ArticlePDF AvailableLiterature Review

Mesenchymal stromal cells in hepatic fibrosis/cirrhosis: from pathogenesis to treatment

Authors:

Abstract and Figures

Hepatic fibrosis/cirrhosis is a significant health burden worldwide, resulting in liver failure or hepatocellular carcinoma (HCC) and accounting for many deaths each year. The pathogenesis of hepatic fibrosis/cirrhosis is very complex, which makes treatment challenging. Endogenous mesenchymal stromal cells (MSCs) have been shown to play pivotal roles in the pathogenesis of hepatic fibrosis. Paradoxically, exogenous MSCs have also been used in clinical trials for liver cirrhosis, and their effectiveness has been observed in most completed clinical trials. There are still many issues to be resolved to promote the use of MSCs in the clinic in the future. In this review, we will examine the controversial role of MSCs in the pathogenesis and treatment of hepatic fibrosis/cirrhosis. We also investigated the clinical trials involving MSCs in liver cirrhosis, summarized the parameters that need to be standardized, and discussed how to promote the use of MSCs from a clinical perspective.
Content may be subject to copyright.
REVIEW ARTICLE OPEN
Mesenchymal stromal cells in hepatic brosis/cirrhosis: from
pathogenesis to treatment
Xue Yang
1,2,3,4,6
, Qing Li
5,6
, Wenting Liu
1,2,6
, Chen Zong
1,2
, Lixin Wei
1,2
, Yufang Shi
3,4
and Zhipeng Han
1,2
© The Author(s), under exclusive licence to CSI and USTC 2023
Hepatic brosis/cirrhosis is a signicant health burden worldwide, resulting in liver failure or hepatocellular carcinoma (HCC) and
accounting for many deaths each year. The pathogenesis of hepatic brosis/cirrhosis is very complex, which makes treatment
challenging. Endogenous mesenchymal stromal cells (MSCs) have been shown to play pivotal roles in the pathogenesis of hepatic
brosis. Paradoxically, exogenous MSCs have also been used in clinical trials for liver cirrhosis, and their effectiveness has been
observed in most completed clinical trials. There are still many issues to be resolved to promote the use of MSCs in the clinic in the
future. In this review, we will examine the controversial role of MSCs in the pathogenesis and treatment of hepatic brosis/cirrhosis.
We also investigated the clinical trials involving MSCs in liver cirrhosis, summarized the parameters that need to be standardized,
and discussed how to promote the use of MSCs from a clinical perspective.
Keywords: Mesenchymal stromal cells; Hepatic brosis/cirrhosis; Pathogenesis; Treatment; Clinical application
Cellular & Molecular Immunology; https://doi.org/10.1038/s41423-023-00983-5
INTRODUCTION
Liver diseases present a signicant threat to human health
globally, resulting in ~2 million deaths each year. Hepatic cirrhosis
accounts for ~50% of liver disease-associated deaths [1]. Liver
cirrhosis is a complication of liver disease, which is a further step
toward hepatic brosis and involves the loss of liver cells and
irreversible liver scarring. Chronic damage due to factors such as
viral hepatitis, alcohol, and drugs impairs hepatocytes. This
damage induces inammatory cell inltration, leading to the
excessive accumulation of collagen and extracellular matrix (ECM)
and destroying liver structure and function [2,3]. Activated
hepatic stellate cells (HSCs) are the primary source of ECM and
contribute to hepatic brosis [4]. Increasing evidence has
demonstrated that MSCs are involved in the pathological
progression of hepatic brosis [5,6] through changes in their
functions and microenvironment regulation.
MSCs were rst discovered in bone marrow (BM) by Frieden-
stein and colleagues in 1968 and were described as an adherent,
broblast-like population in BM in vitro that could differentiate
into bone in vivo [7]. Subsequently, numerous studies have
revealed the plasticity, high proliferation, multidifferentiation, and
immunomodulatory capacity of these cells. MSCs are also present
in other tissues, including adipose (AD) tissue, umbilical cord (UC),
fetal liver, muscle, and lung [812]. MSCs have the ability to
differentiate into multiple lineages, such as chondrocytes,
osteocytes, adipocytes, myocytes, and astrocytes. MSCs express
major histocompatibility complex (MHC) class I but do not express
MHC class II, B7-1, B7-2, CD40, or CD40L. MSCs can be expanded
more than 10
4
-fold in culture without the loss of their multilineage
differentiation potential. Therefore, MSCs could be a potential
source of stem cells for cellular and genetic therapy [9,13].
Exogenous MSCs have been shown to exert antibrotic effects,
and various mechanisms have been examined. MSCs provide an
alternative for patients with liver cirrhosis because there is no
effective traditional treatment other than liver transplantation,
which is expensive and takes a long time to nd a suitable organ.
MSCs are considered to be the most promising cells for cell
therapy due to the following properties: (1) they are easy to obtain
from various tissues and easy to expand in vitro without changing
their properties [8,10] (2) they allow for allogeneic transplantation
because of their low immunogenicity and lack of ethical issues
[14,15]; (3) they migrate to injury sites; [16] (4) they have multiple
differentiation capacity and favorable regeneration and injury
repair effects [9,17]; and (5) they have strong immunoregulatory
abilities [18,19]. There have also been bench studies focusing on
new strategies to improve the therapeutic efcacy of MSCs in liver
cirrhosis. Although MSCs have been used to treat liver cirrhosis in
clinical trials and good outcomes have been observed in most
cases, many problems need to be solved to promote the use of
MSCs in the clinic in the future. In this review, we aimed to
Received: 14 November 2022 Accepted: 29 January 2023
1
Department of Tumor Immunology and Gene Therapy Center, Third Afliated Hospital of Naval Medical University, Shanghai 200438, China.
2
Key Laboratory on Signaling
Regulation and Targeting Therapy of Liver Cancer, Ministry of Education, Eastern Hepatobiliary Surgery Hospital/National Center for Liver Cancer, Naval Medical University,
Shanghai 200438, China.
3
The Third Afliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection,
Key Laboratory of Stem Cells and Medical Biomaterials of Jiangsu Province, Medical College of Soochow University, Soochow University, Suzhou 215000, China.
4
Department of
Experimental Medicine, TOR, University of Rome Tor Vergata, 00133 Rome, Italy.
5
CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and
Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China.
6
These authors contributed equally: Xue Yang, Qing Li, Wenting Liu.
email: shiyufang2@gmail.com; hanzhipeng0311@126.com
www.nature.com/cmi
1234567890();,:
examine the different roles of MSCs in the pathogenesis and
treatment of hepatic brosis/cirrhosis, propose parameters that
need to be standardized or optimized and suggest prospects for
the use of MSCs to treat hepatic brosis/cirrhosis.
ENDOGENOUS MSCS IN THE PATHOGENESIS OF HEPATIC
FIBROSIS
Contribution of MSCs to the population and regulation of
myobroblasts
Myobroblasts: The heart of brosis. Myobroblasts (MFs) were
rst described by the seminal works of Gabbiani et al. in 1971,
who showed that a population of modied broblastswas
present in granulation tissue during wound healing [20]. The
researchers found that these typical broblasts (1) possessed
brillar systems within the cytoplasm; (2) had a deformed
nucleus; and (3) were attached to an extracellular layer of
basement membrane-like material [20]. Due to their similarities
with smooth muscle cells, these unusual broblasts were termed
muscle-broblast intermediate cells and are now referred to as
myobroblasts[21]. The presence of MFs in tissue injury led to a
series of investigations of their roles in wound repair. Myobro-
blast lamentous bers were found to integrate with myosin and
α-smooth muscle actin (α-SMA) proteins through actin [22]. This
unique cytoskeleton of MFs allowed them to exert contractile
forces and to contract the wound edge, and α-SMA is now
regarded as a universal marker of MFs [23,24]. ECM production is
another hallmark of MFs. ECM components, including different
types of brillar collagens, hyaluronan (HA), bronectin (FN), and
extra domain A bronectin, are essential for closing wounds [25]. It
is important to note that dysregulated wound healing, which
frequently occurs in chronic injury, leads to tissue brosis. Due to
awareness of the essential role of MFs in tissue repair, researchers
quickly examined MFs in the context of organ brosis. They
identied MFs as the primary source of excess ECM deposition in
the brotic area [26]. Furthermore, multiple pieces of evidence
suggest that the status of MFs could balance tissue repair and
brosis [27]. Therefore, understanding the origin, modulation, and
remission of MFs is central to developing treatments for hepatic
brosis/cirrhosis.
MSCs are one of the sources of MFs in the liver. The origins of MFs
have long been a central debate in the eld. In the late 1970s, MFs
were initially thought to be derived from smooth muscle cells [28].
Later, investigators showed that MFs could develop directly from
normal broblasts, MSCs, epithelial cells, pericytes, and circulating
brocytes [29]. These contradictory ndings complicated the eld,
and a lack of consensus on the sources of MFs was a signicant
obstacle to understanding organ brosis. The groundbreaking
technological advances in lineage tracing and single-cell analysis
have enabled the detailed characterization of MFs under
homeostatic and disease conditions. The latest conceptual
advance on MFs is that they are a heterogeneous population of
cells whose origins and properties differ in various organs. The
following section focuses on the relationship between MSCs and
MFs during hepatic brosis.
The notion that MSCs were a source of MFs rst came from
in vitro studies [5]. Transforming growth factor β(TGF-β), which is
a major probrotic cytokine, was shown to induce α-SMA
expression in MSCs. These α-SMA
+
MSCs produce ECM, validating
the transition of MSCs to MFs. Notably, the inammatory
properties of MSCs suggest that these in vitro observations may
also occur under pathological conditions. Taking advantage of a
dual uorescent labeling system, Michael Quante et al. found
that BM-derived MSCs were recruited to the site of chronic
inammation and gave rise to α-SMA
+
MFs during Helicobacter
felis-induced gastric cancer [6]. The spatiotemporal coexistence of
BM-MSCs and TGF-βduring liver injury repair was likely to produce
BM-MSC-derived hepatic MFs. In addition to BM niches, MSCs are
found in almost all tissues. These tissue-resident MSCs are located
in perivascular niches and are capable of trilineage differentiation
in vivo [30]. Recent studies using lineage tracing in mice showed
that Gli1 is a marker for resident MSCs in the liver, kidney, lung,
heart, and muscle [30]. Gli1
+
MSCs constitute 0.02% of total live
cells in the liver. These liver-resident MSCs also express CD29,
CD44, CD105, Sca1, and CD34. During carbon tetrachloride (CCl
4
)-
induced hepatic brosis, hepatic Gli1
+
MSCs dramatically expand
and induce the expression of α-SMA in a TGF-β-dependent
manner [30]. Notably, Gli1
+
MSCs contribute to ~39% of interstitial
α-SMA
+
cells in hepatic brosis in Gli1-CreERt2-tdTomato tracing
mice. Ablation of Gli1
+
MSCs was reported to alleviate the severity
of tissue brosis.
MSCs regulate myobroblast generation from hepatic stellate
cells. While the contribution of MSCs to the hepatic myobro-
blast pool was recently validated, it is generally believed that HSCs
are the primary sources of MFs during hepatic brosis. HSCs
are pericyte-like cells with abundant intracellular vitamin A and
lipids [31]. Fate mapping studies using acyltransferase-Cre mice
revealed that more than 80% of hepatic MFs were derived from
HSCs [32]. When we combine the data on Gli1
+
MSCs and HSCs,
there is a potential paradox: the sum of MSC-derived MFs and
HSCs exceeds 100%. A possible explanation for this contradiction
is that MSCs and HSCs have overlapping subpopulations. HSCs
expanded in vitro express MSC-like surface markers and show
adipogenic and osteogenic differentiation potential [33]. Future
studies are required to delineate the similarities between hepatic
MSCs and HSCs.
HSCs are nonproliferative in the steady state, and MF
differentiation in these cells is initiated by inammatory cytokines
released by injured hepatocytes or immune cells. MSCs have been
reported to regulate the activation of HSCs. MSCs have been
reported to express soluble and membrane-bound TGF-β, which
may provide signals for the myobroblast differentiation of HSCs
[34]. However, MSCs were shown to directly suppress the
proliferation of HSCs by the surface expression of Notch1, which
downregulates the PI3K/AKT pathway in HSCs in an in vitro
coculture system [35]. It has also been reported that direct contact
with MSCs induces the expression of proapoptotic proteins, such
as Bax and cleaved caspase-3, in HSCs [36]. In addition to cellcell
contact, MSCs may affect HSCs through paracrine effects.
Hepatocyte growth factor (HGF) and nerve growth factor (NGF),
which are secreted by MSCs, were shown to impair the activation
of HSCs by inhibiting nuclear factor kappa B (NF-κB) signaling [37].
The rationale for these in vitro ndings is that MSCs and HSCs are
both located in perivascular niches, allowing for potential cellcell
communication or paracrine effects. However, the reciprocal
interactions between MSCs and HSCs during the pathogenesis
of hepatic brosis remain to be determined.
Immunomodulation of MSCs in hepatic brosis
Investigations of stem cells have primarily focused on elucidating
the mechanisms underlying their self-renewal and differentiation
properties. However, most studies of MSCs have been related to
their roles in immune regulation [38]. The rst paper linking MSCs
with immunoregulation comes from the observation that the
presence of MSCs effectively suppressed mixed lymphocyte
reactions [39]. The immunosuppressive capabilities of MSCs were
then rmly validated by the successful clinical application of MSCs
to treat a severe steroid-resistant GvHD patient in 2004 [40].
However, when MSC-mediated immunosuppression was inten-
sively studied, controversial results also emerged. The rst FDA-
approved MSC-based clinical trial to treat GvHD failed to achieve
signicant clinical improvements [41]. Later, it was found that the
immunosuppressive effects of MSCs were not intrinsic but were
triggered by inammatory cytokines [42,43]. Depending on the
X. Yang et al.
2
Cellular & Molecular Immunology
context of the cytokine milieu, MSCs can exert proinammatory or
anti-inammatory effects by modulating adaptive and innate
immune cells [38]. Based on the concept that MSCs are plastic in
the context of immunoregulation, we will discuss how the
interaction of MSCs with different types of immune cells affects
the progression of hepatic brosis.
Initiation of early inammation in liver injury. Hepatic brosis is a
dysregulated wound-healing response to liver injury [44]. If not
appropriately controlled, the acute phase of liver injury is the
starting point of hepatic brosis. Damage-associated molecular
patterns (DAMPs) released from apoptotic or necrotic hepatocytes
initiate the inammatory response by recruiting innate immune
cells to the site of tissue injury by secreting chemokines, including
C-C motif chemokine ligand 2 (CCL2), C-C motif chemokine ligand
5 (CCL5), C-X-C motif ligand 1, and C-X-C motif ligand 15 [45].
HSCs and liver-resident macrophages are believed to be the
sources of chemokines induced by liver injury [45]. MSCs are also
immune sentinels that produce chemokines in response to danger
signals or cytokines in vivo [46,47]. It has been reported that BM-
MSCs can sense low levels of circulating microbial molecules and
mediate monocyte mobilization by producing CCL2 [46]. Notably,
most types of liver insult are due to viral infection, alcohol, and
nonalcoholic steatohepatitis (NASH), which do not yield high
levels of danger signals [44]. In this context, the possibility that
liver-resident MSCs initiate inammation in response to liver injury
should be considered since the signaling threshold for chemokine
induction is low in MSCs [48].
Neutrophils are the rst leukocytes to respond to chemokines
released from the injured liver, and the inltration of neutrophils is
commonly observed in patients with steatohepatitis [49,50].
Generally, neutrophils have a short half-life [51]. However, the
viability of neutrophils is increased in the presence of MSCs [52].
MSCs constitutively express interleukin 6 (IL-6) and granulocyte-
macrophage colony-stimulating factors to inhibit neutrophil
apoptosis without affecting the phagocytic function of neutrophils
[52,53]. The postponed death of neutrophils provides a window
to eliminate damaged hepatocytes, which is energy efcient from
an evolutionary perspective.
Resolution of liver inammation. Acute immune responses
corresponding to the accumulation of neutrophils and T cells
lead to the abundant production of inammatory cytokines in
the liver [54]. Activated tissue-resident natural killer T cells and
newly arrived neutrophils express high levels of interferon-
gamma (IFN-γ), IL-1, and tumor necrosis factor α(TNF-α)[
5557].
