NLRP3
inflammasome activation and necroptotic hepatocyte death in
acetaminophen-induced mice acute liver injury. Hum Exp Toxicol.
2021;40(4):673–84.
38. Choi J, Kang S, Kim B, So S, Han J, Kim G-N, Lee M-Y, Roh S, Lee J-Y, Oh
SJ, et al. Efficient hepatic differentiation and regeneration potential
under xeno-free conditions using mass-producible amnion-derived
mesenchymal stem cells. Stem Cell Res Ther. 2021;12(1):569.
39. Zhang S, Yang Y, Fan L, Zhang F, Li L. The clinical application of mesen-
chymal stem cells in liver disease: the current situation and potential
future. Ann Transl Med. 2020;8(8):565.
40. Du XX, Shi Y, Yang Y, Yu Y, Lou HG, Lv FF, Chen Z, Yang Q. DAMP molecu-
lar IL-33 augments monocytic inflammatory storm in hepatitis B-precip-
itated acute-on-chronic liver failure. Liver Int. 2018;38(2):229–38.
41. Li M, Ling T, Teng F, Hu C, Su Z, Zhang C, Li X, Zhao T, Mu X, Li Y. CD5L
deficiency attenuate acetaminophen-induced liver damage in mice
via regulation of JNK and ERK signaling pathway. Cell Death Discov.
2021;7(1):1–11.
42. Weiss ARR, Dahlke MH. Immunomodulation by mesenchymal stem
cells (MSCs): mechanisms of action of living, apoptotic, and dead MSCs.
Front Immunol. 2019;10:1191.
43. Song N, Scholtemeijer M, Shah K. Mesenchymal stem cell immu-
nomodulation: mechanisms and therapeutic potential. Trends Pharma-
col Sci. 2020;41:653–64.
44. Carreras-Planella L, Monguió-Tortajada M, Borràs FE, Franquesa M.
Immunomodulatory effect of MSC on B cells is independent of
secreted extracellular vesicles. Front Immunol. 2019;10:1288.
45. Sun S-J, Lai W-H, Jiang Y, Zhen Z, Wei R, Lian Q, Liao S-Y, Tse H-F.
Immunomodulation by systemic administration of human-induced
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 17 of 21
Shokravietal. Stem Cell Research & Therapy (2022) 13:192
pluripotent stem cell-derived mesenchymal stromal cells to enhance
the therapeutic efficacy of cell-based therapy for treatment of myocar-
dial infarction. Theranostics. 2021;11(4):1641.
46. Bahroudi M, Bakhshi B, Soudi S, Najar-Peerayeh S. Immunomodulatory
effects of mesenchymal stem cell-conditioned media on lipopolysac-
charide of Vibrio cholerae as a vaccine candidate. Stem Cell Res Ther.
2021;12(1):1–14.
47. Putra A, Ridwan FB, Putridewi AI, Kustiyah AR, Wirastuti K, Sadyah NAC,
Rosdiana I, Munir D. The role of TNF-α induced MSCs on suppressive
inflammation by increasing TGF-β and IL-10. Open Access Maced J Med
Sci. 2018;6(10):1779.
48. Wang L, Deng Z, Sun Y, Zhao Y, Li Y, Yang M, Yuan R, Liu Y, Qian Z, Zhou
F. The study on the regulation of Th cells by MSCs through the JAK-STAT
signaling pathway to protect naturally aged sepsis model rats. Front
Immunol. 2021;13:820685. https:// doi. org/ 10. 3389/ fimmu. 2022. 820685.
49. Akiyama K, Chen C, Wang D, Xu X, Qu C, Yamaza T, Cai T, Chen W,
Sun L, Shi S. Mesenchymal-stem-cell-induced immunoregulation
involves FAS-ligand-/FAS-mediated T cell apoptosis. Cell Stem Cell.
2012;10(5):544–55.
50. Miloradovic D, Miloradovic D, Markovic BS, Acovic A, Harrell CR, Djonov
V, Arsenijevic N, Volarevic V. The effects of mesenchymal stem cells on
antimelanoma immunity depend on the timing of their administration.
Stem Cells Int. 2020;2020:1–13.
51. Volarevic V, Nurkovic J, Arsenijevic N, Stojkovic M. Concise review: thera-
peutic potential of mesenchymal stem cells for the treatment of acute
liver failure and cirrhosis. Stem Cells. 2014;32(11):2818–23.
52. Bataller R, Brenner DA. Liver fibrosis. J Clin Investig. 2005;115(2):209–18.
53. Hu C, Wu Z, Li L. Mesenchymal stromal cells promote liver regeneration
through regulation of immune cells. Int J Biol Sci. 2020;16(5):893.
54. Huang B, Cheng X, Wang H, Huang W, Wang D, Zhang K, Zhang H, Xue
Z, Da Y, Zhang N. Mesenchymal stem cells and their secreted molecules
predominantly ameliorate fulminant hepatic failure and chronic liver
fibrosis in mice respectively. J Transl Med. 2016;14(1):1–12.
55. Tsuchiya A, Takeuchi S, Watanabe T, Yoshida T, Nojiri S, Ogawa M, Terai S.
Mesenchymal stem cell therapies for liver cirrhosis: MSCs as “conduct-
ing cells” for improvement of liver fibrosis and regeneration. Inflamm
Regen. 2019;39(1):1–6.
56. Friedenstein A, Piatetzky-Shapiro I, Petrakova K. Osteogenesis in trans-
plants of bone marrow cells. Development. 1966;16(3):381–90.
57. Friedenstein AJ, Deriglasova UF, Kulagina NN, Panasuk AF, Rudakowa SF,
Luriá EA, Ruadkow IA. Precursors for fibroblasts in different populations
of hematopoietic cells as detected by the in vitro colony assay method.
Exp Hematol. 1974;2(2):83–92.
58. Ullah I, Seo K, Wi H, Kim Y, Lee S, Ock SA. Induction of the differentia-
tion of porcine bone marrow mesenchymal stem cells into premature
hepatocyte-like cells in an indirect coculture system with primary
hepatocytes. Anim Cells Syst. 2020;24(5):289–98.
59. Ji R, Zhang N, You N, Li Q, Liu W, Jiang N, Liu J, Zhang H, Wang D, Tao
K. The differentiation of MSCs into functional hepatocyte-like cells in a
liver biomatrix scaffold and their transplantation into liver-fibrotic mice.
Biomaterials. 2012;33(35):8995–9008.
60. Afshari A, Shamdani S, Uzan G, Naserian S, Azarpira N. Different
approaches for transformation of mesenchymal stem cells into
hepatocyte-like cells. Stem Cell Res Ther. 2020;11(1):1–14.
61. Afshari A, Shamdani S, Uzan G, Naserian S, Azarpira N. Different
approaches for transformation of mesenchymal stem cells into
hepatocyte-like cells. Stem Cell Res Ther. 2020;11(1):54.
62. Taléns-Visconti R, Bonora A, Jover R, Mirabet V, Carbonell F, Castell JV,
Gómez-Lechón MJ. Hepatogenic differentiation of human mesenchy-
mal stem cells from adipose tissue in comparison with bone marrow
mesenchymal stem cells. World J Gastroenterol: WJG. 2006;12(36):5834.
63. Ye JS, Su XS, Stoltz JF, de Isla N, Zhang L. Signalling pathways involved
in the process of mesenchymal stem cells differentiating into hepato-
cytes. Cell Prolif. 2015;48(2):157–65.
64. Popovici C, Roubin R, Coulier F, Birnbaum D. An evolutionary history of
the FGF superfamily. BioEssays. 2005;27(8):849–57.
65. Battula VL, Bareiss PM, Treml S, Conrad S, Albert I, Hojak S, Abele H,
Schewe B, Just L, Skutella T. Human placenta and bone marrow derived
MSC cultured in serum-free, b-FGF-containing medium express cell
surface frizzled-9 and SSEA-4 and give rise to multilineage differentia-
tion. Differentiation. 2007;75(4):279–91.
66. Eom YW, Oh J-E, Lee JI, Baik SK, Rhee K-J, Shin HC, Kim YM, Ahn
CM, Kong JH, Kim HS. The role of growth factors in maintenance of
stemness in bone marrow-derived mesenchymal stem cells. Biochem
Biophys Res Commun. 2014;445(1):16–22.
67. Wang Y, Yu X, Chen E, Li L. Liver-derived human mesenchymal stem
cells: a novel therapeutic source for liver diseases. Stem Cell Res Ther.
2016;7(1):1–8.
68. Neuss S, Becher E, Wöltje M, Tietze L, Jahnen-Dechent W. Functional
expression of HGF and HGF receptor/c-met in adult human mesen-
chymal stem cells suggests a role in cell mobilization, tissue repair, and
wound healing. Stem Cells. 2004;22(3):405–14.
69. Park B-W, Jung S-H, Das S, Lee SM, Park J-H, Kim H, Hwang J-W, Lee
S, Kim H-J, Kim H-Y. In vivo priming of human mesenchymal stem
cells with hepatocyte growth factor–engineered mesenchymal
stem cells promotes therapeutic potential for cardiac repair. Sci Adv.
2020;6(13):eaay6994.
70. Forte G, Minieri M, Cossa P, Antenucci D, Sala M, Gnocchi V, Fiaccavento
R, Carotenuto F, De Vito P, Baldini PM. Hepatocyte growth factor effects
on mesenchymal stem cells: proliferation, migration, and differentia-
tion. Stem Cells. 2006;24(1):23–33.
71. Chivu M, Dima SO, Stancu CI, Dobrea C, Uscatescu V, Necula LG, Bleotu
C, Tanase C, Albulescu R, Ardeleanu C. In vitro hepatic differentiation
of human bone marrow mesenchymal stem cells under differential
exposure to liver-specific factors. Transl Res. 2009;154(3):122–32.
72. Dai LJ, Li HY, Guan LX, Ritchie G, Zhou JX. The therapeutic potential of
bone marrow-derived mesenchymal stem cells on hepatic cirrhosis.
Stem Cell Res. 2009;2(1):16–25.
73. Yan Y, Xu W, Qian H, Si Y, Zhu W, Cao H, Zhou H, Mao F. Mesenchymal
stem cells from human umbilical cords ameliorate mouse hepatic
injury in vivo. Liver Int. 2009;29(3):356–65.
74. Zhang GZ, Sun HC, Zheng LB, Guo JB, Zhang XL. In vivo hepatic dif-
ferentiation potential of human umbilical cord-derived mesenchymal
stem cells: Therapeutic effect on liver fibrosis/cirrhosis. World J Gastro-
enterol. 2017;23(46):8152–68.
75. Tsiapalis D, O’Driscoll L. Mesenchymal stem cell derived extracellular
vesicles for tissue engineering and regenerative medicine applications.
Cells. 2020;9(4):991.
76. Chen L, Zhang C, Chen L, Wang X, Xiang B, Wu X, Guo Y, Mou X, Yuan
L, Chen B, et al. Human menstrual blood-derived stem cells ameliorate
liver fibrosis in mice by targeting hepatic stellate cells via paracrine
mediators. Stem Cells Transl Med. 2017;6(1):272–84.
77. Choi JS, Jeong IS, Han JH, Cheon SH, Kim S-W. IL-10-secreting
human MSCs generated by TALEN gene editing ameliorate liver
fibrosis through enhanced anti-fibrotic activity. Biomater Sci.
2019;7(3):1078–87.
78. Xuan J, Feng W, An Z, Yang J, Xu H, Li J, Zhao Z, Wen W. Anti-tgf [beta]-1
receptor inhibitor mediates the efficacy of the human umbilical cord
mesenchymal stem cells against liver fibrosis through tgf [beta]-1/smad
pathway. Mol Cell Biochem. 2017;429(1–2):113.
79. Jang YO, Cho M-Y, Yun C-O, Baik SK, Park K-S, Cha S-K, Chang SJ, Kim MY,
Lim YL, Kwon SO. Effect of function-enhanced mesenchymal stem cells
infected with decorin-expressing adenovirus on hepatic fibrosis. Stem
Cells Transl Med. 2016;5(9):1247–56.