It has been shown that MSCs are highly immunosuppressive in
response to proinammatory cues [58]. Treatment of MSCs with
a combination of IFN-γand TNF-αor IL-1βinduced massive
production of immunosuppressive molecules, including nitric
oxide (NO) in rodent MSCs and indoleamine 2,3-dioxygenase
(IDO) in humans [43,59,60], as well as prostaglandin E2
(PGE2) [61]andTGF-β[62], leading to the subsequent inhibition
of T-cell proliferation and the induction of anti-inammatory
innate immune cells. The immunosuppressive molecules and the
apparent chemokine gradient surrounding activated MSCs
synergistically form MSC-centered immunosuppressive niches
in liver tissue. Indeed, intravital imaging showed that MSCs
colocalized with macrophages in hepatic brosis [63]. The
inammation-induced immunosuppressive functions of MSCs
were thought to reshape the immune microenvironment of the
liver during injury. However, due to a lack of specic markers for
MSCs and their activation status, the MSC-centered immuno-
suppressive population during hepatic brosis has not been
elucidated in vivo. Recent advances in mass spectrometry and
imaging are promising for investigating the localization-
dependent immunosuppressive capabilities of MSCs in hepatic
brosis [64].
To re-establish immune homeostasis in inammatory tissues,
MSCs have been shown to enhance apoptosis in T cells, suppress
the differentiation of T helper 17 (Th17) cells, and induce the
generation and accumulation of regulatory T (Treg) cells through
high expression of inducible nitric oxide synthase (iNOS), IDO,
tumor necrosis factor-stimulated gene-6 (TSG6) and matrix
metalloproteinases (MMPs) [43,48,65,66]. These observations
were consistent with the signicant increase in Treg cells and a
decrease in Th17 cell inltration in brotic liver tissues after the
transfusion of MSCs [67,68]. It was reported that the presence of
IL-17 could enhance the stability of iNOS and programmed
death ligand 1 (PD-L1) mRNA by modulating ARE/poly (U)-
binding/degradation factor 1 (AUF1), thus promoting the
immunosuppressive abilities of MSCs [69]. Therefore, MSCs
activated by acute inammatory cues can control excessive
immune responses and sustain tissue homeostasis.
In addition to T cells, macrophages play essential roles in
immune homeostasis in the liver. Recent studies have shown
that MSCs can impart anti-inammatory properties to macro-
phages during the monocyte-to-macrophage transition [70].
Specically, these effects depend on insulin-like growth factor 2
(IGF2) production by MSCs. IGF2 preprograms maturing macro-
phages toward metabolic commitment to oxidative phosphor-
ylation (OXPHOS) [70].Eveninthepresenceofproinammatory
cues, IGF2-preprogrammed macrophages could exert vital anti-
inammatory effects by upregulating PD-L1 expression [70].
Therefore, the anti-inammatory macrophages trained by MSC-
derived IGF2 could be another layer in the mechanism
that quenches inammation during liver injury. Notably, IGF2
has a dose-dependent effect on training macrophages. At low
concentrations, IGF2 prefers to bind the IGF2 receptor on
monocytes to confer macrophages with anti-inammatory
properties [71]. However, when the amount of IGF2 was
sufcient to ligate the IGF1 receptor on monocytes, the resultant
macrophages were proinammatory. The balance between IGF2
receptor and IGF1 receptor signaling is critical in determining
the functions of macrophages.
Maintenance of chronic inammation in hepatic brosis. During
the chronic inammation phase, low levels of inammatory stimuli
induce MSCs to continuously secrete chemokines and recruit
immune cells to the liver microenvironment [43,48,72]. It was
reported that iNOS and IDO act as switches in MSC-mediated
immunomodulation, and the stimulation intensity triggers a
switch between pro- and anti-inammatory functions [48,59].
When iNOS in murine MSCs or IDO in human MSCs was genetically
ablated, the immunosuppressive effects of IFN-γ- and TNF-α-
treated MSCs were diminished, and a signicant immunostimu-
latory effect was observed [48,59]. As we have previously
discussed, potent inammation in the acute phase initiates the
immunosuppressive functions of MSCs. While the cytokines in liver
tissue are inadequate to induce the expression of iNOS or IDO,
these MSCs can be activated under suboptimal conditions and
secrete chemokines, such as C-X-C motif ligand 9 (CXCL9) and
CCL5, which enables them with immunostimulatory capabilities
[43,48]. The dual functions of MSCs in immunomodulation might
partially account for the chronic inammation during the
progression of hepatic brosis.
TGF-βis a major pathogenic cytokine in hepatic brosis that is
responsible for MF induction. However, TGF-βalso plays an essential
role in immunosuppression, inhibiting the activation of immune
cells and inducing the generation of Treg cells [73]. However, why
TGF-βsignaling fails to resolve chronic inammation in the liver
remains unclear. Interestingly, investigators found that TGF-βmay
promote immune responses through interactions with MSCs [74].
TGF-βcould induce the immunosuppressive effects of MSCs by
inhibiting inammatory cytokine-induced iNOS expression in a
SMAD3-dependent manner, thus sustaining inammation.
X. Yang et al.
3
Cellular & Molecular Immunology
THERAPEUTIC MECHANISMS OF EXOGENOUS MSCS IN
HEPATIC FIBROSIS
As we summarized previously, liver injury-related factors, includ-
ing viruses and alcohol, can lead to hepatocyte damage and
chronic inammation in the liver. During chronic inammatory
injury of the liver, the synergistic effects of inammation and
hepatocyte death promote the development of hepatic brosis.
The role and mechanism of MSCs in the treatment of hepatic
brosis are thoroughly discussed in this section (Fig. 1).
MSCs in the context of hepatocyte injury
Differentiation into hepatocytes. Continuous hepatocyte death is
an essential pathological phenomenon in hepatic brosis. Due to
the multidifferentiation potential of MSCs, targeting the hepatic
differentiation potential of MSCs is expected to become an
alternative strategy for treating hepatic brosis. Several studies
have shown the hepatic differentiation of MSCs in experimental
animal models and humans [7578]. For example, after direct
xenografting into allyl alcohol (AA)-treated rat livers, human bone
marrow-derived MSCs could differentiate into hepatocyte-like
cells [79]. After being infused into mice with CCl
4
-induced
liver injury, MSCs or MSC-derived hepatocytes were found in the
liver [80]. The liver tropism of MSCs in response to hepatocyte
injury is regulated by the coordinated effects of growth factors
and damage signals [81]. HGF is the initial driving force that
recruits MSCs to the liver. The migration of MSCs is terminated
by adenosine, a metabolite that indicates tissue damage, thus
retaining MSCs in the area of hepatocyte injury [81]. The
transplantation of MSC-derived hepatocyte-like cells could effec-
tively attenuate hepatic brosis and protect liver function [82].
It has been reported that cocktail treatment of cytokines,
including HGF, epidermal growth factor (EGF), broblast growth
factor (FGF), leukemia inhibitory factor, dexamethasone (DEX),
and nicotinamide could effectively induce the differentiation of
MSCs toward hepatocyte-like cells [8386]. Coculture with liver
cells or pellet culture can also lead to the differentiation of MSCs
into hepatocytes [87,88]. In the absence or presence of growth
factors, a dynamic cultured scaffold stimulated MSCs to express
endodermal and hepatocyte-specic genes and proteins asso-
ciated with improved functions, and the cells exhibited the
ultrastructural characteristics of mature hepatocytes [77]. In
addition, Maryam et al. demonstrated that Whartons jelly-
derived MSCs could differentiate into hepatocyte-like cells by
permeabilization in the presence of HepG2 cell extracts [89].
However, compared with their differentiation into hepato-
cytes, the differentiation efciency of MSCs into MFs is higher,
which limits their application value in clinical therapy. However,
the transdifferentiation of MSCs into hepatocytes has rarely
been observed in animal models [90]. Therefore, it is necessary
to examine a more efcient method to induce the differentiation
of MSCs into hepatocytes. On the one hand, the biomimetic
microenvironment constructed by human E-cadherin fusion
protein (hE-cad-Fc)-coated poly (lactic-co-glycolic acid) (PLGA)
microparticles (hE-cad-PLGAs) in engineered multicellular aggre-
gates was able to promote endoderm differentiation and the
subsequent hepatic differentiation of human MSCs [91]. On the
other hand, Wang et al. found that the viability and hepatogenic
differentiation of BM-MSCs cultured on plates coated with
Matrigel and ECM were signicantly enhanced compared with
those of cells cultured on noncoated plates [92]. Posttranscrip-
tional regulation also plays an important role in regulating the
differentiation of MSCs into hepatocytes. Zhou et al. screened
the best miRNA candidates for hepatocyte differentiation and
found that ve miRNAs, including miR-122, miR-148a, miR-424,
miR-542-5p, and miR-1246, were essential for hepatocyte
differentiation because omitting anyone from the ve miRNAs
prevented hepatic transdifferentiation [93].
Protection of injured hepatocytes. Increasing evidence has shown
that the production of trophic factors, including cytokines,
chemokines, and growth factors, is essential for MSCs to
contribute to damage repair. Most injured hepatocytes undergo
cell death. However, the distinct forms of cell death and cell
death response pathways used by damaged hepatocytes have
long-term consequences on liver repair [94]. Another pathway
by which MSCs participate in liver damage repair involves the
production of paracrine trophic factors by MSCs through the
paracrine pathway [95,96]. It has been reported that MSC-
derived conditioned medium (CM) effectively inhibits hepato-
cyte death and promotes liver regeneration [97]. The adminis-
tration of MSC-CM to rats with acute liver injury dramatically
prevents apoptotic hepatocellular death [97]. HGF, αFGF, EGF,
and TGF-αcontribute to hepatocyte proliferation, and vascular
endothelial growth factor (VEGF) is the most vital pro-vascular
growth factor that plays a crucial role in angiogenesis during
liver regeneration [98101]. Gokhan Adas et al. showed that
after the injection of MSCs and VEGF-transfected MSCs into the
portal vein following liver resection, the cells engrafted in the
Fig. 1 The role of MSCs in hepatic brosis therapy
X. Yang et al.
4
Cellular & Molecular Immunology
liver, increased bile duct and hepatocyte proliferation and
secreted many growth factors, including HGF, TGF-β,VEGF,
platelet-derived growth factor, EGF, and FGF, via paracrine
effects [102]. In addition, hepatic progenitor cells (HPCs), which
are located in the canals of Hering, are also involved in liver
damage repair by differentiating toward hepatocytes or biliary
lineage cells. EGF and HGF have been reported to play important
roles in HPC differentiation [103]. In addition, MSC-derived
exosomes were shown to suppress ferroptosis in CCl
4
-induced
acute liver injury. Mechanistically, MSC-derived exosomes
stabilized the protein level of SLC7A11 in primary hepatocytes
to downregulate prostaglandin-endoperoxide synthase 2 and
lipoxygenases [104]. Interestingly, MSCs were also shown to
protect hepatocytes by promoting hepatocyte autophagy
through paracrine effects. Let-7a-5p, which is enriched in MSC-
derived exosomes, can activate autophagy by targeting
mitogen-activated protein kinase kinase kinase kinase 3
(MAP4K3) in hepatocytes [105]. MSCs can also exert hepatopro-
tective effects against liver damage by upregulating PTEN-
induced kinase 1-dependent mitophagy [106]. Therefore, MSCs
could shift hepatocyte death toward an ordered mode to limit
tissue damage and inammation, thus reducing the incidence of
hepatic brosis.
Inhibition of HSC activation
Studies on brosis have always used the strategy of inhibiting the
activation of HSCs to achieve an antibrotic effect. Therefore, the
impact of MSCs on HSC activation is particularly important for the
effective treatment of hepatic brosis. It has been reported that in
a CCl
4
-induced rat hepatic brosis model, MSC transplantation
effectively ameliorated hepatic brosis by inhibiting HSC pro-
liferation and promoting HSC apoptosis [107]. Several studies
conrmed the suppressive effect of MSCs on HSCs. TGF-β3 and
HGF derived from MSCs lead to arrest in the G0/G1 phase in HSCs
by decreasing extracellular signal-regulated kinase 1/2 phosphor-
ylation and thereby ameliorating hepatic brosis [108]. MSCs
can also decrease HSC proliferation by activating the Notch
signaling pathway and downregulating the PI3K/AKT signaling
pathway [35]. It has also been reported that hUCMSCs inhibit the
proliferation of HSCs, possibly by inhibiting TGF-β1 and Smad3
expression and increasing Smad7 protein expression [109]. In
addition, MSCs could induce apoptosis in HSCs. Transfer of
circDIDO1 mediated by MSC-derived exosomes suppresses pro-
liferation, reduces probrotic markers, and induces apoptosis and
cell cycle arrest in HSCs through the miR-141-3p/PTEN/AKT
pathway [110]. Lin et al. demonstrated that NGF produced by
MSCs increased HSC apoptosis via the NF-κB- and BCL-xl-
associated signaling pathways [111]. In addition, HSC activation
is triggered by the inammatory microenvironment. MSCs are well
known for their immunosuppressive properties, and MSCs can also
alleviate the expression of inammatory factors that induce HSC
activation and brosis [112].
On the other hand, MSCs play key roles in ECM degradation.
MFs can internalize MSC-derived extracellular vesicles (EVs) to
reduce the mRNA levels of type I collagen, thus decreasing
ECM production by MFs [113]. Analysis of the components of
EVs suggested that these effects were likely mediated by miR-21
and miR-29c, which target the signaling hubs for ECM produc-
tion [113]. In addition, MSCs also produce ECM-remodeling
enzymes, including MMP2, MMP9, and their inhibitors tissue
inhibitors of metalloproteinases 1 and 2 (TIMP-1 and TIMP-2), to
reduce ECM deposition in brotic sites [114]. However, there are
controversial results showing that the activities of MMP-2, MMP-9,
and MMP-13 were reduced in cardiac brosis after MSC treatment
[115]. This effect could be due to the context-specic regulation of
MMPs by MSCs. Janina Burk et al. cultured MSCs on different ECM
substrates and found that brotic-like ECM reduced MMP
production by MSCs [116].
Inammatory microenvironment regulation by MSCs
Accumulating evidence has shown that the immunomodulatory
potential of MSCs plays an important role in alleviating hepatic
brosis. MSCs demonstrated immunosuppressive potential and
effects on innate and adaptive immune responses through cell
contact or the secretion of immunoregulatory factors, including
heme oxygenase-1 (HO-1), NO, PGE2, IDO, IL-6, and HLA-G5
[117122]. Soluble factors are constitutively produced by MSCs or
released after MSC crosstalk with target cells. On the one hand,
these immunoregulatory factors can reduce liver injury caused by
inammation. On the other hand, they also provide a suitable
microenvironment for liver damage repair. MSCs can regulate
several kinds of immune cells, such as T and B lymphocytes,
natural killer (NK) cells, macrophages, Treg cells, neutrophils, and
myeloid-derived suppressor cells (MDSCs). Kazuya Sato et al.
demonstrated that NO produced by MSCs is one of the major
mediators of T-cell suppression by MSCs [120]. iNOS deciency in
MSCs abolishes the therapeutic effects of MSCs in advanced
hepatic brosis models [123]. In addition, it has been reported that
MSC-derived PGE2 and IDO mediate the immunosuppressive
potential of MSCs. On the one hand, PGE2 effectively mediates the
inhibition of NK cells by MSCs [124]. On the other hand, PGE2 and
IDO enhance Treg and Th17 cell differentiation and inhibit Th1 cell
differentiation [125,126]. It has also been reported that MSCs
induce the differentiation of Th17 cells toward Treg cells by
producing HGF, which generates an immunosuppressive environ-
ment [68].
Macrophages are critical inammation-related cells in the
hepatic inammatory microenvironment that play important roles
in regulating the hepatic inammatory microenvironment. Anoop
Babu Vasandan et al. found that MSC-derived PGE2 manipulates
metabolic programs in differentially polarized macrophages to re-
educate these cells [127]. MSCs can induce the differentiation of
macrophages to immunosuppressive phenotypes through direct
interactions [128]. MSC-derived IL-6 plays a crucial role in
repressing M1 polarization during inammation and promoting
M2b polarization under anti-inammatory conditions [121].
Augello et al. found that direct contact between MSCs and
target cells in a cognate fashion inhibited cell proliferation via the
engagement of the inhibitory molecule programmed death 1 with
its ligands PD-L1 and PD-L2, leading the target cells to modulate
the expression of different cytokine receptors and transduction
molecules associated with cytokine signaling [129]. MSCs suppress
the activation of CD4
+
T cells, downregulate IL-2 secretion, and
induce irreversible hyporesponsiveness and cell death via the
expression and secretion of PD-L1 and PD-L2 [130].
BENCH RESEARCH ON IMPROVING THE THERAPEUTIC EFFECT
OF MSCS ON HEPATIC FIBROSIS
Pretreatment of MSCs
The therapeutic effects of MSCs can be improved by changing the
cell culture conditions to further modulate the homing capacity,
survival, hepatogenic differentiation, and paracrine effects of
MSCs. Different attempts have been made to improve the efcacy
of MSCs by modifying the culture conditions, including changing
the culture media with additives, such as growth factors, chemical
agents, lipids, vitamins, and inammatory factors, and other
conditions, such as hypoxia. Pretreatment of MSCs with HGF and
FGF4 before transplantation improved MSC homing, enhanced
the transdifferentiation of MSCs into hepatocyte-like cells in the
recipient mouse liver and increased the therapeutic effects on
CCl
4
-induced brosis [131]. Melatoninpretreated MSCs showed a
high homing capacity to injured liver sites. These cells signicantly
improved the percentage of glycogen storage and decreased
collagen and lipid accumulation in brotic liver tissue [132]. Fathy
et al. showed that eugenol signicantly improved the self-renewal,
migration, and proliferation characteristics of AD-MSCs in vitro and
X. Yang et al.
5
Cellular & Molecular Immunology
greatly enhanced the therapeutic effects against hepatic brosis
induced by CCl
4
in rats [133]. Another recent study reported that
vitamin E pretreatment improved the homing of transplanted
Whartons jelly-derived allogenic MSCs, prevented oxidative stress,
and alleviated hepatic brosis [134]. L-theanine pretreatment
promoted anti-inammatory and antiapoptotic and HGF secretion
by AD-MSCs, which decreased the brosis level in the N-
nitrosodiethylamine-induced liver injury model [135]. TLR4- and
IFN-γ-activated MSCs reduced brosis-related actin alpha 2
(ACTA2), collagen type I alpha 1 chain (COL1A1), TGF-β, and TNF
levels in the liver. TLR4- and IFN-γ-activated MSCs could enhance
the Th1-polarized response, which alleviated liver granulomatous
and brosis in mice infected with Schistosoma japonicum [136].
Gene modication of MSCs
Gene modication of MSCs through viral or nonviral vectors has
been demonstrated to improve stemness, differentiation, immu-
noregulation, homing capacity, and other repair-related abilities
in vitro and in vivo. A series of genes have been overexpressed in
MSCs; among them, HGF was broadly used to modify MSCs and
improve the therapeutic effects on hepatic brosis, since HGF has
been shown to play a critical role in liver regeneration [137140].
Other genes, such as IGF1, hepatic nuclear factor (HNF) 4α, FGF4,
FGF21, IL-10, SERPINA1, PTP4A1, SMAD7, and ECM1, have been
used to modify MSCs and showed improved therapeutic effects in
the treatment of hepatic brosis (Table 1). HNF4α-overexpressing
MSCs inhibited inammation by enhancing iNOS expression
through the NF-κB signaling pathway in a CCl
4
-induced mouse
liver injury model, ultimately reducing inammation in the liver
and alleviating hepatic brosis [141]. Forkhead box A2 (FOXA2)-
overexpressing MSCs promoted the recovery of brotic liver tissue
by enhancing hepatogenic differentiation in MSCs and upregulat-
ing the expression of liver-specic genes, including FOXA2, alpha
fetoprotein, keratin 18 (CK-18), HNF1α, and HGF [142]. ECM1-
overexpressing hair follicle-derived MSCs could had signicantly
improved the homing and differentiation into hepatocytes, which
increased liver function and decreased pathological liver injury in
liver cirrhosis. In addition, HSC activation was signicantly
inhibited in the MSC treatment groups in vivo and in vitro [143].
In addition to the overexpression of target genes, modications of
microRNAs could also improve the effects of MSCs. MiR-122
modication enhanced the therapeutic efcacy of AD-MSCs in the
treatment of hepatic brosis by suppressing the activation of HSCs
and alleviating collagen deposition [144].
Furthermore, genetically modied MSCs can enhance the
immunomodulation of MSCs, thus increasing the therapeutic
effect of MSCs. IL-10 gene-edited amniotic MSCs inhibited the
activation of HSCs and TNF-αexpression in T cells/macrophages
derived from thioacetamide (TAA)-induced brotic livers [145].
Another study indicated that IL-35-overexpressing MSCs had
increased immunosuppressive capabilities, which caused CD4
+
T cells to produce IL-10 [146]. Therefore, modifying specic genes
in MSCs is a potential new strategy for improving their ability to
treat hepatic brosis.
MSC spheroids
There are signicant differences in cell phenotypes and biological
activities in three-dimensional (3D) and two-dimensional (2D) cell
cultures. MSC spheroid culture has been reported to increase the
therapeutic potential of MSCs [147]. Studies have demonstrated
that MSC spheroids have robustly enhanced therapeutic potential
by improvements in MSC stemness, multipotent differentiation,
anti-inammatory effects, angiogenesis, and tissue regeneration
through the secretion of high levels of cytokines. AD-MSC
spheroid formation promotes stemness, including self-renewal
and multidifferentiation capacities, and spheroid-derived AD-
MSCs show improved potential to attenuate liver failure [148]. In
an experimental model, UC-MSC-3D treatment exhibited
enhanced hepatogenic differentiation and improved the func-
tional recovery of the liver with marked decreases in ALT and AST
and a mild increase in albumin in a murine model of CCl
4
-induced
liver cirrhosis [149]. Another study demonstrated that AD-MSC
spheroids had enhanced stemness and anti-inammatory and
immunomodulatory functions, as revealed by the increased
expression of the stem cell markers OCT4, SOX2, and NANOG,
the anti-inammatory factors IL-10, TSG6, and IDO, and the
immunomodulatory molecules HGF, VEGF, and C-X-C motif
chemokine receptor 4 (CXCR4) [150]. It was also suggested that
3D-cultured MSC spheroids signicantly promoted wound healing
through well-organized collagen brils and high expression of the
angiogenesis biomarker CD31 [151]. In addition, stem cells from
human exfoliated deciduous teeth (SHED) are an ideal source of
MSCs. SHED-converted hepatocyte-like cell-based spheroids
improved liver dysfunction in association with antibrotic effects
in CCl
4
-treated mice [152]. Taken together, these ndings suggest
that MSC spheroids may replace MSCs as a novel therapeutic
strategy with excellent application prospects to treat hepatic
brosis.
MSC sheets
Okanos team developed a cell sheet engineering technology
using thermoresponsive culture dishes coated with poly(N-
isopropyl acrylamide) [153]. IC-2, which is a derivative of the
Wnt/β-catenin signaling inhibitor ICG-001, efciently induced
hepatic differentiation in human MSCs. A new therapy combining
IC-2 treatment and MSC sheet technology was developed to
establish a novel therapeutic strategy for liver diseases. Research
has reported that the transplantation of IC-2-engineered BM-MSC
sheets exerted potent regenerative effects and effectively
improved acute liver injury in mice [154]. IC-2 also promoted
MMP-14 secretion by BM-MSCs from elderly patients. Secreted
MMP-14 was a valuable predictive factor in reducing hepatic
brosis. In addition, MMP-13 activity and thioredoxin levels in IC-2
sheets were inversely correlated with hepatic hydroxyproline
levels [155]. Similar effects were also evident in a study by Itaba
et al., which indicated that orthotopic transplantation of IC-2-
engineered MSC sheets markedly reduced hepatic brosis
induced by chronic CCl
4
administration. Mechanistically, IC-2-
engineered MSC sheets produced high levels of MMP-1 and MMP-
14, as well as thioredoxin, which suppressed HSC activation [156].
These studies indicated that IC-2 sheets could be promising
therapeutic options for treating established hepatic brosis.
Cell-free therapy based on MSCs
MSCs secrete many molecules into the culture medium (CM), such
as cytokines/chemokines, free nucleic acids, EVs, and lipids,
effectively repairing tissue injury [157,158]. These MSC-derived
secretomes and EVs avoid the potential tumorigenicity, rejection
of cells, emboli formation, undesired differentiation, and infection
transmission of MSC-based therapies. MSC-CM inhibits HSC
activation by reducing the expression of probrotic genes such
as α-SMA, COL1A1, and MMP2 in vitro, reducing collagen
accumulation and inammation, and increasing hepatocyte
survival in a mouse hepatic brosis model [159,160]. In addition,
proteomic analysis of MSC-CM revealed a broad spectrum of
molecules involved in immunomodulation and liver regeneration
in Gal-N-induced liver-injured rats [161].
Notably, EVs are important paracrine factors secreted by MSCs.
EVs are membrane-bound vesicles that include apoptotic bodies
(504000 nm), microvesicles (1001000 nm), and EXs (40100 nm)
[162]. Accumulating evidence shows that EVs derived from MSCs
have therapeutic effects on hepatic brosis by reducing inam-
mation and hepatocyte apoptosis. Human BM-MSC-derived EVs
effectively targeted inammation and reduced brosis in a mouse
model, which was accompanied by decreased serum levels of ALP,
bile acid (BA), and ALT. EVs reduced liver accumulation of
X. Yang et al.
6
Cellular & Molecular Immunology
Table 1. The application of genetically modied MSCs in animal models of liver brosis/cirrhosis
Genetic
modication
MSCs sources Animal models Dosages Outcomes Mechanisms References
HGF Bone marrow,
umbilical cord
CCl
4
-induced liver brosis in
Rats, DMN-induced liver
brosis in Rats
2×10
6
~
1×10
7
Promote liver function
and attenuate liver
brosis
Upregulate HGF expression; promote
hepatocyte proliferation and inhibit
hepatocyte apoptosis; decrease collagen;
reduce MMP-9, MMP-13, MMP-14 and TIMP-
1 expression; decrease brogenic cytokines
PDGF-bb and TGF-β1
[247250]
IGF1 Bone marrow TAA-induced liver brosis
in mice
5×10
5
Reduce liver brosis Reduce HSC activation; enhance hepatocyte
proliferation; suppress oxidative stress;
decrease the expression of pro-
inammatory and pro-brogenic genes;
activate hepatic macrophages; induce the
expression of growth factors and MMP2 in
hepatic macrophages
[251], [252]
HNF4αBone marrow CCl
4
-induced liver brosis
in mice
1×10
6
Improve liver function
and inhibit liver
cirrhosis
Promote the expression of NF-κB signaling;
enhance anti-inammatory and immune
regulatory effects of MSCs
[253]
FOXA2 Bone marrow CCl
4
-induced liver brosis
in rats
/ Improve liver function
and inhibit liver
brosis
Promote hepatic differentiation of MSCs [254]
FGF4 Bone marrow CCl
4
-induced liver brosis
in rats
/ Improve liver function Promote the location of MSCs; enhance
proliferation of hepatocytes by increasing
the expression of PCNA, EpCAM, and
Jagged1
[255]
FGF21 Adipose tissue TAA-induced liver brosis
in mice
1.5 × 10
6
Improve liver function
and inhibit liver
brosis
Reduce serum HA, α-SMA, collagen, and
TIMP1; suppress p-JNK, NF-κB, and p-
SMAD2/3 signaling pathways
[256]
IL10 Amniotic tissue TAA-induced liver brosis
in mice
1×10
6
Improve liver function
and ameliorate liver
brosis
Promote secretion IL10; suppress activation
of HSCs and the inammation in
brotic liver
[257]
SERPINA1 Adipose tissue CCl
4
-induced liver brosis
in mice
2.5 × 10
5
Improve the survival
time of animals with
liver cirrhosis
Promote hepatic differentiation of MSCs [258]
PTP4A1 Placenta BDL-induced liver cirrhosis
in rats
2×10
6
Alleviate liver brosis Increase ATP production and mitochondrial
biogenesis; enhance the metabolism of
mitochondria via increased mtDNA and ATP
production
[259]
SMAD7 Bone marrow CCl
4
-induced liver brosis
in rats
35×10
6
Improve liver function
and ameliorate liver
brosis
Reduce collagen I and III, TGFβ1, TGFBR1, α-
SMA, TIMP1, laminin, and hyaluronic acid,
increase MMP1.
[260], [261]
ECM1 Hair follicle CCl
4
-induced liver brosis
in mice
1×10
6
Improve liver function Promote hepatic differentiation of MSCs;
inhibit HSC activation; inhibit TGF-β/SMAD
signaling pathway
[262]
X. Yang et al.
7
Cellular & Molecular Immunology
granulocytes and T cells and inhibited VCAM-1 expression [163].
Another study showed that EVs from amnion-derived MSCs
ameliorated inammation and brogenesis in a rat model of
NASH and hepatic brosis, potentially by attenuating HSC and KC
activation [164]. Similarly, human embryonic stem (ES) cell-derived
MSC-EVs exerted immunomodulatory and antiapoptotic effects on
TAA-induced chronic rat liver injury by upregulating anti-
inammatory cytokines (TGF-βand IL-10) and antiapoptotic genes
(BCL-2) and downregulating major contributors to brosis,
including COL1A1, α-SMA, and TIMP1 [165].
EXs from MSC-CM have advantages over corresponding MSCs;
they are smaller and less complex than cells, so they are easier to
produce and store and can potentially avoid some of the
regulatory issues associated with MSCs. Several studies have
focused on the benecial therapeutic effects of MSC-derived
exosomes in animal models of hepatic brosis. An in vivo study
showed that hBM-MSC-EXs effectively alleviated hepatic brosis,
which was characterized by a reduction in collagen accumulation,
enhanced liver functionality, inhibition of inammation, and
increased hepatocyte regeneration. hBM-MSC-EX treatment could
ameliorate CCl
4
-induced hepatic brosis by inhibiting HSC
activation through the Wnt/β-catenin pathway [166]. Tan et al.
found that MSC-derived exosomes contained beclin 1 (BECN1),
which promoted HSC ferroptosis by suppressing glutathione
peroxidase 4 (GPX4) expression. Knockdown of BECN1 in MSCs
inhibited ferroptosis and the antibrotic effects of MSC-derived
EXs in HSCs and brotic livers [167]. Several studies have indicated
that MSC-EXs contain miRNAs such as miR-125b, miR-150-5p, and
miR-486-5p, which suppress the activation of HSCs and reduce
brosis [168170]. In addition, EXs produced by MSCs could be
modied to improve their therapeutic effects on liver failure. MiR-
122-modied AD-MSCs expressed high levels of miR-122, which
could transfer miR-122 to HSCs and further affect the expression
levels of miR-122 target genes, such as insulin-like growth factor
receptor 1, cyclin G1, and prolyl-4-hydroxylase α1, which are
involved in HSC proliferation and collagen maturation [144]. Tang
et al. reported that EXs derived from clinical-grade broblast-like
MSCs were engineered to carry antisense oligonucleotide target-
ing STAT3 and could suppress STAT3 levels and ECM deposition in
mice with established hepatic brosis and signicantly improve
liver function [171]. These animal model-based studies suggested
that EVs may represent effective cell-free therapeutic agents for
treating liver diseases. MSC-derived EVs exert benecial therapeu-
tic effects. However, there are currently no clinical trials on MSC-
derived EVs in liver diseases. Many major obstacles, such as the
application strategies and therapeutic efcacies, as well as stable
and consistent isolation of EVs, remain to be resolved.
CLINICAL APPLICATIONS OF MSCS IN LIVER CIRRHOSIS
TREATMENT
Overview of MSCs in the treatment of liver cirrhosis in clinical
trials
Based on the well-known properties of MSCs, including ease of
isolation and expansion in vitro, self-renewal and multiple
differentiation capacity, injury tropism, immunomodulation, and
low immunogenicity, these cells are considered the most
promising candidates for cell therapy. MSCs have been used in
clinical trials to treat many diseases[172] related to inammation
and injury. Liver diseases are associated with immune responses
and tissue injury. Therefore, liver diseases are ideal targets for MSC
therapy. There are 68 clinical trials for diseases involving MSC
intervention registered at https://clinicaltrials.gov/ as of October
2022, and most these were focused on liver cirrhosis (n=47)
(Fig. 2A). Ten trials had been completed, and only six of them
posted results. Thus, limited data are available to evaluate the
therapeutic effects of MSCs. We also collected 14 published
clinical trial articles on MSC treatment of liver cirrhosis, which
overlapped with the 6 completed clinical trials that had results.
Therefore, we referred to these 14 published articles to evaluate
the outcomes of MSC treatment of liver cirrhosis.
Among the 14 articles, 12 revealed exciting effects of MSCs on
liver cirrhosis by evaluating liver function, liver volume, model for
end-stage liver disease (MELD) scores, and cytokine expression.