80. Pietrosi G, Fernández-Iglesias A, Pampalone M, Ortega-Ribera M, Lozano
JJ, García-Calderó H, Abad-Jordà L, Conaldi PG, Parolini O, Vizzini G.
Human amniotic stem cells improve hepatic microvascular dysfunction
and portal hypertension in cirrhotic rats. Liver Int. 2020;40(10):2500–14.
81. Kim M-D, Kim S-S, Cha H-Y, Jang S-H, Chang D-Y, Kim W, Suh-Kim H,
Lee J-H. Therapeutic effect of hepatocyte growth factor-secreting
mesenchymal stem cells in a rat model of liver fibrosis. Exp Mol Med.
2014;46(8):e110.
82. Rockel JS, Rabani R, Viswanathan S, editors. Anti-fibrotic mechanisms of
exogenously-expanded mesenchymal stromal cells for fibrotic diseases.
Semin Cell Dev Biol. 2020;101:87–103.
83. Motawi TM, Atta HM, Sadik NA, Azzam M. The therapeutic effects of
bone marrow-derived mesenchymal stem cells and simvastatin in a rat
model of liver fibrosis. Cell Biochem Biophys. 2014;68(1):111–25.
84. Raafat N, Aal SMA, Abdo FK, El Ghonaimy NM. Mesenchymal stem
cells: in vivo therapeutic application ameliorates carbon tetrachloride
induced liver fibrosis in rats. Int J Biochem Cell Biol. 2015;68:109–18.
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 18 of 21
Shokravietal. Stem Cell Research & Therapy (2022) 13:192
85. Wang J, Bian C, Liao L, Zhu Y, Li J, Zeng L, Zhao RC. Inhibition of
hepatic stellate cells proliferation by mesenchymal stem cells and
the possible mechanisms. Hepatol Res. 2009;39(12):1219–28.
86. An SY, Jang YJ, Lim H-J, Han J, Lee J, Lee G, Park JY, Park S-Y, Kim JH,
Do B-R. Milk fat globule-EGF factor 8, secreted by mesenchymal
stem cells, protects against liver fibrosis in mice. Gastroenterology.
2017;152(5):1174–86.
87. Chen S, Lin N, Pan W, Hu K, Xu R. Activation of Notch1 signaling
by marrow-derived mesenchymal stem cells through cell–cell
contact inhibits proliferation of hepatic stellate cells. Life Sci.
2011;89(25–26):975–81.
88. Lee C, Kim M, Han J, Yoon M, Jung Y. Mesenchymal stem cells influ-
ence activation of hepatic stellate cells, and constitute a promising
therapy for liver fibrosis. Biomedicines. 2021;9(11):1598.
89. Duman DG, Zibandeh N, Ugurlu MU, Celikel C, Akkoc T, Banzragch
M, Genc D, Ozdogan O, Akkoc T. Mesenchymal stem cells suppress
hepatic fibrosis accompanied by expanded intrahepatic natural killer
cells in rat fibrosis model. Mol Biol Rep. 2019;46(3):2997–3008.
90. De Maria N, Colantonl A, Fagiuoli S, Liu G-J, Rogers BK, Farinati F, Van
Thiel DH, Floyd RA. Association between reactive oxygen species
and disease activity in chronic hepatitis C. Free Radic Biol Med.
1996;21(3):291–5.
91. Takemoto K, Hatano E, Iwaisako K, Takeiri M, Noma N, Ohmae S,
Toriguchi K, Tanabe K, Tanaka H, Seo S. Necrostatin-1 protects against
reactive oxygen species (ROS)-induced hepatotoxicity in acetami-
nophen-induced acute liver failure. FEBS Open Bio. 2014;4:777–87.
92. Choe G, Kim S-W, Park J, Park J, Kim S, Kim YS, Ahn Y, Jung D-W, Wil-
liams DR, Lee JY. Anti-oxidant activity reinforced reduced graphene
oxide/alginate microgels: mesenchymal stem cell encapsulation and
regeneration of infarcted hearts. Biomaterials. 2019;225:119513.
93. Dey R, Kemp K, Gray E, Rice C, Scolding N, Wilkins A. Human mes-
enchymal stem cells increase anti-oxidant defences in cells derived
from patients with Friedreich’s ataxia. Cerebellum. 2012;11(4):861–71.
94. Murakami T, Nagamura Y, Hirano K. The recovering effect of betaine
on carbon tetrachloride-induced liver injury. J Nutr Sci Vitaminol.
1998;44(2):249–55.
95. Mitsuhashi M, Morimura K, Wanibuchi H, Kiyota A, Wada S, Nakatani
T, Fukushima S. Examination of the rat model of liver injury via
thioacetamide (TAA) or carbon tetrachloride (CCl4). J Toxicol Pathol.
2004;17(3):219–22.
96. Eom YW, Shim KY, Baik SK. Mesenchymal stem cell therapy for liver
fibrosis. Korean J Intern Med. 2015;30(5):580.
97. Raafat N, Abdel Aal SM, Abdo FK, El Ghonaimy NM. Mesenchymal
stem cells: in vivo therapeutic application ameliorates carbon
tetrachloride induced liver fibrosis in rats. Int J Biochem Cell Biol.
2015;68:109–18.
98. Cho KA, Woo SY, Seoh JY, Han HS, Ryu KH. Mesenchymal stem cells
restore CCl4-induced liver injury by an antioxidative process. Cell Biol
Int. 2012;36(12):1267–74.
99. Quintanilha LF, Takami T, Hirose Y, Fujisawa K, Murata Y, Yamamoto N,
Goldenberg RC, Terai S, Sakaida I. Canine mesenchymal stem cells
show antioxidant properties against thioacetamide-induced liver injury
in vitro and in vivo. Hepatol Res. 2014;44(10):E206–17.
100. Phinney DG, Pittenger MF. Concise review: msc-derived exosomes for
cell-free therapy. Stem Cells. 2017;35(4):851–8.
101. Razeghian E, Margiana R, Chupradit S, Bokov DO, Abdelbasset WK,
Marofi F, Shariatzadeh S, Tosan F, Jarahian M. Mesenchymal stem/stro-
mal cells as a vehicle for cytokine delivery: an emerging approach for
tumor immunotherapy. Front Med. 2021;8:721174.
102. Rahbaran M, Baghini SS, Mardasi M, Razeghian E. MSCs modifies the
proliferation of leukemia MOLT-4 cells and induces their apoptosis
through up-regulating Bax, caspase-3, and-8, and down-regulating
Bcl-2 expression. Ann Cancer Res Ther. 2021;29(1):79–84.
103. Zhang S, Hou Y, Yang J, Xie D, Jiang L, Hu H, Hu J, Luo C, Zhang Q. Appli-
cation of mesenchymal stem cell exosomes and their drug-loading
systems in acute liver failure. J Cell Mol Med. 2020;24(13):7082–93.
104. Jiang L, Zhang S, Hu H, Yang J, Wang X, Ma Y, Jiang J, Wang J, Zhong L,
Chen M. Exosomes derived from human umbilical cord mesenchymal
stem cells alleviate acute liver failure by reducing the activity of the
NLRP3 inflammasome in macrophages. Biochem Biophys Res Com-
mun. 2019;508(3):735–41.
105. Yan Y, Jiang W, Tan Y, Zou S, Zhang H, Mao F, Gong A, Qian H, Xu W.
hucMSC exosome-derived GPX1 is required for the recovery of hepatic
oxidant injury. Mol Ther. 2017;25(2):465–79.
106. Shao M, Xu Q, Wu Z, Chen Y, Shu Y, Cao X, Chen M, Zhang B, Zhou Y, Yao
R. Exosomes derived from human umbilical cord mesenchymal stem
cells ameliorate IL-6-induced acute liver injury through miR-455-3p.
Stem Cell Res Ther. 2020;11(1):1–13.
107. Zhang S, Jiang L, Hu H, Wang H, Wang X, Jiang J, Ma Y, Yang J, Hou Y, Xie
D, et al. Pretreatment of exosomes derived from hUCMSCs with TNF-α
ameliorates acute liver failure by inhibiting the activation of NLRP3 in
macrophage. Life Sci. 2020;246:117401.
108. Hao NB, Li CZ, Lü MH, Tang B, Wang SM, Wu YY, Liang GP, Yang SM.
SDF-1/CXCR4 axis promotes MSCs to repair liver injury partially through
trans-differentiation and fusion with hepatocytes. Stem Cells Int.
2015;2015:960387.
109. Zhang S, Zhu Z, Wang Y, Liu S, Zhao C, Guan W, Zhao Y. Therapeutic
potential of Bama miniature pig adipose stem cells induced hepato-
cytes in a mouse model with acute liver failure. Cytotechnology.
2018;70(4):1131–41.
110. Wang J, Ren H, Yuan X, Ma H, Shi X, Ding Y. Interleukin-10 secreted by
mesenchymal stem cells attenuates acute liver failure through inhibit-
ing pyroptosis. Hepatol Res. 2018;48(3):E194-e202.
111. Wu X, Dong L, Lin X, Li J. Relevance of the NLRP3 inflammasome in the
pathogenesis of chronic liver disease. Front Immunol. 2017;8:1728.
112. Ma HC, Wang X, Wu MN, Zhao X, Yuan XW, Shi XL. Interleukin-10 con-
tributes to therapeutic effect of mesenchymal stem cells for acute liver
failure via signal transducer and activator of transcription 3 signaling
pathway. Chin Med J (Engl). 2016;129(8):967–75.
113. Liu Y, Ren H, Wang J, Yang F, Li J, Zhou Y, Yuan X, Zhu W, Shi X. Pros-
taglandin E(2) secreted by mesenchymal stem cells protects against
acute liver failure via enhancing hepatocyte proliferation. FASEB J.
2019;33(2):2514–25.
114. Zhou JH, Lu X, Yan CL, Sheng XY, Cao HC. Mesenchymal stromal cell-
dependent immunoregulation in chemically-induced acute liver failure.
World J Stem Cells. 2021;13(3):208–20.
115. Liu L, Song H, Duan H, Chai J, Yang J, Li X, Yu Y, Zhang X, Hu X, Xiao M,
et al. TSG-6 secreted by human umbilical cord-MSCs attenuates severe
burn-induced excessive inflammation via inhibiting activations of P38
and JNK signaling. Sci Rep. 2016;6(1):30121.
116. Wang Y, Wang JL, Ma HC, Tang ZT, Ding HR, Shi XL. Mesenchymal stem
cells increase heme oxygenase 1-activated autophagy in treatment of
acute liver failure. Biochem Biophys Res Commun. 2019;508(3):682–9.
117. Zhang ZH, Zhu W, Ren HZ, Zhao X, Wang S, Ma HC, Shi XL. Mesenchy-
mal stem cells increase expression of heme oxygenase-1 leading to
anti-inflammatory activity in treatment of acute liver failure. Stem Cell
Res Ther. 2017;8(1):70.
118. Zhao X, Shi XL, Zhang ZH, Ma HC, Yuan XW, Ding YT. Role of neutrophils
in treatment of rats with D-galactosamine-induced acute liver failure
with bone marrow mesenchymal stem cells. Zhonghua Gan Zang Bing
Za Zhi. 2016;24(8):601–7.
119. Guo G, Zhuang X, Xu Q, Wu Z, Zhu Y, Zhou Y, Li Y, Lu Y, Zhang B, Talbot P,
et al. Peripheral infusion of human umbilical cord mesenchymal stem
cells rescues acute liver failure lethality in monkeys. Stem Cell Res Ther.
2019;10(1):84.
120. Zhang Y, Li Y, Li W, Cai J, Yue M, Jiang L, Xu R, Zhang L, Li J, Zhu C.
Therapeutic effect of human umbilical cord mesenchymal stem
cells at various passages on acute liver failure in rats. Stem Cells Int.
2018;2018:7159465.
121. He Y, Guo X, Lan T, Xia J, Wang J, Li B, Peng C, Chen Y, Hu X, Meng Z.
Human umbilical cord-derived mesenchymal stem cells improve
the function of liver in rats with acute-on-chronic liver failure via
downregulating Notch and Stat1/Stat3 signaling. Stem Cell Res Ther.