Most of these studies followed up with patients for more than
6 months and veried the safety of MSC transplantation. We
believe that a Phase II study about HBV-related decompensated
liver cirrhosis published in 2021 is the most convincing [173]
because it had the longest follow-up and largest sample size. In
this study, 219 patients were recruited and divided into a control
group (n=111) and a UC-MSC-treated group (n=108). UC-MSCs
were transplanted into patients monthly three times in total, and
the patients were followed up for 75 months. No obvious side
effects were observed, and there was a signicantly increased
overall survival rate and improved liver function in the UC-MSC-
treated group. However, not every study showed positive results.
Two clinical trials did not observe any therapeutic effects of MSCs
on liver function [174,175]. These studies recruited 27 patients
and 12 patients, respectively. This outcome is probably because
the sample size was not very large, and individual differences can
affect the results.
Among the clinical trials, two used hepatocytes derived from
MSCs to treat liver cirrhosis, which provided another possible MSC
application in liver cirrhosis. MSCs were induced to differentiate
into hepatocytes by using HGF, DEX, and oncostatin M (OSM) for
~28 days before being injected [176]. After a follow-up of
24 weeks, liver function was improved, as veried by decreases
in MELD score, prothrombin complex levels, and serum creatinine
and bilirubin and an increase in serum albumin without any
adverse effects. However, during the treatment, which was
~2 months, ten patients died because of the severity of their
illness. In another phase II trial, the researchers compared the
effects of undifferentiated MSCs and hepatocyte-differentiated
MSCs [177]. Follow-up of patients at 3 and 6 months post-infusion
revealed partial improvements in liver function tests with
increases in prothrombin concentrations and serum albumin
levels and decreases in the elevated bilirubin levels and MELD
scores in both groups. However, statistical comparisons between
the undifferentiated and differentiated MSC groups did not
indicate any signicant differences in clinical and laboratory
ndings. Animal experiments showed some contradictory conclu-
sions. In a TAA-induced liver cirrhosis model, MSC therapy could
Fig. 2 Clinical trials of MSCs in liver diseases
X. Yang et al.
8
Cellular & Molecular Immunology
regenerate cirrhotic liver tissue and improve liver function and
hepatic encephalopathy. Moreover, hepatogenic partially differ-
entiated MSCs were more efcient than undifferentiated MSCs.
The superiority of partially differentiated MSCs was most likely
due to the increased homing and differentiation abilities of these
cells [178]. Hanan El Baz et al. compared the therapeutic effect of
undifferentiated human cord blood MSCs, in vitro hepatogenically
differentiated MSCs, or freshly isolated rat hepatocytes in a CCl
4
-
induced liver cirrhosis model [179]. The undifferentiated MSC-
treated group had better outcomes than the in vitro prediffer-
entiated hepatocyte-like cell-treated group. Considering the
outcomes of clinical trials and the animal experiments mentioned
above, it is difcult to conclude whether predifferentiation is
better. However, we should keep in mind that undifferentiated
MSCs have more functions, including immunoregulation and
suppressing extracellular matrix accumulation through various
mechanisms, and these cells can also secrete HGF, which favors
hepatocyte differentiation. To determine a superior strategy for
the use of MSCs, more bench research and clinical trials are
needed. Furthermore, considering that time is precious to
patients, highly efcient MSC differentiation procedures should
be optimized if differentiated MSCs are used.
Parameters that need to be standardized for clinical
applications
In these clinical trials of liver cirrhosis, most (57.45%) are in Phase
II, and there is only one in Phase IV (Fig. 2B). Only ten clinical trials
have been completed (21.28%). Most are recruiting or not yet
recruiting (Fig. 2C). The slow progress in the clinical use of MSCs
may be due to some imprecise treatment standards. As shown in
Fig. 3, there are many clinical parameters used in clinics regarding
both cells and patients, including the sources, cell preparation
procedure, dosages, engraftment routes of MSCs, and condition of
patients, that must be standardized to promote the clinical
application of MSCs.
Sources. Although MSCs can be isolated from various tissues,
such as UC [180], BM [181], AD [182], UC blood [183], placenta [11],
amniotic uid [184], amniotic membrane [185], dental pulp [186],
synovium [187], peripheral blood, liver, lung [188], skeletal muscle
[189,190], and hair follicles [191], UC-MSCs and BM-MSCs have
been primarily used in clinical trials (Fig. 4A). This is probably
because these cells have been most thoroughly studied and are
relatively easy to obtain. Few studies have focused on the
therapeutic effects of MSCs derived from different sources in the
same study. BM-MSCs are mainly suitable for autologous
transplantation. However, there are some limitations to BM-MSC
transplantation. Invasive isolation causes injury and inammation
in donors, and the efciency is low compared with MSCs from
other sources [192]. The amount and therapeutic effect of BM-
MSCs are also affected by the condition of the donors. For
instance, the number and differentiation potential of BM-MSCs
decrease with age [193]. Compared with BM-MSCs, UC-MSCs have
many advantages. They are easy to obtain and easy to expand to
the required amount. UC-MSCs are at an early phase of organic
development and show increased self-renewal and differentiation
capacity [194]. UC-MSCs show higher hepatic differentiation
potential than BM-MSCs, suggesting that UC-MSCs may be
advantageous over BM-MSCs for treating end-stage liver disease
[195]. In addition, UC-MSCs have lower immunogenicity than bone
marrow MSCs [196,197]. AD-MSCs are also easy to obtain, but
these cells have poor proliferation and anti-inammatory abilities
[198]. Therefore, UC-MSCs seem to be the most suitable option for
transplantation.
Autologous or allogeneic transplantation is another problem
challenging doctors. From the clinical trial data, we can see that
Fig. 3 The parameters need to be standardized precisely in clinical application of MSCs in liver cirrhosis
Fig. 4 Clinical trials of liver cirrhosis based on MSCs application
X. Yang et al.
9
Cellular & Molecular Immunology
there is more autologous transplantation than allogeneic trans-
plantation (Fig. 4B). However, due to the advantages of UC-MSCs,
allogeneic transplantation is probably better than autologous
transplantation. Donor variations (age, sex, genetics, and health
status) are important factors that contribute to the clinical
outcomes of MSCs [58]. MSCs isolated from neonatal tissues
exhibit a longer active life and better proliferation and differentia-
tion potential than those derived from adult tissues. Furthermore,
MSCs derived from unhealthy donors may result in negative
clinical outcomes [199].
Cell preparation. Several studies have shown that several
parameters during cell product preparation affect the clinical
outcomes of MSC treatment, including the isolation methods,
culture conditions, cryopreservation and thawing, and product
heterogeneity [200,201]. From the beginning, various isolation
methods have led to robust differences in the potency of MSCs.
For example, techniques used to isolate MSCs from human UC and
placenta vary widely, and there are differences in whether the
specic sections of perivascular tissues are removed [201]. It was
also reported that in UC, MSCs from different parts of the cord
were different [202]. Regardless of the isolation method,
functional and molecular differences exist among MSCs produced
by different centers even when the starting material is the same
and MSCs are isolated from aliquots [203]. Therefore, developing a
worldwide standard to propagate MSCs is very important.
In general, MSCs should be amplied in vitro for at least several
weeks to obtain enough cells for cell therapy; therefore, the
culture conditions, including media, serum, growth factors,
scaffolds and culture systems, are important to maintain the
potency of MSCs [200,204]. Fetal bovine serum (FBS) is commonly
used in research labs to provide nutrients and favor cell
adherence. However, there are several severe limitations in the
use of FBS to culture clinical-grade MSCs. The risk of contamina-
tion and transmission of infectious agents [205], aberrant immune
reactions caused by residual animal proteins [206], antibodies
against FBS produced by MSCs [207], ethical concerns [208] and
batch-to-batch variability [209] make FBS inadequate for culturing
MSCs for clinical applications. Several culture media supplements
have been developed to replace FBS, including human AB serum,
human platelet lysate and chemically dened media (CDM).
However, these alternatives have their own limitations [201].
Serum-free, xenobiotic-free CDM is the preferred alternative to
FBS and has been shown to improve the differentiation and
expansion rates of MSCs [210212]. The oxygen content of the
culture conditions is another important parameter that should be
considered. Typically, 20% O
2
is used in normal culture conditions
in vitro. However, the O
2
concentration in the tissue environment
of native MSCs ranges between 1% and 7%. High oxygen levels
may compromise the therapeutic benets of MSCs [213]. Culturing
MSCs in hypoxic conditions enhances their proliferation, homing,
antiapoptotic, and proangiogenic capacities and minimizes
spontaneous differentiation [214216].
Cryogenic banking is needed to store MSCs before transplanta-
tion, and alterations in MSCs induced by cryopreservation and
subsequent thawing are another challenge regarding the clinical
use of MSCs. Cryopreservation has been demonstrated to stunt
immunosuppressive capabilities [217], postinfusion biodistribu-
tion, engraftment, and clearance kinetics [78,218]. Furthermore,
freezethaw steps and transient warming also affect MSC potency
[217]. Thawed MSCs exhibit diminished structural integrity upon
rewarming, resulting in the cells being recognized and deleted by
activated T cells and signicantly diminishing the lifetime of MSCs
in patients following infusion [78]. However, freshly thawed MSCs
can be fully recovered after 13 days, at least 24 h of postthaw
culture [219,220].
MSC products have robust heterogeneity not only because of
their natural subpopulation diversity but also because of the
donor, cell source, and variable parameters during preparation
[221223]. Heterogeneous cells showed different proliferation,
differentiation, and immunoregulatory capacities [224,225]. To
overcome the heterogeneity of MSCs and control the therapeutic
potency of MSC products, highly homogeneous MSCs were
obtained by using induced pluripotent stem cells (iPSCs) [226].
iPSCs have been shown to be safe and effective without any sign
of tumorigenesis in a clinical trial of GvHD (NCT02923375). On the
other hand, iPSC-MSCs can be used at a low passage number. It
has been demonstrated that long-term expansion of human UC-
MSCs in vitro (passage 11) reduces their functions [227]. Another
study showed that human AD-MSCs at passage 9 exhibited a
senescent phenotype and impaired therapeutic effects in a
murine hindlimb ischemia model [228]. Expanding hMSCs on soft
poly(ethylene glycol) hydrogel matrices [229] and three-
dimensional (3D) [230] culture systems is also benecial for
maintaining the early-passage MSC phenotype during expansion.
A bioactive hydrogel scaffold has also been proven to improve the
therapeutic effect of MSCs [231]. Fibroblast contamination is
probably another source of heterogeneity because these cells can
express most MSC markers and adhere to plastic [232]. However,
the increased expression of CD166 and decreased expression of
CD9 in MSCs could be used to purify these cells from broblasts
by ow-activated cell sorting (FACS) [201]. Taken together,
developing best practices for MSC preparation by considering all
of the crucial parameters is essential for the successful clinical
translation of MSCs.
Routes of administration. In clinical trials, various engraftment
routes have been used for MSC transplantation, primarily the
hepatic artery, followed by intravenous administration and the
portal vein (Fig. 4C). There have also been clinical trials
transplanting MSCs via the intrasplenic route [233]. It is hard to
say which route is the best. A study on acute liver failure
concluded that intraportal injection was better for repairing liver
injury than hepatic intra-arterial injection, peripheral intravenous
infusion, and in situ intrahepatic injection [234]. Unfortunately, no
similar study has been performed on liver cirrhosis. We cannot
draw a conclusion by comparing different studies.
Considering the convenience and side effects, intravenous
injection would be the most convenient and safest route.
However, ~60% of cells accumulate in the lung and are cleared
by the immune system, as indicated by animal experiments [235].
Thus, more cells should be prepared if the cells will be
transplanted intravenously. Intraportal, intrahepatic, and intras-
plenic injection can deliver MSCs faster and avoid off-target
effects. However, we should bear in mind the complications of
each injection route. These injection routes all require surgery and
cause trauma, and some patients will develop a fever [176]. The
condition of the patient should also be considered. Therefore, it is
difcult to determine which method of administration is optimal.
How to choose the injection route should depend on each
patients comprehensive condition.
Doses. Dose selection for MSC transplantation in current clinical
trials is highly irregular. MSC transplantation is performed according
to patient body weight in most clinical trials (0.54×10
6
/kg), while
other clinical trials administer MSCs according to the quantity of
cells (1 × 10
7
5×10
8
cells). Whether MSC therapy should follow
other medicine dosing strategies deserves consideration. Several
clinical trials have focused on the effect of different doses, but they
have not posted the data yet (NCT02705742, NCT03626090,
NCT05080465, NCT05227846, NCT05155657). However, based on
the published data, we can see that as few as 1 × 10
7
cells
administered by intrasplenic injection have apparent therapeutic
effects on liver cirrhosis for six months [233].
Most clinical trials transplant MSCs in one dose, while others
transplant MSCs in several doses, ranging from two to four doses
X. Yang et al.
10
Cellular & Molecular Immunology
with different intervals. One Phase II study compared the effect
of single and two-time transplantation with a monthly interval.
The results showed no signicant difference between single and
double transplantation [236]. Another study showed that one
dose of MSCs could improve liver function for up to two years of
follow-up. No signicant therapeutic effect was observed after 2
years [237]. It was reported that multiple infusions of MSCs may
enhance the effect of transplantation in animal experiments
[238,239]. Based on these studies, several injections with long
intervals are probably benecial for improving the therapeutic
effect of MSCs.
Patient condition. We noticed no trials grouping patients
according to their condition in the clinical trials and published
clinical articles. The properties of MSCs could be affected by their
microenvironment to a great extent, including migration, differ-
entiation, and immunoregulation. MSCs and the microenviron-
ment, especially the inammatory microenvironment, have
substantial crosstalk and interactions [58]. Therefore, the micro-
environment of patients is a crucial parameter that needs to be
considered in clinical trials. The cause of liver cirrhosis, the
inammation level, liver function and the treatment strategies
affect the microenvironment of the patient [240,241]. It should be
mentioned that patient age is another important parameter that
has been demonstrated to affect the therapeutic effect of
MSCs [242]. Based on animal experimental data, we can also see
that the age of the host adversely affects hMDSC-mediated bone
regeneration [243]. In most clinical trials, the enrolled patients are
~1830 years old to 6580 years old (clinicaltrials.gov). This age
span is too broad, resulting in signicant differences in the
microenvironment within a single group. The aging microenvir-
onment, including the senescence-associated secretory pheno-
type (SASP), has been proven to impair the function of
transplanted MSCs [244,245]. We can obtain much more accurate
results if patients are grouped by narrow age ranges. In one
previous study, we found that chronic stress inhibited the
therapeutic effects of MSCs in a mouse model [246]. Another
study showed that DEX abrogated the therapeutic effects of MSCs
on brin deposition, serum levels of bilirubin, albumin, and
aminotransferases, and T-lymphocyte inltration [123]. Combining
the comprehensive conditions of patients and the therapeutic
effects, we can evaluate whether a patient is suitable for MSC
treatment before MSC transplantation. We can also do something
to keep the patients in the best condition to receive MSC
transplantation.
CONCLUSION
In this review, we examined the paradoxical roles of MSCs in the
pathogenesis and treatment of hepatic brosis/cirrhosis. On the
one hand, MSCs are involved in the pathogenesis of hepatic
brosis through various mechanisms, such as differentiation into
MFs and the promotion of HSC activation. MSCs can also affect
hepatic brosis by regulating the inammatory microenviron-
ment. However, in animal experiments and clinical trials, MSCs
are used to treat hepatic brosis/cirrhosis. Increasing evidence
has shown that MSC transplantation can alleviate hepatic
brosis and improve liver function through several mechanisms,
including hepatocyte differentiation and protection, inhibiting
HSC activation, antibrotic factors, and immunoregulation. The
contradictory effects of MSCs on hepatic brosis mainly result
from whether these cells are endogenous or exogenous. Most of
the data suggesting the involvement of MSCs in the pathogen-
esis of hepatic brosis were obtained from endogenous MSCs.
However, the antibrotic effects of MSCs were mainly observed
after the transplantation of exogenous MSCs. Thus, the long-
term brotic microenvironment alters the properties and
functions of MSCs.
Based on studies on MSCs in hepatic brosis regression, several
new methods to enhance the therapeutic effects of MSCs have
been developed, including pretreatment, gene modication, cell-
free therapy, cell sheets, and spheroids. Further study is warranted
to determine whether these are new strategies for use in the
clinic. However, it is necessary to perform more bench research to
verify the validity and safety of these new strategies.
MSCs are the most promising candidates for cell therapy due to
their many advantages. Although some satisfactory outcomes in
some patients with cirrhosis have been observed in clinical trials,
various parameters need to be optimized from cells to recipients,
including the sources and preparation of MSCs and the routes and
doses for MSC transplantation. Patient condition, which is much
easier to ignore, is another important factor that affects the
therapeutic efcacy of MSCs. However, more future work needs to
be done to advance the development of MSC therapy.
REFERENCES
1. Asrani SK, Devarbhavi H, Eaton J, Kamath PS. Burden of liver diseases in the
world. J Hepatol. 2019;70:15171.
2. Llovet JM, Zucman-Rossi J, Pikarsky E, Sangro B, Schwartz M, Sherman M, et al.
Hepatocellular carcinoma. Nat Rev Dis Prim. 2016;2:16018.
3. Affo S, Yu LX, Schwabe RF. The role of cancer-associated broblasts and brosis
in liver cancer. Annu Rev Pathol. 2017;12:15386.
4. Bataller R, Brenner DA. Liver brosis. J Clin Investig. 2005;115:20918.
5. Mishra PJ, Banerjee D. Activation and differentiation of mesenchymal stem cells.
Methods Mol Biol. 2011;717:24553.
6. Quante M, Tu SP, Tomita H, Gonda T, Wang SS, Takashi S, et al. Bone marrow-
derived myobroblasts contribute to the mesenchymal stem cell niche and
promote tumor growth. Cancer Cell. 2011;19:25772.
7. Friedenstein AJ, Petrakova KV, Kurolesova AI, Frolova GP. Heterotopic of bone
marrow. Analysis of precursor cells for osteogenic and hematopoietic tissues.
Transplantation. 1968;6:23047.
8. Bianco P, Robey PG, Simmons PJ. Mesenchymal stem cells: revisiting history,
concepts, and assays. Cell Stem Cell. 2008;2:3139.
9. Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, Mosca JD, et al.
Multilineage potential of adult human mesenchymal stem cells. Science.
1999;284:1437.
10. Anjos-Afonso F, Bonnet D. Nonhematopoietic/endothelial SSEA-1+cells dene
the most primitive progenitors in the adult murine bone marrow mesenchymal
compartment. Blood. 2007;109:1298306.
11. In t Anker PS, Scherjon SA, Kleijburgvan der Keur C, de GrootSwings GMJS,
Claas FHJ, Fibbe WE, et al. Isolation of mesenchymal stem cells of fetal or
maternal origin from human placenta. Stem Cells. 2004;22:133845.
12. Zuk PA, Zhu M, Mizuno H, Huang J, Futrell JW, Katz AJ, et al. Multilineage cells
from human adipose tissue: implications for cell-based therapies. Tissue Eng.
2001;7:21128.
13. Barry FP, Murphy JM. Mesenchymal stem cells: clinical applications and biolo-
gical characterization. Int J Biochem Cell Biol. 2004;36:56884.
14. Barrachina L, Remacha AR, Romero A, Vázquez FJ, Albareda J, Prades M, et al.
Effect of inammatory environment on equine bone marrow derived
mesenchymal stem cells immunogenicity and immunomodulatory properties.
Vet Immunol Immunopathol. 2016;171:5765.
15. Nauta AJ, Westerhuis G, Kruisselbrink AB, Lurvink EG, Willemze R, Fibbe WE.
Donor-derived mesenchymal stem cells are immunogenic in an allogeneic host
and stimulate donor graft rejection in a nonmyeloablative setting. Blood.
2006;108:211420.
16. Mahmood A, Lu D, Lu M, Chopp M. Treatment of traumatic brain injury in adult
rats with intravenous administration of human bone marrow stromal cells.
Neurosurgery. 2003;53:697702.
17. Horwitz EM, Gordon PL, Koo WK, Marx JC, Neel MD, McNall RY, et al. Isolated
allogeneic bone marrow-derived mesenchymal cells engraft and stimulate
growth in children with osteogenesis imperfecta: Implications for cell therapy of
bone. Proc Natl Acad Sci USA. 2002;99:89327.
18. Uccelli A, Moretta L, Pistoia V. Mesenchymal stem cells in health and disease.
Nat Rev Immunol. 2008;8:72636.
19. Gebler A, Zabel O, Seliger B. The immunomodulatory capacity of mesenchymal
stem cells. Trends Mol Med. 2012;18:12834.
20. Gabbiani G, Ryan GB, Majne G. Presence of modied broblasts in granulation
tissue and their possible role in wound contraction. Experientia. 1971;27:54950.
21. Majno G, Gabbiani G, Hirschel BJ, Ryan GB, Statkov PR. Contraction of granu-
lation tissue in vitro: similarity to smooth muscle. Science. 1971;173:54850.
X. Yang et al.
11
Cellular & Molecular Immunology
22. Skalli O, Ropraz P, Trzeciak A, Benzonana G, Gillessen D, Gabbiani G, et al. A
monoclonal antibody against alpha-smooth muscle actin: a new probe for
smooth muscle differentiation. J Cell Biol. 1986;103:278796.
23. Darby I, Skalli O, Gabbiani G. Alpha-smooth muscle actin is transiently expressed
by myobroblasts during experimental wound healing. Lab Invest.
1990;63:219.
24. Serini G, Gabbiani G. Mechanisms of myobroblast activity and phenotypic
modulation. Exp Cell Res. 1999;250:27383.
25. Klingberg F, Hinz B, White ES. The myobroblast matrix: implications for tissue
repair and brosis. J Pathol. 2013;229:298309.
26. El Agha E, Kramann R, Schneider RK, Li X, Seeger W, Humphreys BD, et al.
Mesenchymal stem cells in brotic disease. Cell Stem Cell. 2017;21:16677.
27. Hinz B. The role of myobroblasts in wound healing. Curr Res Transl Med.
2016;64:1717.
28. Eyden B. The myobroblast: phenotypic characterization as a prerequisite to
understanding its functions in translational medicine. J Cell Mol Med.
2008;12:2237.
29. Kisseleva T, Brenner D. Molecular and cellular mechanisms of liver brosis and
its regression. Nat Rev Gastroenterol Hepatol. 2021;18:15166.
30. Kramann R, Schneider RK, DiRocco DP, Machado F, Fleig S, Bondzie PA, et al.
Perivascular Gli1+progenitors are key contributors to injury-induced organ
brosis. Cell Stem Cell. 2015;16:5166.
31. Fujita T, Narumiya S. Roles of hepatic stellate cells in liver inammation: a new
perspective. Inamm Regen. 2016;36:1.
32. Mederacke I, Hsu CC, Troeger JS, Huebener P, Mu X, Dapito DH, et al. Fate
tracing reveals hepatic stellate cells as dominant contributors to liver brosis
independent of its aetiology. Nat Commun. 2013;4:2823.
33. Kordes C, Sawitza I, Götze S, Häussinger D. Hepatic stellate cells support
hematopoiesis and are liver-resident mesenchymal stem cells. Cell Physiol
Biochem. 2013;31:290304.
34. de Araújo Farias V, Carrillo-Gálvez AB, Martín F, Anderson P. TGF-beta and
mesenchymal stromal cells in regenerative medicine, autoimmunity and cancer.
TGF-βand mesenchymal stromal cells in regenerative medicine, autoimmunity
and cancer. Cytokine Growth Factor Rev. 2018;43:2537.
35. Chen S, Xu L, 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;89:97581.
36. Qiao H, Zhou Y, Qin X, Cheng J, He Y, Jiang Y. NADPH oxidase signaling pathway
mediates mesenchymal stem cell-induced inhibition of hepatic stellate cell
activation. Stem Cells Int. 2018;2018:1239143.
37. Lee C, Kim M, Han J, Yoon M, Jung Y. Mesenchymal stem cells inuence acti-
vation of hepatic stellate cells, and constitute a promising therapy for liver
brosis. Biomedicines. 2021;9:1598.
38. Wang Y, Chen X, Cao W, Shi Y. Plasticity of mesenchymal stem cells in immu-
nomodulation: pathological and therapeutic implications. Nat Immunol.
2014;15:100916.
39. Di Nicola M, Carlo-Stella C, Magni M, Milanesi M, Longoni PD, Matteucci P, et al.
Human bone marrow stromal cells suppress T-lymphocyte proliferation induced
by cellular or nonspecic mitogenic stimuli. Blood, Human bone marrow stro-
mal cells suppress T-lymphocyte proliferation induced by cellular or nonspecic
mitogenic stimuli. Blood. 2002;99:383843.
40. Le Blanc K, Rasmusson I, Sundberg B, Götherström C, Hassan M, Uzunel M, et al.
Treatment of severe acute graft-versus-host disease with third party haploi-
dentical mesenchymal stem cells. Lancet, Treatment of severe acute graft-
versus-host disease with third party haploidentical mesenchymal stem cells.
Lancet. 2004;363:143941.
41. Galipeau J, Sensebe L. Mesenchymal stromal cells: clinical challenges and
therapeutic opportunities. Cell Stem Cell. 2018;22:82433.
42. Shi Y, Du L, Lin L, Wang Y. Tumour-associated mesenchymal stem/stromal cells:
emerging therapeutic targets. Nat Rev Drug Discov. 2017;16:3552.
43. Ren G, Zhang L, Zhao X, Xu G, Zhang Y, Roberts AI, et al. Mesenchymal stem cell-
mediated immunosuppression occurs via concerted action of chemokines and
nitric oxide. Cell Stem Cell. 2008;2:14150.
44. Pellicoro A, Ramachandran P, Iredale JP, Falloweld JA. Liver brosis and repair:
immune regulation of wound healing in a solid organ. Nat Rev Immunol.
2014;14:18194.
45. Tacke F, Zimmermann HW. Macrophage heterogeneity in liver injury and
brosis. J Hepatol. 2014;60:10906.
46. Shi C, Jia T, Mendez-Ferrer S, Hohl TM, Serbina NV, Lipuma L, et al. Bone marrow
mesenchymal stem and progenitor cells induce monocyte emigration in
response to circulating toll-like receptor ligands. Immunity. 2011;34:590601.
47. Yu PF, Huang Y, Han YY, Lin LY, Sun WH, Rabson AB, et al. TNFalpha-activated
mesenchymal stromal cells promote breast cancer metastasis by recruiting
CXCR2(+) neutrophils. Oncogene. 2017;36:48290.
48. Li W, Ren G, Huang Y, Su J, Han Y, Li J, et al. Mesenchymal stem cells: a double-
edged sword in regulating immune responses. Cell Death Differ.
2012;19:150513.
49. Liu K, Wang FS, Xu R. Neutrophils in liver diseases: pathogenesis and therapeutic
targets. Cell Mol Immunol. 2021;18:3844.
50. Das S, Maras JS, Hussain MS, Sharma S, David P, Sukriti S, et al. Hyperoxidized
albumin modulates neutrophils to induce oxidative stress and inammation in
severe alcoholic hepatitis. Hepatology. 2017;65:63146.
51. Pillay J, den Braber I, Vrisekoop N, Kwast LM, de Boer RJ, Borghans JA, et al. In
vivo labeling with 2H2O reveals a human neutrophil lifespan of 5.4 days. Blood.
2010;116:6257.
52. Raffaghello L, Bianchi G, Bertolotto M, Montecucco F, Busca A, Dallegri F, et al.
Human mesenchymal stem cells inhibit neutrophil apoptosis: a model for
neutrophil preservation in the bone marrow niche. Stem Cells. 2008;26:15162.
53. Cassatella MA, Mosna F, Micheletti A, Lisi V, Tamassia N, Cont C, et al. Toll-like
receptor-3-activated human mesenchymal stromal cells signicantly prolong
the survival and function of neutrophils. Stem Cells. 2011;29:100111.
54. Liu Q. Role of cytokines in the pathophysiology of acute-on-chronic liver failure.
Blood Purif. 2009;28:33141.
55. Arrenberg P, Maricic I, Kumar V. Sulfatide-mediated activation of type II natural
killer T cells prevents hepatic ischemic reperfusion injury in mice. Gastro-
enterology. 2011;140:64655.
56. Cui K, Yan G, Xu C, Chen Y, Wang J, Zhou R, et al. Invariant NKT cells promote
alcohol-induced steatohepatitis through interleukin-1beta in mice. J Hepatol.
2015;62:13118.
57. Selzner N, Selzner M, Odermatt B, Tian Y, Van Rooijen N, Clavien PA. ICAM-1
triggers liver regeneration through leukocyte recruitment and Kupffer cell-
dependent release of TNF-alpha/IL-6 in mice. Gastroenterology.
2003;124:692700.
58. Wang Y, Fang J, Liu B, Shao C, Shi Y. Reciprocal regulation of mesenchymal stem
cells and immune responses. Cell Stem Cell. 2022;29:151530.
59. Ren G, Su J, Zhang L, Zhao X, Ling W, L'huillie A, et al. Species variation in the
mechanisms of mesenchymal stem cell-mediated immunosuppression. Stem
Cells. 2009;27:195462.
60. Su J, Chen X, Huang Y, Li W, Li J, Cao K, et al. Phylogenetic distinction of iNOS
and IDO function in mesenchymal stem cell-mediated immunosuppression in
mammalian species. Cell Death Differ. 2014;21:38896.
61. Hegyi B, Kudlik G, Monostori E, Uher F. Activated T-cells and pro-inammatory
cytokines differentially regulate prostaglandin E2 secretion by mesenchymal
stem cells. Biochem Biophys Res Commun. 2012;419:21520.
62. Putra A, Ridwan FB, Putridewi AI, Kustiyah AR, Wirastuti K, Sadyah N, et al. The
Role of TNF-alpha induced MSCs on Suppressive Inammation by Increasing
TGF-beta and IL-10. Open Access Maced J Med Sci. 2018;6:177983.
63. Watanabe Y, Tsuchiya A, Seino S, Kawata Y, Kojima Y, Ikarashi S, et al.
Mesenchymal stem cells and induced bone marrow-derived macrophages
synergistically improve liver brosis in mice. Stem Cells Transl Med.
2019;8:27184.
64. David BA, Rezende RM, Antunes MM, Santos MM, Freitas Lopes MA, Diniz AB,
et al. Combination of mass cytometry and imaging analysis reveals origin,
location, and functional repopulation of liver myeloid cells in mice. Gastro-
enterology. 2016;151:117691.
65. Wang G, Cao K, Liu K, Xue Y, Roberts AI, Li F, et al. Kynurenic acid, an IDO
metabolite, controls TSG-6-mediated immunosuppression of human mesench-
ymal stem cells. Cell Death Differ. 2018;25:120923.
66. Ding Y, Xu D, Feng G, Bushell A, Muschel RJ, Wood KJ. Mesenchymal stem cells
prevent the rejection of fully allogenic islet grafts by the immunosuppressive
activity of matrix metalloproteinase-2 and -9. Diabetes. 2009;58:1797806.
67. Milosavljevic N, Gazdic M, Simovic Markovic B, Arsenijevic A, Nurkovic J, Doli-
canin Z, et al. Mesenchymal stem cells attenuate liver brosis by suppressing
Th17 cells - an experimental study. Transpl Int. 2018;31:10215.
68. Chen QH, Wu F, Liu L, Chen HB, Zheng RQ, Wang HL, et al. Mesenchymal stem
cells regulate the Th17/Treg cell balance partly through hepatocyte growth
factor in vitro. Stem Cell Res Ther. 2020;11:91.
69. Han X, Yang Q, Lin L, Xu C, Zheng C, Chen X, et al. Interleukin-17 enhances
immunosuppression by mesenchymal stem cells. Cell Death Differ.
2014;21:175868.
70. Du L, Lin L, Li Q, Liu K, Huang Y, Wang X, et al. IGF-2 preprograms maturing
macrophages to acquire oxidative phosphorylation-dependent anti-inamma-
tory properties. Cell Metab. 2019;29:136375.e8.
71. Wang X, Lin L, Lan B, Wang Y, Du L, Chen X, et al. IGF2R-initiated proton
rechanneling dictates an anti-inammatory property in macrophages. Sci Adv.
2020;6:eabb7389.
72. Hu C, Wu Z, Li L. Mesenchymal stromal cells promote liver regeneration through
regulation of immune cells. Int J Biol Sci. 2020;16:893903.
X. Yang et al.
12
Cellular & Molecular Immunology
73. Yang L, Pang Y, Moses HL. TGF-beta and immune cells: an important regulatory
axis in the tumor microenvironment and progression. Trends Immunol.
2010;31:2207.
74. Xu C, Yu P, Han X, Du L, Gan J, Wang Y, et al. TGF-beta promotes immune
responses in the presence of mesenchymal stem cells. J Immunol.
2014;192:1039.
75. Chen Z, Kuang Q, Lao XJ, Yang J, Huang W, Zhou D. Differentiation of UC-MSCs
into hepatocyte-like cells in partially hepatectomized model rats. Exp Ther Med.
2016;12:17759.
76. Wang J, Fu X, Yan Y, Li S, Duan Y, Marie Inglis B, et al. In vitro differentiation of
rhesus macaque bone marrow- and adipose tissue-derived MSCs into
hepatocyte-like cells. Exp Ther Med. 2020;20:25160.