2021;12(1):396.
122. Ding HR, Wang JL, Tang ZT, Wang Y, Zhou G, Liu Y, Ren HZ, Shi XL.
Mesenchymal stem cells improve glycometabolism and liver regenera-
tion in the treatment of post-hepatectomy liver failure. Front Physiol.
2019;10:412.
123. Sang JF, Shi XL, Han B, Huang T, Huang X, Ren HZ, Ding YT. Intraportal
mesenchymal stem cell transplantation prevents acute liver failure
through promoting cell proliferation and inhibiting apoptosis. Hepato-
biliary Pancreat Dis Int. 2016;15(6):602–11.
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 19 of 21
Shokravietal. Stem Cell Research & Therapy (2022) 13:192
124. Putra A, Rosdiana I, Darlan DM, Alif I, Hayuningtyas F, Wijaya I, Aryanti R,
Makarim FR, Antari AD. Intravenous administration is the best route of
mesenchymal stem cells migration in improving liver function enzyme
of acute liver failure. Folia Med (Plovdiv). 2020;62(1):52–8.
125. Zare H, Jamshidi S, Dehghan MM, Saheli M, Piryaei A. Bone marrow or
adipose tissue mesenchymal stem cells: comparison of the thera-
peutic potentials in mice model of acute liver failure. J Cell Biochem.
2018;119(7):5834–42.
126. Ali M, Payne SL. Biomaterial-based cell delivery strategies to promote
liver regeneration. Biomater Res. 2021;25(1):5.
127. Liu M, Yang J, Hu W, Zhang S, Wang Y. Superior performance of co-
cultured mesenchymal stem cells and hepatocytes in poly(lactic acid-
glycolic acid) scaffolds for the treatment of acute liver failure. Biomed
Mater. 2016;11(1):015008.
128. Kang HT, Jun DW, Jang K, Hoh JK, Lee JS, Saeed WK, Chae YJ, Lee JH.
Effect of stem cell treatment on acute liver failure model using scaffold.
Dig Dis Sci. 2019;64(3):781–91.
129. Ghaedi M, Soleimani M, Shabani I, Duan Y, Lotfi AS. Hepatic differentia-
tion from human mesenchymal stem cells on a novel nanofiber scaf-
fold. Cell Mol Biol Lett. 2012;17(1):89–106.
130. Xu L, Wang S, Sui X, Wang Y, Su Y, Huang L, Zhang Y, Chen Z, Chen Q,
Du H, et al. Mesenchymal stem cell-seeded regenerated silk fibroin
complex matrices for liver regeneration in an animal model of acute
liver failure. ACS Appl Mater Interfaces. 2017;9(17):14716–23.
131. Wang L, Li S, Wang HY, Zeng J, Zhang ZZ, Lv DY, Kuang WH. In a rat
model of acute liver failure, Icaritin improved the therapeutic effect
of mesenchymal stem cells by activation of the hepatocyte growth
factor/c-met pathway. Evid Based Complement Alternat Med.
2019;2019:4253846.
132. Chen H, Tang S, Liao J, Liu M, Lin Y. Therapeutic effect of human
umbilical cord blood mesenchymal stem cells combined with G-CSF
on rats with acute liver failure. Biochem Biophys Res Commun.
2019;517(4):670–6.
133. Chavez-Tapia NC, Mendiola-Pastrana I, Ornelas-Arroyo VJ, Noreña-
Herrera C, Vidaña-Perez D, Delgado-Sanchez G, Uribe M, Barrientos-
Gutierrez T. Granulocyte-colony stimulating factor for acute-on-chronic
liver failure: systematic review and meta-analysis. Ann Hepatol.
2015;14(5):631–41.
134. Nam HH, Jun DW, Jang K, Saeed WK, Lee JS, Kang HT, Chae YJ.
Granulocyte colony stimulating factor treatment in non-alcoholic
fatty liver disease: beyond marrow cell mobilization. Oncotarget.
2017;8(58):97965–76.
135. Sang JF, Shi XL, Han B, Huang X, Huang T, Ren HZ, Ding YT. Combined
mesenchymal stem cell transplantation and interleukin-1 recep-
tor antagonism after partial hepatectomy. World J Gastroenterol.
2016;22(16):4120–35.
136. Xiao JQ, Shi XL, Tan JJ, Zhang L, Xu Q, Ding YT. A novel treatment regi-
men for acute liver failure based on a combination of mesenchymal
stem cells transplantation and IL-lRa-loaded chitosan nanoparticles.
Zhonghua Gan Zang Bing Za Zhi. 2013;21(4):308–14.
137. Nasiri E, Alizadeh A, Roushandeh AM, Gazor R, Hashemi-Firouzi N,
Golipoor Z. Melatonin-pretreated adipose-derived mesenchymal stem
cells efficeintly improved learning, memory, and cognition in an animal
model of Alzheimer’s disease. Metab Brain Dis. 2019;34(4):1131–43.
138. Huang P, Wang L, Li Q, Tian X, Xu J, Xu J, Xiong Y, Chen G, Qian H, Jin C,
et al. Atorvastatin enhances the therapeutic efficacy of mesenchymal
stem cells-derived exosomes in acute myocardial infarction via up-reg-
ulating long non-coding RNA H19. Cardiovasc Res. 2020;116(2):353–67.
139. Nie H, An F, Mei J, Yang C, Zhan Q, Zhang Q. IL-1β Pretreatment
improves the efficacy of mesenchymal stem cells on acute liver failure
by enhancing CXCR4 expression. Stem Cells Int. 2020;2020:1498315.
140. Mohsin S, Shams S, Ali Nasir G, Khan M, Javaid Awan S, Khan SN, Riazud-
din S. Enhanced hepatic differentiation of mesenchymal stem cells after
pretreatment with injured liver tissue. Differentiation. 2011;81(1):42–8.
141. Li QY, Chen J, Luo YH, Zhang W, Xiao EH. Sodium Butyrate Pre-Treat-
ment Enhance Differentiation of Bone Marrow Mesenchymal Stem
Cells (BM-MSCs) into Hepatocytes and Improve Liver Injury. Curr Mol
Med. 2021;1.
142. Ma HC, Shi XL, Ren HZ, Yuan XW, Ding YT. Targeted migration of mesen-
chymal stem cells modified with CXCR4 to acute failing liver improves
liver regeneration. World J Gastroenterol. 2014;20(40):14884–94.
143. Chen H, Tang S, Liao J, Liu M, Lin Y. VEGF(165) gene-modified human
umbilical cord blood mesenchymal stem cells protect against acute
liver failure in rats. J Gene Med. 2021;23(10):e3369.
144. Wang W, Guo H, Li H, Yan Y, Wu C, Wang X, He X, Zhao N. Interleu-
kin-35 gene-modified mesenchymal stem cells protect concanavalin
A-induced fulminant hepatitis by decreasing the interferon gamma
level. Hum Gene Ther. 2018;29(2):234–41.
145. Zheng YB, Zhang XH, Huang ZL, Lin CS, Lai J, Gu YR, Lin BL, Xie DY, Xie
SB, Peng L, et al. Amniotic-fluid-derived mesenchymal stem cells over-
expressing interleukin-1 receptor antagonist improve fulminant hepatic
failure. PLoS ONE. 2012;7(7):e41392.
146. Zhang J, Gao J, Lin D, Xiong J, Wang J, Chen J, Lin B, Gao Z. Potential
networks regulated by MSCs in acute-on-chronic liver failure: exosomal
miRNAs and intracellular target genes. Front Genet. 2021;12:650536.
147. Zheng J, Lu T, Zhou C, Cai J, Zhang X, Liang J, Sui X, Chen X, Chen L,
Sun Y, et al. Extracellular vesicles derived from human umbilical cord
mesenchymal stem cells protect liver ischemia/reperfusion injury by
reducing CD154 expression on CD4+ T cells via CCT2. Adv Sci (Weinh).
2020;7(18):1903746.
148. Wei S, Wang Q, Zhou H, Qiu J, Li C, Shi C, Zhou S, Liu R, Lu L. miR-455-3p
alleviates hepatic stellate cell activation and liver fibrosis by suppress-
ing HSF1 expression. Mol Ther Nucleic Acids. 2019;16:758–69.
149. Jun JH, Kim JY, Choi JH, Lim JY, Kim K, Kim GJ. Exosomes from placenta-
derived mesenchymal stem cells are involved in liver regenera-
tion in hepatic failure induced by bile duct ligation. Stem Cells Int.
2020;2020:5485738.
150. Damania A, Jaiman D, Teotia AK, Kumar A. Mesenchymal stromal cell-
derived exosome-rich fractionated secretome confers a hepatoprotec-
tive effect in liver injury. Stem Cell Res Ther. 2018;9(1):31.
151. Chen L, Xiang B, Wang X, Xiang C. Exosomes derived from human
menstrual blood-derived stem cells alleviate fulminant hepatic failure.
Stem Cell Res Ther. 2017;8(1):9.
152. Dang S, Zhou J, Wang Z, Wang K, Dai S, He S. MiR-299-3p functions as
a tumor suppressor via targeting Sirtuin 5 in hepatocellular carcinoma.
Biomed Pharmacother. 2018;106:966–75.
153. Temnov A, Rogov K, Zhalimov V, Igor P, Pekov S, Bader A, Sklifas A,
Giri S. The effect of a mesenchymal stem cell conditioned medium
fraction on morphological characteristics of hepatocytes in acetami-
nophen-induced acute liver failure: a preliminary study. Hepat Med.
2019;11:89–96.
154. Chen L, Zhang J, Yang L, Zhang G, Wang Y, Zhang S. The effects of
conditioned medium derived from mesenchymal stem cells cocultured
with hepatocytes on damaged hepatocytes and acute liver failure in
rats. Stem Cells Int. 2018;2018:9156560.
155. Foroutan T, Kabiri F, Motamedi E. Silica magnetic graphene oxide
improves the effects of stem cell-conditioned medium on acute liver
failure. ACS Omega. 2021;6(33):21194–206.
156. Lotfinia M, Kadivar M, Piryaei A, Pournasr B, Sardari S, Sodeifi N,
Sayahpour FA, Baharvand H. Effect of secreted molecules of human
embryonic stem cell-derived mesenchymal stem cells on acute hepatic
failure model. Stem Cells Dev. 2016;25(24):1898–908.
157. Huang B, Cheng X, Wang H, Huang W, la Ga HuZ, Wang D, Zhang K,
Zhang H, Xue Z, Da Y, et al. Mesenchymal stem cells and their secreted
molecules predominantly ameliorate fulminant hepatic failure and
chronic liver fibrosis in mice respectively. J Transl Med. 2016;14:45.
158. Zagoura DS, Roubelakis MG, Bitsika V, Trohatou O, Pappa KI, Kapelou-
zou A, Antsaklis A, Anagnou NP. Therapeutic potential of a distinct
population of human amniotic fluid mesenchymal stem cells and
their secreted molecules in mice with acute hepatic failure. Gut.
2012;61(6):894–906.
159. Cho PS, Messina DJ, Hirsh EL, Chi N, Goldman SN, Lo DP, Harris IR,
Popma SH, Sachs DH, Huang CA. Immunogenicity of umbilical cord
tissue–derived cells. Blood. 2008;111(1):430–8.
160. Deuse T, Stubbendorff M, Tang-Quan K, Phillips N, Kay MA, Eiermann T,
Phan TT, Volk H-D, Reichenspurner H, Robbins RC. Immunogenicity and
immunomodulatory properties of umbilical cord lining mesenchymal
stem cells. Cell Transplant. 2011;20(5):655–67.
161. Strioga M, Viswanathan S, Darinskas A, Slaby O, Michalek J. Same or
not the same? Comparison of adipose tissue-derived versus bone
marrow-derived mesenchymal stem and stromal cells. Stem Cells Dev.
2012;21(14):2724–52.