77. Ji R, Zhang N, You N, Li Q, Liu W, Jiang N, et al. The differentiation of MSCs into
functional hepatocyte-like cells in a liver biomatrix scaffold and their trans-
plantation into liver-brotic mice. Biomaterials. 2012;33:89959008.
78. Chinnadurai R, Garcia MA, Sakurai Y, Lam WA, Kirk AD, Galipeau J, et al. Actin
cytoskeletal disruption following cryopreservation alters the biodistribution of
human mesenchymal stromal cells in vivo. Stem Cell Rep. 2014;3:6072.
79. Sato Y, Araki H, Kato J, Nakamura K, Kawano Y, Kobune M, et al. Human
mesenchymal stem cells xenografted directly to rat liver are differentiated into
human hepatocytes without fusion. Blood. 2005;106:75663.
80. Banas A, Teratani T, Yamamoto Y, Tokuhara M, Takeshita F, Quinn G, et al.
Adipose tissue-derived mesenchymal stem cells as a source of human hepa-
tocytes. Hepatology. 2007;46:21928.
81. Mohamadnejad M, Sohail MA, Watanabe A, Krause DS, Swenson ES, Mehal WZ.
Adenosine inhibits chemotaxis and induces hepatocyte-specic genes in bone
marrow mesenchymal stem cells. Hepatology. 2010;51:96373.
82. Health care in rural America: the crisis unfolds. Joint Task Force of the National
Association of Community Health Centers and the National Rural Health Asso-
ciation. J Public Health Policy. 1989;10:99116.
83. Schwartz RE, Reyes M, Koodie L, Jiang Y, Blackstad M, Lund T, et al. Multipotent
adult progenitor cells from bone marrow differentiate into functional
hepatocyte-like cells. J Clin Investig. 2002;109:1291302.
84. Hong SH, Gang EJ, Jeong JA, Ahn C, Hwang SH, Yang IH, et al. In vitro differ-
entiation of human umbilical cord blood-derived mesenchymal stem cells into
hepatocyte-like cells. Biochem Biophys Res Commun. 2005;330:115361.
85. Chivu M, Dima SO, Stancu CI, Dobrea C, Uscatescu V, Necula LG, et al. In vitro
hepatic differentiation of human bone marrow mesenchymal stem cells under
differential exposure to liver-specic factors. Transl Res. 2009;154:12232.
86. Kang XQ, Zang WJ, Song TS, Xu XL, Yu XJ, Li DL, et al. Rat bone marrow
mesenchymal stem cells differentiate into hepatocytes in vitro. World J Gas-
troenterol. 2005;11:347984.
87. Lange C, Bassler P, Lioznov MV, Bruns H, Kluth D, Zander AR, et al. Liver-specic
gene expression in mesenchymal stem cells is induced by liver cells. World J
Gastroenterol. 2005;11:4497504.
88. Ong SY, Dai H, Leong KW. Inducing hepatic differentiation of human
mesenchymal stem cells in pellet culture. Biomaterials. 2006;27:408797.
89. Borhani-Haghighi M, Talaei-Khozani T, Ayatollahi M, Vojdani Z. Whartons jelly-
derived mesenchymal stem cells can differentiate into hepatocyte-like cells by
HepG2 cell line extract. Iran J Med Sci. 2015;40:14351.
90. 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;2:1625.
91. Cao L, Zhang Y, Qian M, Wang X, Shuai Q, Gao C, et al. Construction of multicellular
aggregate by E-cadherin coated microparticles enhancing the hepatic specic
differentiation of mesenchymal stem cells. Acta Biomater. 2019;95:38294.
92. Wang B, Li W, Dean D, Mishra MK, Wekesa KS. Enhanced hepatogenic differ-
entiation of bone marrow derived mesenchymal stem cells on liver ECM
hydrogel. J Biomed Mater Res A. 2018;106:82938.
93. Zhou X, Cui L, Zhou X, Yang Q, Wang L, Guo G, et al. Induction of hepatocyte-like
cells from human umbilical cord-derived mesenchymal stem cells by dened
microRNAs. J Cell Mol Med. 2017;21:88193.
94. Schwabe RF, Luedde T. Apoptosis and necroptosis in the liver: a matter of life
and death. Nat Rev Gastroenterol Hepatol. 2018;15:73852.
95. Liang X, Ding Y, Zhang Y, Tse HF, Lian Q. Paracrine mechanisms of mesenchymal
stem cell-based therapy: current status and perspectives. Cell Transpl.
2014;23:104559.
96. Driscoll J, Patel T. The mesenchymal stem cell secretome as an acellular
regenerative therapy for liver disease. J Gastroenterol. 2019;54:76373.
97. van Poll D, Parekkadan B, Cho CH, Berthiaume F, Nahmias Y, Tilles AW, et al.
Mesenchymal stem cell-derived molecules directly modulate hepatocellular
death and regeneration in vitro and in vivo. Hepatology. 2008;47:163443.
98. Michalopoulos GK. Liver regeneration after partial hepatectomy: critical analysis
of mechanistic dilemmas. Am J Pathol. 2010;176:213.
99. Marsden ER, Hu Z, Fujio K, Nakatsukasa H, Thorgeirsson SS, Evarts RP. Expression
of acidic broblast growth factor in regenerating liver and during hepatic dif-
ferentiation. Lab Invest. 1992;67:42733.
100. Webber EM, Godowski PJ, Fausto N. In vivo response of hepatocytes to growth
factors requires an initial priming stimulus. Hepatology. 1994;19:48997.
101. Yu J, Yin S, Zhang W, Gao F, Liu Y, Chen Z, et al. Hypoxia preconditioned bone
marrow mesenchymal stem cells promote liver regeneration in a rat massive
hepatectomy model. Stem Cell Res Ther. 2013;4:83.
102. Adas G, Koc B, Adas M, Duruksu G, Subasi C, Kemik O, et al. Effects of
mesenchymal stem cells and VEGF on liver regeneration following major
resection. Langenbecks Arch Surg. 2016;401:72540.
103. Li WL, Su J, Yao YC, Tao XR, Yan YB, Yu HY, et al. Isolation and characterization of
bipotent liver progenitor cells from adult mouse. Stem Cells. 2006;24:32232.
104. Lin F, Chen W, Zhou J, Zhu J, Yao Q, Feng B, et al. Mesenchymal stem cells
protect against ferroptosis via exosome-mediated stabilization of SLC7A11 in
acute liver injury. Cell Death Dis. 2022;13:271.
105. Lin D, Chen H, Xiong J, Zhang J, Hu Z, Gao J, et al. Mesenchymal stem cells
exosomal let-7a-5p improve autophagic ux and alleviate liver injury in acute-
on-chronic liver failure by promoting nuclear expression of TFEB. Cell Death Dis.
2022;13:865.
106. Zheng J, Chen L, Lu T, Zhang Y, Sui X, Li Y, et al. MSCs ameliorate hepatocellular
apoptosis mediated by PINK1-dependent mitophagy in liver ischemia/reperfu-
sion injury through AMPKalpha activation. Cell Death Dis. 2020;11:256.
107. Yu F, Ji S, Su L, Wan L, Zhang S, Dai C, et al. Adipose-derived mesenchymal stem
cells inhibit activation of hepatic stellate cells in vitro and ameliorate rat liver
brosis in vivo. J Formos Med Assoc. 2015;114:1308.
108. Wang J, Bian C, Liao L, Zhu Y, Li J, Zeng L, et al. Inhibition of hepatic stellate cells
proliferation by mesenchymal stem cells and the possible mechanisms. Hepatol
Res. 2009;39:121928.
109. Zhang LT, Peng XB, Fang XQ, Li JF, Chen H, Mao XR. Human umbilical cord
mesenchymal stem cells inhibit proliferation of hepatic stellate cells in vitro. Int
J Mol Med. 2018;41:254552.
110. Ma L, Wei J, Zeng Y, Liu J, Xiao E, Kang Y, et al. Mesenchymal stem cell-
originated exosomal circDIDO1 suppresses hepatic stellate cell activation by
miR-141-3p/PTEN/AKT pathway in human liver brosis. Drug Deliv.
2022;29:44053.
111. Lin N, Hu K, Chen S, Xie S, Tang Z, Lin J, et al. Nerve growth factor-mediated
paracrine regulation of hepatic stellate cells by multipotent mesenchymal
stromal cells. Life Sci. 2009;85:2915.
112. Meier RP, Mahou R, Morel P, Meyer J, Montanari E, Muller YD, et al. Micro-
encapsulated human mesenchymal stem cells decrease liver brosis in mice. J
Hepatol. 2015;62:63441.
113. Basalova N, Sagaradze G, Arbatskiy M, Evtushenko E, Kulebyakin K, Grigorieva O,
et al. Secretome of mesenchymal stromal cells prevents myobroblasts differ-
entiation by transferring brosis-associated microRNAs within extracellular
vesicles. Cells. 2020;9:1272.
114. Lozito TP, Tuan RS. Mesenchymal stem cells inhibit both endogenous and
exogenous MMPs via secreted TIMPs. J Cell Physiol. 2011;226:38596.
115. Li L, Zhang Y, Li Y, Yu B, Xu Y, Zhao S, et al. Mesenchymal stem cell trans-
plantation attenuates cardiac brosis associated with isoproterenol-induced
global heart failure. Transpl Int. 2008;21:11819.
116. Burk J, Sassmann A, Kasper C, Nimptsch A, Schubert S. Extracellular matrix
synthesis and remodeling by mesenchymal stromal cells is context-sensitive. Int
J Mol Sci. 2022;23:1758.
117. Chabannes D, Hill M, Merieau E, Rossignol J, Brion R, Soulillou JP, et al. A role for
heme oxygenase-1 in the immunosuppressive effect of adult rat and human
mesenchymal stem cells. Blood. 2007;110:36914.
118. Jones BJ, Brooke G, Atkinson K, McTaggart SJ. Immunosuppression by placental
indoleamine 2,3-dioxygenase: a role for mesenchymal stem cells. Placenta.
2007;28:117481.
119. Selmani Z, Naji A, Zidi I, Favier B, Gaiffe E, Obert L, et al. Human leukocyte
antigen-G5 secretion by human mesenchymal stem cells is required to suppress
T lymphocyte and natural killer function and to induce CD4+CD25highFOXP3+
regulatory T cells. Stem Cells. 2008;26:21222.
120. Sato K, Ozaki K, Oh I, Meguro A, Hatanaka K, Nagai T, et al. Nitric oxide plays a
critical role in suppression of T-cell proliferation by mesenchymal stem cells.
Blood. 2007;109:22834.
121. Philipp D, Suhr L, Wahlers T, Choi YH, Paunel-Görgülü A. Preconditioning of
bone marrow-derived mesenchymal stem cells highly strengthens their
potential to promote IL-6-dependent M2b polarization. Stem Cell Res Ther.
2018;9:286.
122. Chen K, Wang D, Du WT, Han ZB, Ren H, Chi Y, et al. Human umbilical cord
mesenchymal stem cells hUC-MSCs exert immunosuppressive activities through
a PGE2-dependent mechanism. Clin Immunol. 2010;135:44858.
X. Yang et al.
13
Cellular & Molecular Immunology
123. Chen X, Gan Y, Li W, Su J, Zhang Y, Huang Y, et al. The interaction between
mesenchymal stem cells and steroids during inammation. Cell Death Dis.
2014;5:e1009.
124. Sotiropoulou PA, Perez SA, Gritzapis AD, Baxevanis CN, Papamichail M. Inter-
actions between human mesenchymal stem cells and natural killer cells. Stem
Cells. 2006;24:7485.
125. Yu Y, Yoo SM, Park HH, Baek SY, Kim YJ, Lee S, et al. Preconditioning with
interleukin-1 beta and interferon-gamma enhances the efcacy of human
umbilical cord blood-derived mesenchymal stem cells-based therapy via
enhancing prostaglandin E2 secretion and indoleamine 2,3-dioxygenase activity
in dextran sulfate sodium-induced colitis. J Tissue Eng Regen Med.
2019;13:1792804.
126. Rozenberg A, Rezk A, Boivin MN, Darlington PJ, Nyirenda M, Li R, et al. Human
mesenchymal stem cells impact Th17 and Th1 responses through a pros-
taglandin E2 and myeloid-dependent mechanism. Stem Cells Transl Med.
2016;5:150614.
127. Vasandan AB, Jahnavi S, Shashank C, Prasad P, Kumar A, Prasanna SJ. Human
mesenchymal stem cells program macrophage plasticity by altering their
metabolic status via a PGE2-dependent mechanism. Sci Rep. 2016;6:38308.
128. Özdemir R, Özdemir AT, SarıboyacıAE, Uysal O, Tuğlu Mİ,Kırmaz C. The
investigation of immunomodulatory effects of adipose tissue mesenchymal
stem cell educated macrophages on the CD4 T cells. Immunobiology.
2019;224:58594.
129. Augello A, Tasso R, Negrini SM, Amateis A, Indiveri F, Cancedda R, et al. Bone
marrow mesenchymal progenitor cells inhibit lymphocyte proliferation by
activation of the programmed death 1 pathway. Eur J Immunol.
2005;35:148290.
130. Davies LC, Heldring N, Kadri N, Le Blanc K. Mesenchymal stromal cell secretion
of programmed death-1 ligands regulates T cell mediated immunosuppression.
Stem Cells. 2017;35:76676.
131. Shams S, Mohsin S, Nasir GA, Khan M, Khan SN. Mesenchymal stem cells pre-
treated with HGF and FGF4 can reduce liver brosis in mice. Stem Cells Int.
2015;2015:747245.
132. Mortezaee K, Khanlarkhani N, Sabbaghziarani F, Nekoonam S, Majidpoor J,
Hosseini A, et al. Preconditioning with melatonin improves therapeutic out-
comes of bone marrow-derived mesenchymal stem cells in targeting liver
brosis induced by CCl4. Cell Tissue Res. 2017;369:30312.
133. Fathy M, Okabe M, M. Othman E, Saad Eldien HM, Yoshida T. Preconditioning of
adipose-derived mesenchymal stem-like cells with eugenol potentiates their
migration and proliferation in vitro and therapeutic abilities in rat hepatic
brosis. Molecules. 2020;25:2020.
134. Baig MT, Ghufran H, Mehmood A, Azam M, Humayun S, Riazuddin S. Vitamin E
pretreated Whartons jelly-derived mesenchymal stem cells attenuate CCl4-
induced hepatocyte injury in vitro and liver brosis in vivo. Biochem Pharm.
2021;186:114480.
135. Lai YJ, Sung YT, Lai YA, Chen LN, Chen TS, Chien CT. L-Theanine-treated adipose-
derived mesenchymal stem cells alleviate the cytotoxicity induced by
N-nitrosodiethylamine in liver. Tissue Eng Regen Med. 2022;19:120721.
136. Liu C, Zhang YS, Chen F, Wu XY, Zhang BB, Wu ZD, et al. Immunopathology in
schistosomiasis is regulated by TLR2,4- and IFN-gamma-activated MSC through
modulating Th1/Th2 responses. Stem Cell Res Ther. 2020;11:217.
137. Lai L, Chen J, Wei X, Huang M, Hu X, Yang R, et al. Transplantation of MSCs
overexpressing HGF into a rat model of liver brosis. Mol Imaging Biol.
2016;18:4351.
138. Seo KW, Sohn SY, Bhang DH, Nam MJ, Lee HW, Youn HY. Therapeutic effects of
hepatocyte growth factor-overexpressing human umbilical cord blood-derived
mesenchymal stem cells on liver brosis in rats. Cell Biol Int. 2014;38:10616.
139. Moon SH, Lee CM, Park SH, Jin Nam M. Effects of hepatocyte growth factor
gene-transfected mesenchymal stem cells on dimethylnitrosamine-induced
liver brosis in rats. Growth Factors. 2019;37:10519.
140. Kim MD, Kim SS, Cha HY, Jang SH, Chang DY, Kim W, et al. Therapeutic effect of
hepatocyte growth factor-secreting mesenchymal stem cells in a rat model of
liver brosis. Exp Mol Med. 2014;46:e110.
141. Ye Z, Lu W, Liang L, Tang M, Wang Y, Li Z, et al. Mesenchymal stem cells
overexpressing hepatocyte nuclear factor-4 alpha alleviate liver injury by
modulating anti-inammatory functions in mice. Stem Cell Res Ther.
2019;10:149.
142. Cho JW, Lee CY, Ko Y. Therapeutic potential of mesenchymal stem cells over-
expressing human forkhead box A2 gene in the regeneration of damaged liver
tissues. J Gastroenterol Hepatol. 2012;27:136270.