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 20 of 21
Shokravietal. Stem Cell Research & Therapy (2022) 13:192
162. Shi M, Zhang Z, Xu R, Lin H, Fu J, Zou Z, Zhang A, Shi J, Chen L, Lv S,
et al. Human mesenchymal stem cell transfusion is safe and improves
liver function in acute-on-chronic liver failure patients. Stem Cells Transl
Med. 2012;1(10):725–31.
163. Amer ME, El-Sayed SZ, El-Kheir WA, Gabr H, Gomaa AA, El-Noomani N,
Hegazy M. Clinical and laboratory evaluation of patients with end-stage
liver cell failure injected with bone marrow-derived hepatocyte-like
cells. Eur J Gastroenterol Hepatol. 2011;23(10):936–41.
164. Lin BL, Chen JF, Qiu WH, Wang KW, Xie DY, Chen XY, Liu QL, Peng L,
Li JG, Mei YY, et al. Allogeneic bone marrow-derived mesenchymal
stromal cells for hepatitis B virus-related acute-on-chronic liver failure: a
randomized controlled trial. Hepatology. 2017;66(1):209–19.
165. Peng L, Xie DY, Lin BL, Liu J, Zhu HP, Xie C, Zheng YB, Gao ZL. Autolo-
gous bone marrow mesenchymal stem cell transplantation in liver fail-
ure patients caused by hepatitis B: short-term and long-term outcomes.
Hepatology. 2011;54(3):820–8.
166. Tsochatzis EA, Bosch J, Burroughs AK. Liver cirrhosis. Lancet.
2014;383(9930):1749–61.
167. Mohamadnejad M, Alimoghaddam K, Mohyeddin-Bonab M, Bagheri
M, Bashtar M, Ghanaati H, Baharvand H, Ghavamzadeh A, Malekzadeh
R. Phase 1 trial of autologous bone marrow mesenchymal stem cell
transplantation in patients with decompensated liver cirrhosis. Arch
Iran Med. 2007;10(4):459–66.
168. Wang L, Li J, Liu H, Li Y, Fu J, Sun Y, Xu R, Lin H, Wang S, Lv S, et al. Pilot
study of umbilical cord-derived mesenchymal stem cell transfusion
in patients with primary biliary cirrhosis. J Gastroenterol Hepatol.
2013;28(Suppl 1):85–92.
169. El-Ansary M, Abdel-Aziz I, Mogawer S, Abdel-Hamid S, Hammam O,
Teaema S, Wahdan M. Phase II trial: undifferentiated versus dif-
ferentiated autologous mesenchymal stem cells transplantation in
Egyptian patients with HCV induced liver cirrhosis. Stem Cell Rev Rep.
2012;8(3):972–81.
170. Amin MA, Sabry D, Rashed LA, Aref WM, el-Ghobary MA, Farhan MS,
Fouad HA, Youssef YA. Short-term evaluation of autologous transplanta-
tion of bone marrow-derived mesenchymal stem cells in patients with
cirrhosis: Egyptian study. Clin Transplant. 2013;27(4):607–12.
171. Xu L, Gong Y, Wang B, Shi K, Hou Y, Wang L, Lin Z, Han Y, Lu L, Chen D,
et al. Randomized trial of autologous bone marrow mesenchymal stem
cells transplantation for hepatitis B virus cirrhosis: regulation of Treg/
Th17 cells. J Gastroenterol Hepatol. 2014;29(8):1620–8.
172. Kharaziha P, Hellström PM, Noorinayer B, Farzaneh F, Aghajani K, Jafari
F, Telkabadi M, Atashi A, Honardoost M, Zali MR, et al. Improvement of
liver function in liver cirrhosis patients after autologous mesenchymal
stem cell injection: a phase I-II clinical trial. Eur J Gastroenterol Hepatol.
2009;21(10):1199–205.
173. Liang J, Zhang H, Zhao C, Wang D, Ma X, Zhao S, Wang S, Niu L, Sun
L. Effects of allogeneic mesenchymal stem cell transplantation in
the treatment of liver cirrhosis caused by autoimmune diseases. Int J
Rheum Dis. 2017;20(9):1219–26.
174. Mohamadnejad M, Alimoghaddam K, Bagheri M, Ashrafi M, Abdol-
lahzadeh L, Akhlaghpoor S, Bashtar M, Ghavamzadeh A, Malekzadeh R.
Randomized placebo-controlled trial of mesenchymal stem cell trans-
plantation in decompensated cirrhosis. Liver Int. 2013;33(10):1490–6.
175. Ball SG, Shuttleworth CA, Kielty CM. Mesenchymal stem cells and neo-
vascularization: role of platelet-derived growth factor receptors. J Cell
Mol Med. 2007;11(5):1012–30.
176. Kim DH, Yoo KH, Choi KS, Choi J, Choi S-Y, Yang S-E, Yang Y-S, Im HJ, Kim
KH, Jung HL. Gene expression profile of cytokine and growth factor
during differentiation of bone marrow-derived mesenchymal stem cell.
Cytokine. 2005;31(2):119–26.
177. Zhou YF, Bosch-Marce M, Okuyama H, Krishnamachary B, Kimura
H, Zhang L, Huso DL, Semenza GL. Spontaneous transformation of
cultured mouse bone marrow-derived stromal cells. Cancer Res.
2006;66(22):10849–54.
178. Røsland GV, Svendsen A, Torsvik A, Sobala E, McCormack E, Immervoll
H, Mysliwietz J, Tonn JC, Goldbrunner R, Lønning PE, et al. Long-term
cultures of bone marrow-derived human mesenchymal stem cells
frequently undergo spontaneous malignant transformation. Cancer
Res. 2009;69(13):5331–9.
179. Dahl J-A, Duggal S, Coulston N, Millar D, Melki J, Shahdadfar A, Brinch-
mann JE, Collas P. Genetic and epigenetic instability of human bone
marrow mesenchymal stem cells expanded in autologous serum or
fetal bovine serum. Int J Dev Biol. 2002;52(8):1033–42.
180. Zhang Z, Lin H, Shi M, Xu R, Fu J, Lv J, Chen L, Lv S, Li Y, Yu S, et al.
Human umbilical cord mesenchymal stem cells improve liver function
and ascites in decompensated liver cirrhosis patients. J Gastroenterol
Hepatol. 2012;27(Suppl 2):112–20.
181. Kang SH, Kim MY, Eom YW, Baik SK. Mesenchymal stem cells for
the treatment of liver disease: present and perspectives. Gut Liver.
2020;14(3):306–15.
182. Kim JY, Rhim W-K, Yoo Y-I, Kim D-S, Ko K-W, Heo Y, Park CG, Han DK.
Defined MSC exosome with high yield and purity to improve regenera-
tive activity. J Tissue Eng. 2021;12:20417314211008624.
183. Wang L, Abhange KK, Wen Y, Chen Y, Xue F, Wang G, Tong J, Zhu C, He
X, Wan Y. Preparation of engineered extracellular vesicles derived from
human umbilical cord mesenchymal stem cells with ultrasonication for
skin rejuvenation. ACS Omega. 2019;4(27):22638–45.
184. Cao J, Wang B, Tang T, Lv L, Ding Z, Li Z, Hu R, Wei Q, Shen A, Fu Y. Three-
dimensional culture of MSCs produces exosomes with improved yield
and enhanced therapeutic efficacy for cisplatin-induced acute kidney
injury. Stem Cell Res Ther. 2020;11(1):1–13.
185. Yan L, Wu X. Exosomes produced from 3D cultures of umbilical cord
mesenchymal stem cells in a hollow-fiber bioreactor show improved
osteochondral regeneration activity. Cell Biol Toxicol. 2020;36(2):165–78.
186. Haraszti RA, Miller R, Stoppato M, Sere YY, Coles A, Didiot M-C, Wollacott
R, Sapp E, Dubuke ML, Li X. Exosomes produced from 3D cultures of
MSCs by tangential flow filtration show higher yield and improved
activity. Mol Ther. 2018;26(12):2838–47.
187. Qazi TH, Mooney DJ, Duda GN, Geissler S. Biomaterials that promote
cell-cell interactions enhance the paracrine function of MSCs. Biomate-
rials. 2017;140:103–14.
188. Phan J, Kumar P, Hao D, Gao K, Farmer D, Wang A. Engineering mes-
enchymal stem cells to improve their exosome efficacy and yield for
cell-free therapy. J Extracell Vesicles. 2018;7(1):1522236.
189. Lee B-C, Kang K-S. Functional enhancement strategies for immu-
nomodulation of mesenchymal stem cells and their therapeutic
application. Stem Cell Res Ther. 2020;11(1):1–10.
190. Cunningham CJ, Redondo-Castro E, Allan SM. The therapeutic potential
of the mesenchymal stem cell secretome in ischaemic stroke. J Cereb
Blood Flow Metab. 2018;38(8):1276–92.
191. Nakao Y, Fukuda T, Zhang Q, Sanui T, Shinjo T, Kou X, Chen C, Liu D,
Watanabe Y, Hayashi C. Exosomes from TNF-α-treated human gingiva-
derived MSCs enhance M2 macrophage polarization and inhibit peri-
odontal bone loss. Acta Biomater. 2021;122:306–24.
192. Amer M-EM, El-Sayed SZ, Abou El-Kheir W, Gabr H, Gomaa AA, El-
Noomani N, Hegazy M. Clinical and laboratory evaluation of patients
with end-stage liver cell failure injected with bone marrow-derived
hepatocyte-like cells. Eur J Gastroenterol Hepatol. 2011;23(10):936–41.
193. Saleh M, Taher M, Sohrabpour AA, Vaezi AA, Nasiri Toosi M, Kavianpour
M, Ghazvinian Z, Abdolahi S, Verdi J. Perspective of placenta derived
mesenchymal stem cells in acute liver failure. Cell Biosci. 2020;10:71.
194. Tautenhahn HM, Brückner S, Baumann S, Winkler S, Otto W, von Bergen
M, Bartels M, Christ B. Attenuation of postoperative acute liver failure by
mesenchymal stem cell treatment due to metabolic implications. Ann
Surg. 2016;263(3):546–56.
195. Gilsanz C, Aller MA, Fuentes-Julian S, Prieto I, Blázquez-Martinez A,
Argudo S, Fernández-Delgado J, Beleña J, Arias J, De Miguel MP.
Adipose-derived mesenchymal stem cells slow disease progression of
acute-on-chronic liver failure. Biomed Pharmacother. 2017;91:776–87.
196. Lin NC, Wu HH, Ho JH, Liu CS, Lee OK. Mesenchymal stem cells prolong
survival and prevent lethal complications in a porcine model of fulmi-
nant liver failure. Xenotransplantation. 2019;26(6):e12542.
197. Wang J-L, Ding H-R, Pan C-Y, Shi X-L, Ren H-Z. Mesenchymal stem cells
ameliorate lipid metabolism through reducing mitochondrial damage
of hepatocytes in the treatment of post-hepatectomy liver failure. Cell
Death Dis. 2021;12(1):111.
198. Amiri F, Molaei S, Bahadori M, Nasiri F, Deyhim MR, Jalili MA, Nourani
MR, Habibi RM. Autophagy-modulated human bone marrow-derived
mesenchymal stem cells accelerate liver restoration in mouse models
of acute liver failure. Iran Biomed J. 2016;20(3):135–44.
199. Hao N-B, Li C-Z, Lü M-H, Tang B, Wang S-M, Wu Y-Y, Liang G-P, Yang S-M.
SDF-1/CXCR4 axis promotes MSCs to repair liver injury partially through
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 21 of 21
Shokravietal. Stem Cell Research & Therapy (2022) 13:192
fast, convenient online submission
thorough peer review by experienced researchers in your field
rapid publication on acceptance
support for research data, including large and complex data types
gold Open Access which fosters wider collaboration and increased citations
maximum visibility for your research: over 100M website views per year
At BMC, research is always in progress.
Learn more biomedcentral.com/submissions
Ready to submit your research
Ready to submit your research
? Choose BMC and benefit from:
? Choose BMC and benefit from:
trans-differentiation and fusion with hepatocytes. Stem Cells Int.
2015;2015:960387.
200. Ramanathan R, Rupert S, Selvaraj S, Satyanesan J, Vennila R, Rajagopal
S. Role of human Wharton’s Jelly derived mesenchymal stem cells (WJ-
MSCs) for rescue of d-galactosamine induced acute liver injury in mice.
J Clin Exp Hepatol. 2017;7(3):205–14.
201. Huang YJ, Chen P, Lee CY, Yang SY, Lin MT, Lee HS, Wu YM. Protec-
tion against acetaminophen-induced acute liver failure by omentum
adipose tissue derived stem cells through the mediation of Nrf2 and
cytochrome P450 expression. J Biomed Sci. 2016;23:5.
202. Zhang S, Chen L, Liu T, Zhang B, Xiang D, Wang Z, Wang Y. Human
umbilical cord matrix stem cells efficiently rescue acute liver failure
through paracrine effects rather than hepatic differentiation. Tissue Eng
Part A. 2012;18(13–14):1352–64.
203. Liao H, Du S, Jiang T, Zheng M, Xiang Z, Yang J. UMSCs attenuate
LPS/D-GalN-induced acute liver failure in mice by down-regulating the
MyD88/NF-κB pathway. J Clin Transl Hepatol. 2021;9(5):690–701.
204. Milosavljevic N, Gazdic M, Simovic Markovic B, Arsenijevic A, Nurkovic
J, Dolicanin Z, Djonov V, Lukic ML, Volarevic V. Mesenchymal stem
cells attenuate acute liver injury by altering ratio between interleu-
kin 17 producing and regulatory natural killer T cells. Liver Transpl.
2017;23(8):1040–50.
205. Fathy M, Okabe M, Saad Eldien HM, Yoshida T. AT-MSCs antifibrotic
activity is improved by eugenol through modulation of TGF-β/Smad
signaling pathway in rats. Molecules. 2020;25(2):348.
206. Zhao X, Shi X, Zhang Z, Ma H, Yuan X, Ding Y. Combined treatment with
MSC transplantation and neutrophil depletion ameliorates D-GalN/
LPS-induced acute liver failure in rats. Clin Res Hepatol Gastroenterol.
2016;40(6):730–8.
207. Kong D, Xu H, Chen M, Yu Y, Qian Y, Qin T, Tong Y, Xia Q, Hang H. Co-
encapsulation of HNF4α overexpressing UMSCs and human primary
hepatocytes ameliorates mouse acute liver failure. Stem Cell Res Ther.
2020;11(1):449.
208. Feng J, Yao W, Zhang Y, Xiang AP, Yuan D, Hei Z. Intravenous anesthet-
ics enhance the ability of human bone marrow-derived mesenchymal
stem cells to alleviate hepatic ischemia-reperfusion injury in a receptor-
dependent manner. Cell Physiol Biochem. 2018;47(2):556–66.
209. Despeyroux A, Duret C, Gondeau C, Perez-Gracia E, Chuttoo L, de
Boussac H, Briolotti P, Bony C, Noël D, Jorgensen C, et al. Mesenchymal
stem cells seeded on a human amniotic membrane improve liver
regeneration and mouse survival after extended hepatectomy. J Tissue
Eng Regen Med. 2018;12(4):1062–73.
210. Parekkadan B, van Poll D, Suganuma K, Carter EA, Berthiaume F, Tilles
AW, Yarmush ML. Mesenchymal stem cell-derived molecules reverse
fulminant hepatic failure. PLoS ONE. 2007;2(9):e941.
211. Kantarcıoğlu M, Demirci H, Avcu F, Karslıoğlu Y, Babayiğit MA, Karaman
B, Öztürk K, Gürel H, Akdoğan Kayhan M, Kaçar S, et al. Efficacy of
autologous mesenchymal stem cell transplantation in patients with
liver cirrhosis. Turk J Gastroenterol. 2015;26(3):244–50.
212. Jang YO, Kim YJ, Baik SK, Kim MY, Eom Y W, Cho MY, Park HJ, Park SY,
Kim BR, Kim JW, et al. Histological improvement following administra-
tion of autologous bone marrow-derived mesenchymal stem cells for
alcoholic cirrhosis: a pilot study. Liver Int. 2014;34(1):33–41.
213. Salama H, Zekri AR, Medhat E, Al Alim SA, Ahmed OS, Bahnassy AA,
Lotfy MM, Ahmed R, Musa S. Peripheral vein infusion of autologous
mesenchymal stem cells in Egyptian HCV-positive patients with end-
stage liver disease. Stem Cell Res Ther. 2014;5(3):70.
214. Fang X, Liu L, Dong J, Zhang J, Song H, Song Y, Huang Y, Cui X, Lin J,
Chen C, et al. A study about immunomodulatory effect and efficacy
and prognosis of human umbilical cord mesenchymal stem cells in
patients with chronic hepatitis B-induced decompensated liver cirrho-
sis. J Gastroenterol Hepatol. 2018;33(4):774–80.
215. Zhang YC, Liu W, Fu BS, Wang GY, Li HB, Yi HM, Jiang N, Wang G, Zhang
J, Yi SH, et al. Therapeutic potentials of umbilical cord-derived mesen-
chymal stromal cells for ischemic-type biliary lesions following liver
transplantation. Cytotherapy. 2017;19(2):194–9.
216. Suk KT, Yoon JH, Kim MY, Kim CW, Kim JK, Park H, Hwang SG, Kim DJ,
Lee BS, Lee SH, et al. Transplantation with autologous bone marrow-
derived mesenchymal stem cells for alcoholic cirrhosis: phase 2 trial.
Hepatology. 2016;64(6):2185–97.
217. 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(12):2053–61.
218. Detry O, Vandermeulen M, Delbouille MH, Somja J, Bletard N, Briquet
A, Lechanteur C, Giet O, Baudoux E, Hannon M, et al. Infusion of mes-
enchymal stromal cells after deceased liver transplantation: a phase I-II,
open-label, clinical study. J Hepatol. 2017;67(1):47–55.
Publisher’s Note
Springer Nature remains neutral with regard to jurisdictional claims in pub-
lished maps and institutional affiliations.
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
1.
2.
3.
4.
5.
6.
Terms and Conditions
Springer Nature journal content, brought to you courtesy of Springer Nature Customer Service Center GmbH (“Springer Nature”).
Springer Nature supports a reasonable amount of sharing of research papers by authors, subscribers and authorised users (“Users”), for small-
scale personal, non-commercial use provided that all copyright, trade and service marks and other proprietary notices are maintained. By
accessing, sharing, receiving or otherwise using the Springer Nature journal content you agree to these terms of use (“Terms”). For these
purposes, Springer Nature considers academic use (by researchers and students) to be non-commercial.
These Terms are supplementary and will apply in addition to any applicable website terms and conditions, a relevant site licence or a personal
subscription. These Terms will prevail over any conflict or ambiguity with regards to the relevant terms, a site licence or a personal subscription
(to the extent of the conflict or ambiguity only). For Creative Commons-licensed articles, the terms of the Creative Commons license used will
apply.
We collect and use personal data to provide access to the Springer Nature journal content. We may also use these personal data internally within
ResearchGate and Springer Nature and as agreed share it, in an anonymised way, for purposes of tracking, analysis and reporting. We will not
otherwise disclose your personal data outside the ResearchGate or the Springer Nature group of companies unless we have your permission as
detailed in the Privacy Policy.
While Users may use the Springer Nature journal content for small scale, personal non-commercial use, it is important to note that Users may
not:
use such content for the purpose of providing other users with access on a regular or large scale basis or as a means to circumvent access
control;
use such content where to do so would be considered a criminal or statutory offence in any jurisdiction, or gives rise to civil liability, or is
otherwise unlawful;
falsely or misleadingly imply or suggest endorsement, approval , sponsorship, or association unless explicitly agreed to by Springer Nature in
writing;
use bots or other automated methods to access the content or redirect messages
override any security feature or exclusionary protocol; or
share the content in order to create substitute for Springer Nature products or services or a systematic database of Springer Nature journal
content.
In line with the restriction against commercial use, Springer Nature does not permit the creation of a product or service that creates revenue,
royalties, rent or income from our content or its inclusion as part of a paid for service or for other commercial gain. Springer Nature journal
content cannot be used for inter-library loans and librarians may not upload Springer Nature journal content on a large scale into their, or any
other, institutional repository.
These terms of use are reviewed regularly and may be amended at any time. Springer Nature is not obligated to publish any information or
content on this website and may remove it or features or functionality at our sole discretion, at any time with or without notice. Springer Nature
may revoke this licence to you at any time and remove access to any copies of the Springer Nature journal content which have been saved.
To the fullest extent permitted by law, Springer Nature makes no warranties, representations or guarantees to Users, either express or implied
with respect to the Springer nature journal content and all parties disclaim and waive any implied warranties or warranties imposed by law,
including merchantability or fitness for any particular purpose.
Please note that these rights do not automatically extend to content, data or other material published by Springer Nature that may be licensed
from third parties.
If you would like to use or distribute our Springer Nature journal content to a wider audience or on a regular basis or in any other manner not
expressly permitted by these Terms, please contact Springer Nature at
onlineservice@springernature.com
... Their pleiotropic effects extend to various therapeutic effects in liver diseases, including immunomodulation, anti-fibrotic, and anti-apoptotic activities [33]. While clinical studies support the safety and efficacy of MSCs in treating liver disorders, challenges such as uncontrolled differentiation/proliferation and potential immunogenicity persist [34]. EVs derived from MSCs offer a viable alternative [34]. ...
... While clinical studies support the safety and efficacy of MSCs in treating liver disorders, challenges such as uncontrolled differentiation/proliferation and potential immunogenicity persist [34]. EVs derived from MSCs offer a viable alternative [34]. These nanoscale vesicles, varying in size, enclose a wide array of bioactive molecules, such as micro RNAs, messenger RNAs, and lipids [35]. ...
Article
Full-text available
Inflammation plays a critical role in conditions such as acute liver failure, acute-on-chronic liver failure, and ischemia–reperfusion-induced liver injury. Various pathogenic pathways contribute to liver inflammation, involving inflammatory polarization of macrophages and Küpffer cells, neutrophil infiltration, dysregulation of T cell subsets, oxidative stress, and activation of hepatic stellate cells. While mesenchymal stromal cells (MSCs) have demonstrated beneficial properties, their clinical translation is limited by their cellular nature. However, MSC-derived extracellular vesicles (MSC-EVs) have emerged as a promising cell-free therapeutic approach for immunomodulation. MSC-EVs naturally mirror their parental cell properties, overcoming the limitations associated with the use of MSCs. In vitro and in vivo preclinical studies have demonstrated that MSC-EVs replicate the beneficial effects of MSCs in liver injury. This includes the reduction of cell death and oxidative stress, improvement of hepatocyte function, induction of immunomodulatory effects, and mitigation of cytokine storm. Nevertheless, MSC-EVs face challenges regarding the necessity of defining consistent isolation methods, optimizing MSCs culture conditions, and establishing quality control measures for EV characterization and functional assessment. By establishing standardized protocols, guidelines, and affordable cost mass production, clinicians and researchers will have a solid foundation to conduct further studies, validate the therapeutic efficacy of MSC-EVs, and ultimately pave the way for their clinical implementation in acute liver injury.
... Compared to animal/human cells, PDVLNs have higher production rates and shorter extraction periods, thus making PDVLNs a promising biological material with significant application potential. In contrast, mammalian exosomes often have lower yields and limited sources such as milk, urine, blood, and bile [83,84]. Therefore, the emergence of PDVLNs provides new possibilities to overcome these limitations. ...