143. Liu Q, Lv C, Huang Q, Zhao L, Sun X, Ning D, et al. ECM1 modied HF-MSCs
targeting HSC attenuate liver cirrhosis by inhibiting the TGF-beta/Smad sig-
naling pathway. Cell Death Discov. 2022;8:51.
144. Lou G, Yang Y, Liu F, Ye B, Chen Z, Zheng M, et al. MiR-122 modication
enhances the therapeutic efcacy of adipose tissue-derived mesenchymal stem
cells against liver brosis. J Cell Mol Med. 2017;21:296373.
145. Choi JS, Jeong IS, Han JH, Cheon SH, Kim SW. IL-10-secreting human MSCs
generated by TALEN gene editing ameliorate liver brosis through enhanced
anti-brotic activity. Biomater Sci. 2019;7:107887.
146. Guo H, Zhao N, Gao H, He X. Mesenchymal stem cells overexpressing
interleukin-35 propagate immunosuppressive effects in mice. Scand J Immunol.
2017;86:38995.
147. Zhang X, Hu MG, Pan K, Li CH, Liu R. 3D spheroid culture enhances the
expression of antibrotic factors in human adipose-derived MSCs and improves
their therapeutic effects on hepatic brosis. Stem Cells Int. 2016;2016:4626073.
148. Zhang S, Liu P, Chen L, Wang Y, Wang Z, Zhang B. The effects of spheroid
formation of adipose-derived stem cells in a microgravity bioreactor on stem-
ness properties and therapeutic potential. Biomaterials. 2015;41:1525.
149. El Baz H, Demerdash Z, Kamel M, Hammam O, Abdelhady DS, Mahmoud S, et al.
Induction of hepatic regeneration in an experimental model using hepatocyte-
differentiated mesenchymal stem cells. Cell Reprogram. 2020;22:13446.
150. Lee S, Kim HS, Min BH, Kim BG, Kim SA, Nam H, et al. Enhancement of anti-
inammatory and immunomodulatory effects of adipose-derived human
mesenchymal stem cells by making uniform spheroid on the new nano-
patterned plates. Biochem Biophys Res Commun. 2021;552:1649.
151. Nilforoushzadeh MA, Khodadadi Yazdi M, Baradaran Ghavami S, Farokhimanesh
S, Mohammadi Amirabad L, Zarrintaj P, et al. Mesenchymal stem cell spheroids
embedded in an injectable thermosensitive hydrogel: an in situ drug formation
platform for accelerated wound healing. ACS Biomater Sci Eng.
2020;6:5096109.
152. Takahashi Y, Yuniartha R, Yamaza T, Sonoda S, Yamaza H, Kirino K, et al. Ther-
apeutic potential of spheroids of stem cells from human exfoliated deciduous
teeth for chronic liver brosis and hemophilia A. Pediatr Surg Int.
2019;35:137988.
153. Fukumori K, Akiyama Y, Kumashiro Y, Kobayashi J, Yamato M, Sakai K, et al.
Characterization of ultra-thin temperature-responsive polymer layer and its
polymer thickness dependency on cell attachment/detachment properties.
Macromol Biosci. 2010;10:111729.
154. Itaba N, Noda I, Oka H, Kono Y, Okinaka K, Yokobata T, et al. Hepatic cell sheets
engineered from human mesenchymal stem cells with a single small molecule
compound IC-2 ameliorate acute liver injury in mice. Regen Ther. 2018;9:4557.
155. Fukushima K, Itaba N, Kono Y, Okazaki S, Enokida S, Kuranobu N, et al. Secreted
matrix metalloproteinase-14 is a predictor for antibrotic effect of IC-2-
engineered mesenchymal stem cell sheets on liver brosis in mice. Regen
Ther. 2021;18:292301.
156. Itaba N, Kono Y, Watanabe K, Yokobata T, Oka H, Osaki M, et al. Reversal of
established liver brosis by IC-2-engineered mesenchymal stem cell sheets. Sci
Rep. 2019;9:6841.
157. Corrigendum to: Concise Review: MSC-Derived Exosomes for Cell-Free Therapy.
Stem Cells. 2017;35:2103.
158. Zhao T, Sun F, Liu J, Ding T, She J, Mao F, et al. Emerging role of mesenchymal
stem cell-derived exosomes in regenerative medicine. Curr Stem Cell Res Ther.
2019;14:48294.
159. Fu Q, Ohnishi S, Sakamoto N. Conditioned medium from human amnion-
derived mesenchymal stem cells regulates activation of primary hepatic stellate
cells. Stem Cells Int. 2018;2018:4898152.
160. Wang S, Lee JS, Hyun J, Kim J, Kim SU, Cha HJ, et al. Tumor necrosis factor-
inducible gene 6 promotes liver regeneration in mice with acute liver injury.
Stem Cell Res Ther. 2015;6:20.
161. Parekkadan B, van Poll D, Suganuma K, Carter EA, Berthiaume F, Tilles AW, et al.
Mesenchymal stem cell-derived molecules reverse fulminant hepatic failure.
PLoS One. 2007;2:e941.
162. Doyle LM, Wang MZ. Overview of extracellular vesicles, their origin, composi-
tion, purpose, and methods for exosome isolation and analysis. Cells.
2019;8:727.
163. Angioni R, Calì B, Vigneswara V, Crescenzi M, Merino A, Sánchez-Rodríguez R,
et al. Administration of human MSC-derived extracellular vesicles for the
treatment of primary sclerosing cholangitis: preclinical data in MDR2 knockout
mice. Int J Mol Sci. 2020;21:8874.
164. Ohara M, Ohnishi S, Hosono H, Yamamoto K, Yuyama K, Nakamura H, et al.
Extracellular vesicles from amnion-derived mesenchymal stem cells ameliorate
hepatic inammation and brosis in rats. Stem Cells Int. 2018;2018:3212643.
165. Mardpour S, Hassani SN, Mardpour S, Sayahpour F, Vosough M, Ai J, et al.
Extracellular vesicles derived from human embryonic stem cell-MSCs ameliorate
cirrhosis in thioacetamide-induced chronic liver injury. J Cell Physiol.
2018;233:933044.
X. Yang et al.
14
Cellular & Molecular Immunology
166. Rong X, Liu J, Yao X, Jiang T, Wang Y, Xie F. Human bone marrow mesenchymal
stem cells-derived exosomes alleviate liver brosis through the Wnt/beta-
catenin pathway. Stem Cell Res Ther. 2019;10:98.
167. Tan Y, Huang Y, Mei R, Mao F, Yang D, Liu J, et al. HucMSC-derived exosomes
delivered BECN1 induces ferroptosis of hepatic stellate cells via regulating the
xCT/GPX4 axis. Cell Death Dis. 2022;13:319.
168. Hyun J, Wang S, Kim J, Kim GJ, Jung Y. MicroRNA125b-mediated Hedgehog
signaling inuences liver regeneration by chorionic plate-derived mesenchymal
stem cells. Sci Rep. 2015;5:14135.
169. Du Z, Wu T, Liu L, Luo B, Wei C. Extracellular vesicles-derived miR-150-5p
secreted by adipose-derived mesenchymal stem cells inhibits CXCL1 expression
to attenuate hepatic brosis. J Cell Mol Med. 2021;25:70115.
170. Kim J, Lee C, Shin Y, Wang S, Han J, Kim M, et al. sEVs from tonsil-derived
mesenchymal stromal cells alleviate activation of hepatic stellate cells and liver
brosis through miR-486-5p. Mol Ther. 2021;29:147186.
171. Tang M, Chen Y, Li B, Sugimoto H, Yang S, Yang C, et al. Therapeutic targeting of
STAT3 with small interference RNAs and antisense oligonucleotides embedded
exosomes in liver brosis. FASEB J. 2021;35:e21557.
172. Squillaro T, Peluso G, Galderisi U. Clinical trials with mesenchymal stem cells: an
update. Cell Transpl. 2016;25:82948.
173. Shi M, Li YY, Xu RN, Meng FP, Yu SJ, Fu JL, et al. Mesenchymal stem cell therapy
in decompensated liver cirrhosis: a long-term follow-up analysis of the rando-
mized controlled clinical trial. Hepatol Int. 2021;15:143141.
174. Kantarcıoğlu M, Demirci H, Avcu F, Karslıoğlu Y, Babayiğit MA, Karaman B, et al.
Efcacy of autologous mesenchymal stem cell transplantation in patients with
liver cirrhosis. Turk J Gastroenterol. 2015;26:24450.
175. Mohamadnejad M, Alimoghaddam K, Bagheri M, AshraM, Abdollahzadeh L,
Akhlaghpoor S, et al. Randomized placebo-controlled trial of mesenchymal stem
cell transplantation in decompensated cirrhosis. Liver Int. 2013;33:14906.
176. Kharaziha P, Hellström PM, Noorinayer B, Farzaneh F, Aghajani K, Jafari F, 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;21:1199205.
177. El-Ansary M, Abdel-Aziz I, Mogawer S, Abdel-Hamid S, Hammam O, Teaema S,
et al. Phase II trial: undifferentiated versus differentiated autologous mesench-
ymal stem cells transplantation in Egyptian patients with HCV induced liver
cirrhosis. Stem Cell Rev Rep. 2012;8:97281.
178. Elberry DA, Amin SN, Esmail RS, Rashed LA, Gamal MM. Effect of undifferentiated
versus hepatogenic partially differentiated mesenchymal stem cells on hepatic
and cognitive functions in liver cirrhosis. EXCLI J. 2016;15:65270.
179. El Baz H, Demerdash Z, Kamel M, Atta S, Salah F, Hassan S, et al. Transplant of
hepatocytes, undifferentiated mesenchymal stem cells, and in vitro hepatocyte-
differentiated mesenchymal stem cells in a chronic liver failure experimental
model: a comparative study. Exp Clin Transpl. 2018;16:819.
180. Tsai PC, Fu TW, Chen YM, Ko TL, Chen TH, Shih YH, et al. The therapeutic
potential of human umbilical mesenchymal stem cells from Whartons jelly in
the treatment of rat liver brosis. Liver Transpl. 2009;15:48495.
181. Friedenstein AJ, Chailakhjan RK, Lalykina KS. The development of broblast
colonies in monolayer cultures of guinea-pig bone marrow and spleen cells. Cell
Tissue Kinet. 1970;3:393403.
182. Seki A, Sakai Y, Komura T, Nasti A, Yoshida K, Higashimoto M, et al. Adipose
tissue-derived stem cells as a regenerative therapy for a mouse steatohepatitis-
induced cirrhosis model. Hepatology. 2013;58:113342.
183. Erices A, Conget P, Minguell JJ. Mesenchymal progenitor cells in human
umbilical cord blood. Br J Haematol. 2000;109:23542.
184. In t Anker PS, Scherjon SA, Kleijburg-van der Keur C, Noort WA, Claas FHJ,
Willemze R, et al. Amniotic uid as a novel source of mesenchymal stem cells for
therapeutic transplantation. Blood. 2003;102:15489.
185. Yan K, Zhang R, Chen L, Chen F, Liu Y, Peng L, et al. Nitric oxide-mediated
immunosuppressive effect of human amniotic membrane-derived mesenchy-
mal stem cells on the viability and migration of microglia. Brain Res.
2014;1590:19.
186. Lei M, Li K, Li B, Gao LN, Chen FM, Jin Y. Mesenchymal stem cell characteristics of
dental pulp and periodontal ligament stem cells after in vivo transplantation.
Biomaterials. 2014;35:633243.
187. Ogata Y, Mabuchi Y, Yoshida M, Suto EG, Suzuki N, Muneta T, et al. Puried
human synovium mesenchymal stem cells as a good resource for cartilage
regeneration. PLoS One. 2015;10:e0129096.
188. in t Anker PS, Noort WA, Scherjon SA, Kleijburg-van der Keur C, Kruisselbrink AB,
van Bezooijen RL. Mesenchymal stem cells in human second-trimester bone
marrow, liver, lung, and spleen exhibit a similar immunophenotype but a het-
erogeneousmultilineage differentiationpotential. Haematologica. 2003;88:84552.
189. Gressner AM, Weiskirchen R, Breitkopf K, Dooley S. Roles of TGF-beta in hepatic
brosis. Front Biosci. 2002;7:d793807.
190. Williams JT, Southerland SS, Souza J, Calcutt AF, Cartledge RG. Cells isolated
from adult human skeletal muscle capable of differentiating into multiple
mesodermal phenotypes. Am Surg. 1999;65:226.
191. Ohyama M, Zheng Y, Paus R, Stenn KS. The mesenchymal component of hair
follicle neogenesis: background, methods and molecular characterization. Exp
Dermatol. 2010;19:8999.
192. Batsali AK, Kastrinaki MC, Papadaki HA, Pontikoglou C. Mesenchymal stem cells
derived from Whartons Jelly of the umbilical cord: biological properties and
emerging clinical applications. Curr Stem Cell Res Ther. 2013;8:14455.
193. D'ippolito G, Schiller PC, Ricordi C, Roos BA, Howard GA. Age-related osteogenic
potential of mesenchymal stromal stem cells from human vertebral bone
marrow. J Bone Min Res. 1999;14:111522.
194. Hsieh JY, Fu YS, Chang SJ, Tsuang YH, Wang HW. Functional module analysis
reveals differential osteogenic and stemness potentials in human mesenchymal
stem cells from bone marrow and Whartons jelly of umbilical cord. Stem Cells
Dev. 2010;19:1895910.
195. Yu YB, Song Y, Chen Y, Zhang F, Qi FZ. Differentiation of umbilical cord
mesenchymal stem cells into hepatocytes in comparison with bone marrow
mesenchymal stem cells. Mol Med Rep. 2018;18:200916.
196. Cho PS, Messina DJ, Hirsh EL, Chi N, Goldman SN, Lo DP, et al. Immunogenicity
of umbilical cord tissue derived cells. Blood. 2008;111:4308.
197. Deuse T, Stubbendorff M, Tang-Quan K, Phillips N, Kay MA, Eiermann T, et al.
Immunogenicity and immunomodulatory properties of umbilical cord lining
mesenchymal stem cells. Cell Transpl. 2011;20:65567.
198. 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;21:272452.
199. Donders R, Bogie J, Ravanidis S, Gervois P, Vanheusden M, Marée R, et al. Human
Whartons jelly-derived stem cells display a distinct immunomodulatory and
proregenerative transcriptional signature compared to bone marrow-derived
stem cells. Stem Cells Dev. 2018;27:6584.
200. Levy O, Kuai R, Siren E, Bhere D, Milton Y, Nissar N, et al. Shattering barriers
toward clinically meaningful MSC therapies. Sci Adv. 2020;6:eaba6884.
201. Yin JQ, Zhu J, Ankrum JA. Manufacturing of primed mesenchymal stromal cells
for therapy. Nat Biomed Eng. 2019;3:90104.
202. Davies JE, Walker JT, Keating A. Concise Review: Whartons Jelly: the rich, but
enigmatic, source of mesenchymal stromal cells. Stem Cells Transl Med.
2017;6:162030.
203. Stroncek DF, Jin P, McKenna DH, Takanashi M, Fontaine MJ, Pati S, et al. Human
mesenchymal stromal cell (MSC) characteristics vary among laboratories when
manufactured from the same source material: a report by the cellular therapy
team of the Biomedical Excellence for Safer Transfusion (BEST) collaborative.
Front Cell Dev Biol. 2020;8:458.
204. Qin L, Liu N, Bao CL, Yang DZ, Ma GX, Yi WH, et al., Mesenchymal stem cells in
brotic diseases-the two sides of the same coin. Acta Pharmacol Sin.
2023;44:26887.
205. Gregory CA, Reyes E, Whitney MJ, Spees JL. Enhanced engraftment of
mesenchymal stem cells in a cutaneous wound model by culture in allogenic
species-specic serum and administration in brin constructs. Stem Cells.
2006;24:223243.
206. Gstraunthaler G, Lindl T, van der Valk J. A plea to reduce or replace fetal bovine
serum in cell culture media. Cytotechnology. 2013;65:7913.
207. Sundin M, Ringdén O, Sundberg B, Nava S, Götherström C, Le Blanc K. No
alloantibodies against mesenchymal stromal cells, but presence of anti-fetal calf
serum antibodies, after transplantation in allogeneic hematopoietic stem cell
recipients. Haematologica. 2007;92:120815.
208. Tekkatte C, Gunasingh GP, Cherian KM, Sankaranarayanan K. Humanizedstem
cell culture techniques: the animal serum controversy. Stem Cells Int.
2011;2011:504723.