Article
Full-text available
A wide variety of natural bioactive compounds derived from plants have demonstrated significant clinical relevance in the treatment of various diseases such as cancer, chronic disease, and inflammation. An increasing number of studies have surfaced that give credence to the potential of plant-derived vesicle-like nanoparticles (PDVLNs) as compelling candidates for a drug delivery system (DDS). PDVLNs are cost-effective production, non-toxicity and non-immunogenicity and fascinating bi-ocompatibility. In this review, we attempt to comprehensively review and consolidate the position of PDVLNs as next-generation drug delivery nanoplatforms. We aim to give a quick glance to readers of the current developments of PDVLNs, including their biogenesis, characteristic features, composition, administration routes, advantages, and application. Further, we discuss the advantages and limitations of PDVLNs. We expect that the role of PDVLNs in drug delivery will be significantly enhanced, thus positioning them as the next generation of therapeutic modalities in the foreseeable future.
... Novel stem cell technologies are currently being developed to improve the efficacy of stem cells for liver regeneration. Advancements in this area are mostly focused on MSCs and include techniques such as priming, genetic manipulation, and extracellular vesicle (EV)-based therapy [92,93]. Priming is a pre-conditioning process that involves the exposure of cells to cytokines, growth factors, and selected microenvironments to enhance their survival and augment their valuable properties. ...
Article
Full-text available
Portal vein embolization with stem cell augmentation (PVESA) is an emerging approach for enhancing the growth of the liver segment that will remain after surgery (i.e., future liver remnant, FLR) in patients with liver cancer. Conventional portal vein embolization (PVE) aims to induce preoperative FLR growth, but it has a risk of failure in patients with underlying liver dysfunction and comorbid illnesses. PVESA combines PVE with stem cell therapy to potentially improve FLR size and function more effectively and efficiently. Various types of stem cells can help improve liver growth by secreting paracrine signals for hepatocyte growth or by transforming into hepatocytes. Mesenchymal stem cells (MSCs), unrestricted somatic stem cells, and small hepatocyte-like progenitor cells have been used to augment liver growth in preclinical animal models, while clinical studies have demonstrated the benefit of CD133 + bone marrow–derived MSCs and hematopoietic stem cells. These investigations have shown that PVESA is generally safe and enhances liver growth after PVE. However, optimizing the selection, collection, and application of stem cells remains crucial to maximize benefits and minimize risks. Additionally, advanced stem cell technologies, such as priming, genetic modification, and extracellular vesicle-based therapy, that could further enhance efficacy outcomes should be evaluated. Despite its potential, PVESA requires more investigations, particularly mechanistic studies that involve orthotopic animal models of liver cancer with concomitant liver injury as well as larger human trials.
Article
Introduction: The emergence of antiparasitic drug resistance poses a concerning threat to animals and humans. Mesenchymal Stem Cells (MSCs) have been widely used to treat infections in humans, pets, and livestock. Although this is an emerging field of study, the current review outlines possible mechanisms and examines potential synergism in combination therapies and the possible harmful effects of such an approach. Areas covered: The present study delved into the latest pre-clinical research on utilizing MSCs to treat parasitic infections. As per investigations, the introduction of MSCs to patients grappling with parasitic diseases like schistosomiasis, malaria, cystic echinococcosis, toxoplasmosis, leishmaniasis, and trypanosomiasis has shown a reduction in parasite prevalence. This intervention also alters the levels of both pro- and anti-inflammatory cytokines. Furthermore, the combined administration of MSCs and antiparasitic drugs has demonstrated enhanced efficacy in combating parasites and modulating the immune response. Expert opinion: Mesenchymal stem cells are a potential solution for addressing parasitic drug resistance. This is mainly because of their remarkable immunomodulatory abilities, which can potentially help combat parasites' resistance to drugs.
Article
Full-text available
Exosomes are the smallest subset of extracellular signaling vesicles secreted by most cells with the ability to communicate with other tissues and cell types over long distances. Their use in regenerative medicine has gained tremendous momentum recently due to their ability to be utilized as therapeutic options for a wide array of diseases/conditions. Over 5000 publications are currently being published yearly on this topic, and this number is only expected to dramatically increase as novel therapeutic strategies continue to be developed. Today exosomes have been applied in numerous contexts including neurodegenerative disorders (Alzheimer's disease, central nervous system, depression, multiple sclerosis, Parkinson's disease, post‐traumatic stress disorders, traumatic brain injury, peripheral nerve injury), damaged organs (heart, kidney, liver, stroke, myocardial infarctions, myocardial infarctions, ovaries), degenerative processes (atherosclerosis, diabetes, hematology disorders, musculoskeletal degeneration, osteoradionecrosis, respiratory disease), infectious diseases (COVID‐19, hepatitis), regenerative procedures (antiaging, bone regeneration, cartilage/joint regeneration, osteoarthritis, cutaneous wounds, dental regeneration, dermatology/skin regeneration, erectile dysfunction, hair regrowth, intervertebral disc repair, spinal cord injury, vascular regeneration), and cancer therapy (breast, colorectal, gastric cancer and osteosarcomas), immune function (allergy, autoimmune disorders, immune regulation, inflammatory diseases, lupus, rheumatoid arthritis). This scoping review is a first of its kind aimed at summarizing the extensive regenerative potential of exosomes over a broad range of diseases and disorders.
Article
Full-text available
Introduction Osteoporotic-related fractures remains a significant public health concern, thus imposing substantial burdens on our society. Excessive activation of osteoclastic activity is one of the main contributing factors for osteoporosis-related fractures. While polylactic acid (PLA) is frequently employed as a biodegradable scaffold in tissue engineering, it lacks sufficient biological activity. Microdroplets (MDs) have been explored as an ultrasound-responsive drug delivery method, and mesenchymal stem cell (MSC)-derived exosomes have shown therapeutic effects in diverse preclinical investigations. Thus, this study aimed to develop a novel bioactive hybrid PLA scaffold by integrating MDs-NFATc1-silencing siRNA to target osteoclast formation and MSCs-exosomes (MSC-Exo) to influence osteogenic differentiation (MDs-NFATc1/PLA-Exo). Methods Human bone marrow-derived mesenchymal stromal cells (hBMSCs) were used for exosome isolation. Transmission electron microscopy (TEM) and confocal laser scanning microscopy were used for exosome and MDs morphological characterization, respectively. The MDs-NFATc1/PLA-Exo scaffold was fabricated through poly(dopamine) and fibrin gel coating. Biocompatibility was assessed using RAW 264.7 macrophages and hBMSCs. Osteoclast formations were examined via TRAP staining. Osteogenic differentiation of hBMSCs and cytokine expression modulation were also investigated. Results MSC-Exo exhibited a cup-shaped structure and effective internalization into cells, while MDs displayed a spherical morphology with a well-defined core-shell structure. Following ultrasound stimulation, the internalization study demonstrated efficient delivery of bioactive MDs into recipient cells. Biocompatibility studies indicated no cytotoxicity of MDs-NFATc1/PLA-Exo scaffolds in RAW 264.7 macrophages and hBMSCs. Both MDs-NFATc1/PLA and MDs-NFATc1/PLA-Exo treatments significantly reduced osteoclast differentiation and formation. In addition, our results further indicated MDs-NFATc1/PLA-Exo scaffold significantly enhanced osteogenic differentiation of hBMSCs and modulated cytokine expression. Discussion These findings suggest that the bioactive MDs-NFATc1/PLA-Exo scaffold holds promise as an innovative structure for bone tissue regeneration. By specifically targeting osteoclast formation and promoting osteogenic differentiation, this hybrid scaffold may address key challenges in osteoporosis-related fractures.
Article
Full-text available
Platelet-derived growth factor-BB (PDGF-BB) is a polypeptide growth factor generated by platelet granules faced to cytokines. It plays a role in forming and remodeling various tissue types, including epithelial tissue, through interaction with cell-surface receptors on most mesenchymal origin cells. However, it breaks down quickly in biological fluids, emphasizing the importance of preserving them from biodegradation. To address this challenge, we formulated and evaluated PDGF-encapsulated nanospheres (PD@PCEC) using polycaprolactone-polyethylene glycol-polycaprolactone. PD@PCECs were fabricated through the triple emulsion methodology and optimized by using the Box–Behnken design. The encapsulation efficiency (EE) of nanoencapsulated PDGF-BB was investigated concerning four variables: stirring rate (X1), stirring duration (X2), poly(vinyl alcohol) concentration (X3), and PDGF-BB concentration (X4). The selected optimized nanospheres were integrated into a gelatin-collagen scaffold (PD@PCEC@GC) and assessed for morphology, biocompatibility, in vitro release, and differentiation-inducing activity in human adipose-derived stem cells (hADSCs). The optimized PD@PCEC nanospheres exhibited a particle size of 177.9 ± 91 nm, a zeta potential of 5.2 mV, and an EE of 87.7 ± 0.44%. The release profile demonstrated approximately 85% of loaded PDGF-BB released during the first 360 h, with a sustained release over the entire 504 h period, maintaining bioactivity of 87.3%. The study also included an evaluation of the physicochemical properties of the scaffolds and an assessment of hADSC adhesion to the scaffold’s surface. Additionally, hADSCs cultivated within the scaffold effectively differentiated into keratinocyte-like cells (KLCs) over 21 days, evidenced by morphological changes and upregulation of keratinocyte-specific genes, including cytokeratin 18, cytokeratin 19, and involucrin, at both transcriptional and protein levels.
Article
Acute liver failure (ALF) is a rare and serious condition characterized by major hepatocyte death and liver dysfunction. Owing to the limited therapeutic options, this disease generally has a poor prognosis and a high mortality rate. When ALF cannot be reversed by medications, liver transplantation is often needed. However, transplant rejection and the shortage of donor organs still remain major challenges. Most recently, stem cell therapy has emerged as a promising alternative for the treatment of liver diseases. However, the limited cell delivery routes and poor stability of live cell products have greatly hindered the feasibility and therapeutic efficacy of stem cell therapy. Inspired by the functions of mesenchymal stem cells (MSCs) primarily through the secretion of several factors, we developed an MSC-inspired biomimetic multifunctional nanoframework (MBN) that encapsulates the growth-promoting factors secreted by MSCs via combination with hydrophilic or hydrophobic drugs. The red blood cell (RBC) membrane was coated with the MBN to enhance its immunological tolerance and prolong its circulation time in blood. Importantly, the MBN can respond to the oxidative microenvironment, where it accumulates and degrades to release the payload. In this work, two biomimetic nanoparticles, namely, rhein-encapsulated MBN (RMBN) and N-acetylcysteine (NAC)-encapsulated MBN (NMBN), were designed and synthesized. In lipopolysaccharide (LPS)/d-galactosamine (D-GalN)-induced and acetaminophen (APAP)-induced ALF mouse models, RMBN and NMBN could effectively target liver lesions, relieve the acute symptoms of ALF, and promote liver cell regeneration by virtue of their strong antioxidative, anti-inflammatory, and regenerative activities. This study demonstrated the feasibility of the use of an MSC-inspired biomimetic nanoframework for treating ALF.
Article
Full-text available
Pro-inflammatory cytokines can effectively be used for tumor immunotherapy, affecting every step of the tumor immunity cycle. Thereby, they can restore antigen priming, improve the effector immune cell frequencies in the tumor microenvironment (TME), and eventually strengthen their cytolytic function. A renewed interest in the anticancer competencies of cytokines has resulted in a substantial promotion in the number of trials to address the safety and efficacy of cytokine-based therapeutic options. However, low response rate along with the high toxicity associated with high-dose cytokine for reaching desired therapeutic outcomes negatively affect their clinical utility. Recently, mesenchymal stem/stromal cells (MSCs) due to their pronounced tropism to tumors and also lower immunogenicity have become a promising vehicle for cytokine delivery for human malignancies. MSC-based delivery of the cytokine can lead to the more effective immune cell-induced antitumor response and provide sustained release of target cytokines, as widely evidenced in a myriad of xenograft models. In the current review, we offer a summary of the novel trends in cytokine immunotherapy using MSCs as a potent and encouraging carrier for antitumor cytokines, focusing on the last two decades' animal reports.
Article
Full-text available
Background: Acute liver failure (ALF) is a complicated condition that is characterized by global hepatocyte death and often requires immediate liver transplantation. However, this therapy is limited by shortage of donor organs. Mesenchymal stem cells (MSCs) and hepatocytes are two attractive sources of cell-based therapies to treat ALF. The combined transplantation of hepatocytes and MSCs is considered to be more effective for the treatment of ALF than single-cell transplantation. We have previously demonstrated that HNF4α-overexpressing human umbilical cord MSCs (HNF4α-UMSCs) promoted the expression of hepatic-specific genes. In addition, microencapsulation allows exchange of nutrients, forming a protective barrier to the transplanted cells. Moreover, encapsulation of hepatocytes improves the viability and synthetic ability of hepatocytes and circumvents immune rejection. This study aimed to investigate the therapeutic effect of microencapsulation of hepatocytes and HNF4α-UMSCs in ALF mice. Methods: Human hepatocytes and UMSCs were obtained separately from liver and umbilical cord, followed by co-encapsulation and transplantation into mice by intraperitoneal injection. LPS/D-gal was used to induce ALF by intraperitoneal injection 24 h after transplantation. In addition, Raw 264.7 cells (a macrophage cell line) were used to elucidate the effect of HNF4α-UMSCs-hepatocyte microcapsules on polarization of macrophages. The protein chip was used to define the important paracrine factors in the conditioned mediums (CMs) of UMSCs and HNF4α-UMSCs and investigate the possible mechanism of HNF4α-UMSCs for the treatment of ALF in mice. Results: HNF4α-UMSCs can enhance the function of primary hepatocytes in alginate-poly-L-lysine-alginate (APA) microcapsules. The co-encapsulation of both HNF4α-UMSCs and hepatocytes achieved better therapeutic effects in ALF mice by promoting M2 macrophage polarization and reducing inflammatory response mainly mediated by the paracrine factor HB-EGF secreted by HNF4α-UMSCs. Conclusions: The present study confirms that the co-encapsulation of HNF4α-UMSC and hepatocytes could exert therapeutic effect on ALF mainly by HB-EGF secreted by HNF4α-UMSCs and provides a novel strategy for the treatment of ALF.
Article
Full-text available
Sepsis is the leading cause of death among patients, especially elderly patients, in intensive care units worldwide. In this study, we established a sepsis model using naturally aged rats and injected 5×10⁶ umbilical cord-derived MSCs via the tail vein. Each group of rats was analyzed for survival, examined for biochemical parameters, stained for organ histology, and analyzed for the Th cell subpopulation ratio and inflammatory cytokine levels by flow cytometry. Western blotting was performed to detect the activity of the JAK-STAT signaling pathway. We designed the vitro experiments to confirm the regulatory role of MSCs, and verified the possible mechanism using JAK/STAT inhibitors. It was revealed from the experiments that the 72 h survival rate of sepsis rats treated with MSCs was significantly increased, organ damage and inflammatory infiltration were reduced, the levels of organ damage indicators were decreased, the ratios of Th1/Th2 and Th17/Treg in peripheral blood and spleen were significantly decreased, the levels of pro-inflammatory cytokines such as IL-6 were decreased, the levels of anti-inflammatory cytokines such as IL-10 were increased, and the levels of STAT1 and STAT3 phosphorylation were reduced. These results were validated in in vitro experiments. Therefore, this study confirms that MSCs can control the inflammatory response induced by sepsis by regulating Th cells and inflammatory factors, and that this leads to the reduction of tissue damage, protection of organ functions and ultimately the improvement of survival in aged sepsis model rats. Inhibition of the JAK-STAT signaling pathway was surmised that it may be an important mechanism for their action.
Article
Full-text available
Background Amnion-derived mesenchymal stem cells (AM-MSCs) are an attractive source of stem cell therapy for patients with irreversible liver disease. However, there are obstacles to their use due to low efficiency and xeno-contamination for hepatic differentiation. Methods We established an efficient protocol for differentiating AM-MSCs into hepatic progenitor cells (HPCs) by analyzing transcriptome-sequencing data. Furthermore, to generate the xeno-free conditioned differentiation protocol, we replaced fetal bovine serum (FBS) with polyvinyl alcohol (PVA). We investigated the hepatocyte functions with the expression of mRNA and protein, secretion of albumin, and activity of CYP3A4. Finally, to test the transplantable potential of HPCs, we transferred AM-MSCs along with hepatic progenitors after differentiated days 11, 12, and 13 based on the expression of hepatocyte-related genes and mitochondrial function. Further, we established a mouse model of acute liver failure using a thioacetamide (TAA) and cyclophosphamide monohydrate (CTX) and transplanted AM-HPCs in the mouse model through splenic injection. Results We analyzed gene expression from RNA sequencing data in AM-MSCs and detected downregulation of hepatic development-associated genes including GATA6, KIT, AFP, c-MET, FGF2, EGF, and c-JUN, and upregulation of GSK3 . Based on this result, we established an efficient hepatic differentiation protocol using the GSK3 inhibitor, CHIR99021. Replacing FBS with PVA resulted in improved differentiation ability, such as upregulation of hepatic maturation markers. The differentiated hepatocyte-like cells (HLCs) not only synthesized and secreted albumin, but also metabolized drugs by the CYP3A4 enzyme. The best time for translation of AM-HPCs was 12 days from the start of differentiation. When the AM-HPCs were transplanted into the liver failure mouse model, they settled in the damaged livers and differentiated into hepatocytes. Conclusion This study offers an efficient and xeno-free conditioned hepatic differentiation protocol and shows that AM-HPCs could be used as transplantable therapeutic materials. Thus, we suggest that AM-MSC-derived HPCs are promising cells for treating liver disease.
Article
Full-text available
CD5 molecule like (CD5L), a member of the scavenger receptor cysteine-rich domain superfamily, plays a critical role in immune homeostasis and inflammatory disease. Acetaminophen (APAP) is a safe and effective antipyretic analgesic. However, overdose may cause liver damage or even liver failure. APAP hepatotoxicity is characterized by extensive necrotic cell death and a sterile inflammatory response, in which the role of CD5L remains to be investigated. In this study, we found that the expression of CD5L was increased in the livers of mice after APAP overdose. Furthermore, CD5L deficiency reduced the increase of alanine transaminase (ALT) level, histopathologic lesion area, c-Jun N-terminal kinase (JNK)/extracellular signal-regulated kinase (ERK) phosphorylation level, Transferase-Mediated dUTP Nick End-Labeling positive (TUNEL+) cells proportion, vascular endothelial cell permeability and release of inflammatory cytokines induced by excess APAP. Therefore, our findings reveal that CD5L may be a potential therapeutic target for prevention and treatment of APAP-induced liver injury.
Article
Full-text available
Background Vibrio cholerae is the causative agent of cholera, which is commonly associated with high morbidity and mortality, and presents a major challenge to healthcare systems throughout the world. Lipopolysaccharide (LPS) is required for full protection against V. cholerae but can induce inflammation and septic shock. Mesenchymal stem cells (MSCs) are currently used to treat infectious and inflammatory diseases. Therefore, this study aimed to evaluate the immune-modulating effects of the LPS‐MSC‐conditioned medium (CM) on V. cholerae LPS immunization in a murine model. Methods After preconditioning MSCs with LPS, mice were immunized intraperitoneally on days 0 and 14 with the following combinations: LPS + LPS-MSC-CM; detoxified LPS (DLPS) + MSC-CM; LPS + MSC sup; LPS; LPS-MSC-CM; MSC supernatant (MSC sup); and PBS. The mouse serum and saliva samples were collected to evaluate antibody (serum IgG and saliva IgA) and cytokine responses (TNF-α, IL-10, IL-6, TGF-β, IL-4, IL-5, and B-cell activating factor (BAFF)). Results The LPS + LPS-MSC-CM significantly increased total IgG and IgA compared to other combinations ( P < 0.001). TNF-α levels, in contrast to IL-10 and TGF-β, were reduced significantly in mice receiving the LPS + LPS-MSC-CM compared to mice receiving only LPS. IL-4, IL-5, and BAFF levels significantly increased in mice receiving increased doses of LPS + LPS-MSC-CM compared to those who received only LPS. The highest vibriocidal antibody titer (1:64) was observed in LPS + LPS-MSC-CM-immunized mice and resulted in a significant improvement in survival in infant mice infected by V. cholerae O1. Conclusions The LPS-MSC-CM modulates the immune response to V. cholerae LPS by regulating inflammatory and anti-inflammatory responses and inducing vibriocidal antibodies, which protect neonate mice against V. cholerae infection.
Article
Full-text available
Liver fibrosis is a common feature of chronic liver disease. Activated hepatic stellate cells (HSCs) are the main drivers of extracellular matrix accumulation in liver fibrosis. Hence, a strategy for regulating HSC activation is crucial in treating liver fibrosis. Mesenchymal stem cells (MSCs) are multipotent stem cells derived from various post-natal organs. Therapeutic approaches involving MSCs have been studied extensively in various diseases, including liver disease. MSCs modulate hepatic inflammation and fibrosis and/or differentiate into hepatocytes by interacting directly with immune cells, HSCs, and hepatocytes and secreting modulators, thereby contributing to reduced liver fibrosis. Cell-free therapy including MSC-released secretomes and extracellular vesicles has elicited extensive attention because they could overcome MSC transplantation limitations. Herein, we provide basic information on hepatic fibrogenesis and the therapeutic potential of MSCs. We also review findings presenting the effects of MSC itself and MSC-based cell-free treatments in liver fibrosis, focusing on HSC activation. Growing evidence supports the anti-fibrotic function of either MSC itself or MSC modulators, although the mechanism underpinning their effects on liver fibrosis has not been established. Further studies are required to investigate the detailed mechanism explaining their functions to expand MSC therapies using the cell itself and cell-free treatments for liver fibrosis.
Article
Full-text available
There is still a lively debate about whether mesenchymal stem cells (MSCs) promote or suppress antitumor immune response. Although several possible explanations have been proposed, including different numbers of injected and engrafted MSCs, heterogeneity in phenotype, and function of tumor cells, the exact molecular mechanisms responsible for opposite effects of MSCs in modulation of antitumor immunity are still unknown. Herewith, we used a B16F10 murine melanoma model to investigate whether timing of MSC administration in tumor-bearing mice was crucially important for their effects on antitumor immunity. MSCs, intravenously injected 24 h after melanoma induction (B16F10+MSC1d-treated mice), significantly enhanced natural killer (NK) and T cell-driven antitumor immunity, suppressed tumor growth, and improved survival of melanoma-bearing animals. Significantly higher plasma levels of antitumorigenic cytokines (TNF-α and IFN-γ), remarkably lower plasma levels of immunosuppressive cytokines (TGF-β and IL-10), and a significantly higher number of tumor-infiltrating, IFN-γ-producing, FasL- and granzyme B-expressing NK cells, IL-17-producing CD4+Th17 cells, IFN-γ- and TNF-α-producing CD4+Th1 cells, and CD8+cytotoxic T lymphocytes (CTLs) were observed in B16F10+MSC1d-treated mice. On the contrary, MSCs, injected 14 days after melanoma induction (B16F10+MSC14d-treated mice), promoted tumor growth by suppressing antigen-presenting properties of tumor-infiltrating dendritic cells (DCs) and macrophages and by reducing tumoricidal capacity of NK cells and T lymphocytes. Significantly higher plasma levels of TGF-β and IL-10, remarkably lower plasma levels of TNF-α and IFN-γ, and significantly reduced number of tumor-infiltrating, I-A-expressing, and IL-12-producing macrophages, CD80- and I-A-expressing DCs, granzyme B-expressing CTLs and NK cells, IFN-γ- and IL-17-producing CTLs, CD4+Th1, and Th17 cells were observed in B16F10+MSC14d-treated animals. In summing up, the timing of MSC administration into the tumor microenvironment was crucially important for MSC-dependent modulation of antimelanoma immunity. MSCs transplanted during the initial phase of melanoma growth exerted tumor-suppressive effect, while MSCs injected during the progressive stage of melanoma development suppressed antitumor immunity and enhanced tumor expansion. 1. Introduction Melanoma is nowadays considered as one of the most aggressive and the fastest growing malignant tumors worldwide [1]. Although a primary cutaneous melanoma can be managed by surgery, the advanced metastatic melanoma requires use of modern molecular mechanism-based therapeutic approaches [1]. The immuno- and targeted drug therapies, which interfere with oncoprotein and immune checkpoint pathways, were able to positively impact survival of patients with advanced melanoma [2]. Unfortunately, the success rate is being hampered by a number of factors including drug resistance, heterogeneous phenotype of melanoma cells, and impaired activation of antitumor immune response [2]. Therefore, new and more effective strategies are needed for patients who did not receive optimal benefit from currently used therapeutic approaches. Mesenchymal stem cells (MSCs) are nonhematopoietic, multipotent stem cells that reside in almost all postnatal tissues [3]. As cells of mesodermal origin, MSCs are considered as an integral part of the tumor stromal microenvironment, where, together with malignant cells, fibroblasts, pericytes, and endothelial cells, it produces trophic and growth factors and immuno- and angiomodulatory molecules and regulates tumor development [4]. Additionally, MSCs express a large number of chemokine receptors and exhibit strong tropism towards cancer cells [5]. After systemic administration, MSCs engraft in the tumor microenvironment where, in a juxtacrine and paracrine manner, it regulates expansion of malignant cells and modulates antitumor immunity [6]. Due to their tumor-homing capacity, MSCs were used as a vehicle to deliver cytotoxic drugs, proinflammatory cytokines, and cell cycle-interfering microRNAs in the tumors, attenuating their growth and progression [7]. MSCs modulate phenotype and function of all immune cells that play an important role in antitumor immune response [8]. MSCs regulate antigen-presenting properties of macrophages and dendritic cells (DCs), cytotoxicity of natural killer (NK) and CD8+T cells (CTLs), and cytokine production in CD4+T helper cells [8]. Accordingly, effects of MSC-dependent modulation of antitumor immunity have been explored in a large number of experimental studies, but surprisingly, opposite results were reported. While several research groups demonstrated that MSCs suppressed antitumor immune response and enhanced tumor progression [9–11], experimental findings presented by other researchers indicated that MSC-based therapy favored development of strong antitumor immunity that inhibited expansion of malignant cells [9–13]. Although several possible explanations for these contradictory findings have been proposed, including different numbers of injected and engrafted MSCs, diverse route of their administration, heterogeneity in phenotype, and function of tumor cells [12, 13], the exact molecular mechanisms responsible for opposite effects of MSCs in modulation of tumor growth are still unknown. Recently, Zong and colleagues indicated that MSC-based effects on progression and metastasis of hepatocellular carcinoma (HCC) depended on the time of MSC administration in the tumor-bearing animals [14]. Injection of MSCs in the initial phase of HCC development resulted in tumor suppression, while MSCs administered in the progressive stage of tumor growth promoted progression and metastasis of HCC [14]. In line with these findings, herewith, we used a murine model of melanoma to investigate whether the timing of MSC administration in melanoma-bearing mice was crucially important for MSC-dependent modulation of antitumor immunity and melanoma progression. 2. Material and Methods 2.1. Cells MSCs isolated from bone marrow of C57BL/6 mice were purchased from Gibco (Catalog number S1502-100). The murine melanoma cell line B16F10, which is syngeneic to the C57BL/6 background, was purchased from the American Type Culture Collection (CRL-6475; ATCC, USA). Both types of cells were cultured in complete Dulbecco’s modified Eagle medium (DMEM) containing 10% heat-inactivated fetal bovine serum (FBS), 100 IU/mL penicillin G, and 100 μg/mL streptomycin (Sigma-Aldrich, Munich, Germany), at 37°C in a 5% CO2 incubator. MSCs in passage 4 and B16F10 cells in passage 4 were used throughout the experiments. 2.2. Animals Eight- to ten-week-old C57BL/6 mice were used. Mice were maintained in animal breeding facilities of the Faculty of Medical Sciences, University of Kragujevac, Serbia. All procedures were performed in accordance with the guidelines for the Principles of Laboratory Animal Care and the Guide for the Care and Use of Laboratory Animals, and all animals received humane care according to the criteria outlined in the Guide for the Care and Use of Laboratory Animals (National Institutes of Health publication 86-23, 1985 revision). All experiments were approved by the Animal Ethical Review Board of the Faculty of Medical Sciences, University of Kragujevac, Serbia. Mice were housed in a temperature-controlled environment with a 12-hour light-dark cycle and were administered with standard laboratory chow and water ad libitum. At least eight mice per group were used in each experiment. 2.3. Melanoma Induction and Injection of MSCs B16F10 cells ( cells, suspended in 200 μL of phosphate-buffered saline (PBS)) were subcutaneously injected in the left flank of C57BL/6 mice. Immediately after, mice were divided into four experimental groups. The first experimental group of mice, 1 day after injection of B16F10 cells, intravenously received MSCs ( cells, suspended in 200 μL of PBS; B16F10+MSC1d-treated mice). The second experimental group of B16F10-treated animals, 14 days after administration of B16F10 cells, intravenously received MSCs ( cells, suspended in 200 μL of PBS; B16F10+MSC14d-treated mice). Mice from the third and fourth experimental groups intravenously received 200 μL of PBS at appropriate time points (1 day (B16F10+PBS1d-treated mice) or 14 days after B16F10 administration (B16F10+PBS14d-treated animals)). All animals were sacrificed 28 days after the injection of B16F10 cells. 2.4. Measurement of Tumor Growth and Progression Once the tumors were palpable, they were measured daily and tumor volume was calculated with the following formula: (, , and ) [15]. 2.5. Measurement of Cytokines in Plasma Samples of Tumor-Bearing Mice Blood samples were collected from the facial vein at days 1, 14, and 28 after the injection of B16F10 cells. Mouse blood was kept in anticoagulant-containing tubes and centrifuged for 10 minutes at 2000 g at 4°C. Supernatants were stored at -20°C until needed. Concentration of tumor necrosis factor alpha (TNF-α), interferon gamma (IFN-γ), transforming growth factor beta (TGF-β), and interleukin- (IL-) 10 in mouse plasma samples were measured by using enzyme-linked immunosorbent assay (ELISA) sets (R&D Systems, Minneapolis, MN, USA), according to the manufacturer’s instructions [16]. 2.6. Isolation of Tumor-Infiltrating Leucocytes By using forceps and scissors, subcutaneous tumors were resected en bloc, including overlying and surrounding skin. After the removal of surrounding skin, tumors were measured and weighed. By using scissors, the tumors were minced, until all large sections were processed into 1-2 mm pieces which are digested in 5 mL of DMEM containing 1 mg/mL collagenase I, 1 mM EDTA, and 2% FBS (all from Sigma-Aldrich, Munich, Germany). After incubation of 2 hr at 37°C, the digested tumor tissue was incubated with 4 mL of trypsin and DNase I (Roche Diagnostics), followed by passing through a 40 μm nylon filter. Single-cell suspensions were then processed for flow cytometry analysis [17]. 2.7. Flow Cytometry Analysis and Intracellular Staining of Tumor-Infiltrating Leucocytes Tumor-infiltrating leucocytes were investigated for different cell surface and intracellular markers with flow cytometry. Briefly, cells were incubated with anti-mouse F4/80, CD4, CD8, CD11c, NK1.1, CD80, I-A, granzyme B, and Fas ligand (FasL) monoclonal antibodies conjugated with fluorescein isothiocyanate (FITC), phycoerythrin (PE), peridinin chlorophyll protein (PerCP), or allophycocyanin (APC) (all from BD Biosciences, San Jose, CA, USA) following the manufacturer’s instructions. Immune cells derived from the tumors were concomitantly stained for the intracellular content of TNF-α, IFN-γ, IL-12, IL-4, and IL-17 by using the fixation/permeabilization kit and anti-mouse monoclonal antibodies conjugated with FITC, PE, PerCP, and APC (BD Biosciences). For intracellular cytokine staining, cells were stimulated with 50 ng/mL PMA and 500 ng/mL ionomycin for 5 h, and GolgiStop (BD Biosciences) was added. Cells were fixed in Cytofix/Cytoperm, permeated with 0.1% saponin, and stained with fluorescent Abs. Flow cytometric analysis was conducted on a BD Biosciences’ FACSCalibur and analyzed by using the Flowing Software analysis program [17]. 2.8. Statistical Analyses The data were analyzed using statistical package SPSS, version 21. The normality of distribution was tested by the Kolmogorov-Smirnov test. The results were analyzed using the Student -test. All data in this study were expressed as the . Values of were considered as statistically significant. 3. Results 3.1. MSC-Based Modulation of Melanoma Growth Depends on the Time of MSC Administration First, we examined whether systemic application of MSCs affected melanoma growth. As it is shown in Figure 1(a), tumors become palpable in B16F10+MSC1d-treated mice 8 days later compared with other experimental groups, suggesting that MSCs, intravenously injected 24 h after melanoma induction, prevented rapid tumor growth. Starting from day 18, average tumor volumes were significantly lower in B16F10+MSC1d-treated mice than in B16F10+PBS1d-treated animals (; Figure 1(a)). Additionally, the average volume and weight of tumors removed from B16F10+MSC1d-treated mice at day 28 were significantly lower than melanomas taken from B16F10+PBS1d-treated animals (Figures 1(b) and 1(c)), confirming that MSCs, intravenously injected 24 h after melanoma induction, efficiently suppressed tumor growth and progression. (a)
Article
Objective The treatment of liver failure by stem cell transplantation has attracted growing interest. Herein, we aim to explore the role of sodium butyrate (NaB) in the hepatic differentiation of bone marrow mesenchymal stem cells (BM-MSCs) under liver-specific factors induction in vitro and vivo. Materials & Methods We isolated BM-MSCs from the mononuclear cell fraction of rabbit bone marrow samples, and identified the cells by Immunophenotypic analysis. We investigated the effects of different concentrations and induction conditions. The histone deacetylase inhibitor NaB induced hepatic differentiation of BM-MSCs under liver-specific factors induction in vitro. Morphological features, liver-specific gene and protein expression, and functional analyses in vitro and vivo were performed to evaluate the hepatic differentiation of BM-MSCs. Results Our results showed that pre-treated NaB inhibited the expression of liver-specific protein in a dose-dependent manner. The induction efficiency of NaB with 24h pre-treatment was higher than that of NaB continuous intervention. 0.5 mM 24h NaB pre-treated cells can improve liver tissue damage in vivo. And the liver ALB, AAT and the serum TP were significantly increased, while the serum ALT was significantly reduced. Conclusion Continuous NaB treatment can inhibit BM-MSCs proliferation in a dose-dependent manner at a certain concentration range. 0.5 mM 24h pre-treatment of NaB enhanced differentiation of BM-MSCs into hepatocytes and improves liver injury in vitro and vivo.
Article
Background & aims: Liver dysfunctions are classified into acute and chronic diseases, which comprise a heterogeneous group of pathological features and a high mortality rate. Liver transplantation still remains the gold standard therapy for most of liver diseases with concomitant limitations related to donor organ shortage and life-long immunosuppressive therapy. Novel concept in liver therapy intends to overcome these limitations based on the secreted extracellular vesicles (EVs; microvesicles and exosomes) by mesenchymal stem/stromal cells (MSCs). A significant number of studies have shown that factors released by MSCs could induce liver repair and ameliorate systemic inflammation through paracrine effects. It is well known that this paracrine action is based not only on the secretion of cytokines and growth factors, but also on EVs, which regulate pathways associated with inflammation, hepatic fibrosis, integrin-linked protein kinase (ILK) signaling and apoptosis. Approach & results: Herein, we aim to extensively discuss the differential effects of MSC-EVs on different liver diseases, on cellular and animal models, and to address the complex molecular mechanisms involved in therapeutic potential of EVs. In addition, we aim to cover the crucial information regarding the type of molecules contained in MSC-EVs that can be effective in the context of liver diseases. Conclusion: In conclusion, outcomes on MSC-EV mediated therapy are expected to lead to an innovative, cell free, non-invasive, less immunogenic and non-toxic alternative strategy for liver treatment and to provide important new mechanistic information on the reparative function of liver cells.