209. Witzeneder K, Lindenmair A, Gabriel C, Höller K, Theiß D, Redl H, et al. Human-
derived alternatives to fetal bovine serum in cell culture. Transfus Med
Hemother. 2013;40:41723.
210. Chase LG, Lakshmipathy U, Solchaga LA, Rao MS, Vemuri MC. A novel serum-
free medium for the expansion of human mesenchymal stem cells. Stem Cell
Res Ther. 2010;1:8.
211. Oikonomopoulos A, van Deen WK, Manansala AR, Lacey PN, Tomakili TA, Ziman
A, et al. Optimization of human mesenchymal stem cell manufacturing: the
effects of animal/xeno-free media. Sci Rep. 2015;5:16570.
212. Usta SN, Scharer CD, Xu J, Frey TK, Nash RJ. Chemically dened serum-free and
xeno-free media for multiple cell lineages. Ann Transl Med. 2014;2:97.
213. Fernández-Francos S, Eiro N, González-Galiano N, Vizoso FJ. Mesenchymal stem
cell-based therapy as an alternative to the treatment of acute respiratory dis-
tress syndrome: current evidence and future perspectives. Int J Mol Sci.
2021;22:15.
X. Yang et al.
15
Cellular & Molecular Immunology
214. Simon MC, Keith B. The role of oxygen availability in embryonic development
and stem cell function. Nat Rev Mol Cell Biol. 2008;9:28596.
215. Das B, Bayat-Mokhtari R, Tsui M, LotS, Tsuchida R, Felsher DW, et al. HIF-2alpha
suppresses p53 to enhance the stemness and regenerative potential of human
embryonic stem cells. Stem Cells. 2012;30:168595.
216. Rosová I, Dao M, Capoccia B, Link D, Nolta JA. Hypoxic preconditioning results in
increased motility and improved therapeutic potential of human mesenchymal
stem cells. Stem Cells. 2008;26:217382.
217. François M, Copland IB, Yuan S, Romieu-Mourez R, Waller EK, Galipeau J.
Cryopreserved mesenchymal stromal cells display impaired immunosuppressive
properties as a result of heat-shock response and impaired interferon-gamma
licensing. Cytotherapy. 2012;14:14752.
218. Ko IK, Kean TJ, Dennis JE. Targeting mesenchymal stem cells to activated
endothelial cells. Biomaterials. 2009;30:370210.
219. Moll G, Alm JJ, Davies LC, von Bahr L, Heldring N, Stenbeck-Funke L, et al. Do
cryopreserved mesenchymal stromal cells display impaired immunomodulatory
and therapeutic properties? Stem Cells. 2014;32:243042.
220. Panés J, García-Olmo D, Van Assche G, Colombel JF, Reinisch W, Baumgart DC,
et al. Expanded allogeneic adipose-derived mesenchymal stem cells (Cx601) for
complex perianal stulas in Crohns disease: a phase 3 randomised, double-
blind controlled trial. Lancet. 2016;388:128190.
221. Hass R, Kasper C, Böhm S, Jacobs R. Different populations and sources of human
mesenchymal stem cells (MSC): A comparison of adult and neonatal tissue-
derived MSC. Cell Commun Signal. 2011;9:12.
222. Shi Y, Wang Y, Li Q, Liu K, Hou J, Shao C, et al. Immunoregulatory mechanisms of
mesenchymal stem and stromal cells in inammatory diseases. Nat Rev Nephrol.
2018;14:493507.
223. de Wolf C, van de Bovenkamp M, Hoefnagel M. Regulatory perspective on
in vitro potency assays for human mesenchymal stromal cells used in immu-
notherapy. Cytotherapy. 2017;19:78497.
224. Chen JY, Mou XZ, Du XC, Xiang C. Comparative analysis of biological char-
acteristics of adult mesenchymal stem cells with different tissue origins. Asian
Pac J Trop Med. 2015;8:73946.
225. Costa LA, Eiro N, Fraile M, Gonzalez LO, Saá J, Garcia-Portabella P, et al. Func-
tional heterogeneity of mesenchymal stem cells from natural niches to culture
conditions: implications for further clinical uses. Cell Mol Life Sci.
2021;78:44767.
226. Ozay EI, Vijayaraghavan J, Gonzalez-Perez G, Shanthalingam S, Sherman HL, Gar-
rigan DT Jr, et al. Cymerus iPSC-MSCs signicantly prolong survival in a pre-clinical,
humanized mouse model of Graft-vs-host disease. Stem Cell Res. 2019;35:101401.
227. Gu Y, Li T, Ding Y, Sun L, Tu T, Zhu W, et al. Changes in mesenchymal stem cells
following long-term culture in vitro. Mol Med Rep. 2016;13:520715.
228. Lee JH, Yoon YM, Song KH, Noh H, Lee SH. Melatonin suppresses senescence-
derived mitochondrial dysfunction in mesenchymal stem cells via the HSPA1L-
mitophagy pathway. Aging Cell. 2020;19:e13111.
229. Rao VV, Vu MK, Ma H, Killaars AR, Anseth KS. Rescuing mesenchymal stem cell
regenerative properties on hydrogel substrates post serial expansion. Bioeng
Transl Med. 2019;4:5160.
230. Bunpetch V, Zhang ZY, Zhang X, Han S, Zongyou P, Wu H, et al. Strategies for
MSC expansion and MSC-based microtissue for bone regeneration. Biomaterials.
2019;196:6779.
231. Sivaraj D, Chen K, Chattopadhyay A, Henn D, Wu W, Noishiki C, et al. Hydrogel
scaffolds to deliver cell therapies for wound healing. Front Bioeng Biotechnol.
2021;9:660145.
232. Halfon S, Abramov N, Grinblat B, Ginis I. Markers distinguishing mesenchymal
stem cells from broblasts are downregulated with passaging. Stem Cells Dev.
2011;20:5366.
233. Amin MA, Sabry D, Rashed LA, Aref WM, el-Ghobary MA, Farhan MS, et al. Short-
term evaluation of autologous transplantation of bone marrow-derived
mesenchymal stem cells in patients with cirrhosis: Egyptian study. Clin
Transpl. 2013;27:60712.
234. Sang JF, Shi XL, Han B, Huang T, Huang X, Ren HZ, et al. Intraportal mesench-
ymal stem cell transplantation prevents acute liver failure through promoting
cell proliferation and inhibiting apoptosis. Hepatobiliary Pancreat Dis Int.
2016;15:60211.
235. Eggenhofer E, Benseler V, Kroemer A, Popp FC, Geissler EK, Schlitt HJ, et al.
Mesenchymal stem cells are short-lived and do not migrate beyond the lungs
after intravenous infusion. Front Immunol. 2012;3:297.
236. Suk KT, Yoon JH, Kim MY, Kim CW, Kim JK, Park H, et al. Transplantation with
autologous bone marrow-derived mesenchymal stem cells for alcoholic cir-
rhosis: phase 2 trial. Hepatology. 2016;64:218597.
237. Liang J, Zhang H, Zhao C, Wang D, Ma X, Zhao S, et al. Effects of allogeneic
mesenchymal stem cell transplantation in the treatment of liver cirrhosis caused
by autoimmune diseases. Int J Rheum Dis. 2017;20:121926.
238. Wei L, Zhang J, Xiao XB, Mai HX, Zheng K, Sun WL, et al. Multiple injections of
human umbilical cord-derived mesenchymal stromal cells through the tail vein
improve microcirculation and the microenvironment in a rat model of radiation
myelopathy. J Transl Med. 2014;12:246.
239. Zhang Y, Xia Y, Ni S, Gu Z, Liu H. Transplantation of umbilical cord mesenchymal
stem cells alleviates pneumonitis of MRL/lpr mice. J Thorac Dis. 2014;6:10917.
240. Boland L, Bitterlich LM, Hogan AE, Ankrum JA, English K. Translating MSC
therapy in the age of obesity. Front Immunol. 2022;13:943333.
241. Purtill D, Cirillo M, Fogarty J, Tan D, Cooney J, Wright M, et al. Early cessation of a
randomised study in acute graft versus host disease: upfront mesenchymal
stromal cells with corticosteroids versus corticosteroids alone. Bone Marrow
Transpl. 2020;55:2199201.
242. Chen H, Liu O, Chen S, Zhou Y. Aging and mesenchymal stem cells: therapeutic
opportunities and challenges in the older group. Gerontology. 2022;68:33952.
243. Gao X, Lu A, Tang Y, Schneppendahl J, Liebowitz AB, Scibetta AC, et al. Inu-
ences of donor and host age on human muscle-derived stem cell-mediated
bone regeneration. Stem Cell Res Ther. 2018;9:316.
244. Bickford PC, Kaneko Y, Grimmig B, Pappas C, Small B, Sanberg CD, et al.
Nutraceutical intervention reverses the negative effects of blood from aged rats
on stem cells. Age. 2015;37:103.
245. Rebo J, Mehdipour M, Gathwala R, Causey K, Liu Y, Conboy MJ, et al. A single
heterochronic blood exchange reveals rapid inhibition of multiple tissues by old
blood. Nat Commun. 2016;7:13363.
246. Yang X, Han ZP, Zhang SS, Zhu PX, Hao C, Fan TT, et al. Chronic restraint stress
decreases the repair potential from mesenchymal stem cells on liver injury by
inhibiting TGF-beta1 generation. Cell Death Dis. 2014;5:e1308.
247. Lai L, Chen J, Wei X, Huang M, Hu X, Yang R, et al. Transplantation of MSCs
Overexpressing HGF into a Rat Model of Liver Fibrosis. Mol Imaging Biol.
2016;18:4351.
248. Seo K-W, Sohn S-Y, Bhang D-H, Nam M-J, Lee H-W, Youn H-Y. Therapeutic effects
of hepatocyte growth factor-overexpressing human umbilical cord blood-
derived mesenchymal stem cells on liver brosis in rats. Cell Biol Int.
2014;38:10616.
249. Moon SH, Lee CM, Park S-H, Jin Nam M. Effects of hepatocyte growth factor
gene-transfected mesenchymal stem cells on dimethylnitrosamine-induced
liver brosis in rats. Growth Factors. 2019;37:10519.
250. Kim M-D, Kim S-S, Cha H-Y, Jang S-H, Chang D-Y, Kim W, et al. Therapeutic effect
of hepatocyte growth factor-secreting mesenchymal stem cells in a rat model of
liver brosis. Exp Mol Med. 2014;46:e110e110.
251. Fiore EJ, Bayo JM, Garcia MG, Malvicini M, Lloyd R, Piccioni F, et al. Mesenchymal
Stromal Cells Engineered to Produce IGF-I by Recombinant Adenovirus Ame-
liorate Liver Fibrosis in Mice. Stem Cells Dev. 2015;24:791801.
252. Fiore E, Malvicini M, Bayo J, Peixoto E, Atorrasagasti C, Sierra R, et al. Involve-
ment of hepatic macrophages in the antibrotic effect of IGF-I-overexpressing
mesenchymal stromal cells. Stem Cell Res Ther. 2016;7:172.
253. Ye Z, Lu W, Liang L, Tang M, Wang Y, Li Z, et al. Mesenchymal stem cells
overexpressing hepatocyte nuclear factor-4 alpha alleviate liver injury by
modulating anti-inammatory functions in mice. Stem Cell Research & Therapy.
2019;10:149.
254. Cho J-W, Lee C-Y, Ko Y. Therapeutic potential of mesenchymal stem cells
overexpressing human forkhead box A2 gene in the regeneration of damaged
liver tissues. J Gastroenterol Hepatol. 2012;27:136270.
255. Wang J, Xu L, Chan Q, Zhang Y, Hu Y, Yan L. Bone mesenchymal stem cells
overexpressing FGF4 contribute to liver regeneration in an animal model of liver
cirrhosis. Int J Clin Exp Med. 2015;8:1277482.
256. Kang H, Seo E, Park J-M, Han N-Y, Lee H, Jun H-S. Effects of FGF21-secreting
adipose-derived stem cells in thioacetamide-induced hepatic brosis. J Cell Mol
Med. 2018;22:51659.
257. Choi JS, Jeong IS, Han JH, Cheon SH, Kim S-W. IL-10-secreting human MSCs
generated by TALEN gene editing ameliorate liver brosis through enhanced
anti-brotic activity. Biomater Sci. 2019;7:107887.
258. Di Rocco G, Gentile A, Antonini A, Truffa S, Piaggio G, Capogrossi MC, et al.
Analysis of Biodistribution and Engraftment into the Liver of Genetically Mod-
ied Mesenchymal Stromal Cells Derived from Adipose Tissue. Cell Transplant.
2012;21:19972008.
259. Kim JY, Choi JH, Jun JH, Park S, Jung J, Bae SH, et al. Enhanced PRL-1 expression
in placenta-derived mesenchymal stem cells accelerates hepatic function via
mitochondrial dynamics in a cirrhotic rat model. Stem Cell Res Ther.
2020;11:512.
260. Wu S-P, Yang Z, Li F-R, Liu X-D, Chen H-T, Su D-N. Smad7-overexpressing rat
BMSCs inhibit the brosis of hepatic stellate cells by regulating the TGF-β1/
Smad signaling pathway. Exp Ther Med. 2017;14:256876.
261. Su D-N, Wu S-P, Xu S-Z. Mesenchymal stem cell-based Smad7 gene therapy for
experimental liver cirrhosis. Stem Cell Res Ther. 2020;11:395.
X. Yang et al.
16
Cellular & Molecular Immunology
262. Liu Q, Lv C, Huang Q, Zhao L, Sun X, Ning D, et al. ECM1 modied HF-MSCs
targeting HSC attenuate liver cirrhosis by inhibiting the TGF-β/Smad signaling
pathway. Cell Death Discov. 2022;8:51.
ACKNOWLEDGEMENTS
This project was supported by the National Key R&D Program of China (Grant No.
2018YFA0107500), the National Natural Science Foundation of China (Grant Nos.
82173276, 81972599, 81930085, 82073032, 82073037, and 81872243) and Grant No.
2022YFA0807300.
COMPETING INTERESTS
The authors declare no competing interests.
ADDITIONAL INFORMATION
Correspondence and requests for materials should be addressed to Yufang Shi or
Zhipeng Han.
Reprints and permission information is available at http://www.nature.com/
reprints
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 license, and indicate if changes were made. The images or other third party
material in this article are included in the articles Creative Commons license, unless
indicated otherwise in a credit line to the material. If material is not included in the
articles Creative Commons license 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 license, visit http://
creativecommons.org/licenses/by/4.0/.
© The Author(s), under exclusive licence to CSI and USTC 2023
X. Yang et al.
17
Cellular & Molecular Immunology
... NF-κB, a heterodimer comprising subunits p65 and p50, binds specifically to gene promoters or enhancer sequences, fostering transcription and expression [7]. The experiment have affirmed [8,9] that NFKB1 exhibits a specific affinity for distinct sites on gene promoters or enhancer sequences within various cells, consequently facilitating transcription and expression. The NFKB1 gene codes for subunits p105 and p50 within the NF-κB family. ...
... Ribavirin can delay CCL4-induced liver fibrosis by elevating the level of hepatic prostaglandin E2 and the anti-inflammatory cytokine IL-10. Recent animal studies have demonstrated that various TCM formulations, such as Biejia Xiaozheng Pills [8,9], can reverse liver fibrosis by inhibiting inflammatory factors and promoting antioxidation. TCM formulations have made Colchicine group (E-H). ...
Article
Full-text available
Si Ni San combined with Astragalus (SNSQ) has demonstrated significant efficacy in the treatment of hepatic fibrosis (HF), as confirmed by clinical practice. However, its pharmacological mechanism remains unclear. This study employs network pharmacology to identify key targets and proteins for molecular docking. Additionally, animal experiments were conducted to validate the network pharmacology results, providing further insights into the mechanism of SNSQ in treating HF. Effective compounds of SNSQ were screened from the Traditional Chinese Medicine Systems Pharmacology (TCMSP) and Encyclopedia of Traditional Chinese Medicine (ETCM) databases. Molecular formula structures of these effective compounds were obtained from the PubChem database. Partial target proteins with a probability greater than 0.6 were sourced from the SWISS database. Uniprot IDs corresponding to these target proteins were retrieved from the SUPERPRED database. The remaining target proteins of the compounds were obtained from the Uniprot database based on the Uniprot IDs. The drug target proteins were then summarized. Target points related to HF were selected from the GeneCards and OMIM databases. Common target points were identified in the Venn diagram and imported into Cytoscape 3.9.1 software to construct the “SNSQ-effective compound-target pathway-HF” network. AutoDock software was used for molecular docking of compounds and target proteins with high-degree values. The common target points underwent GO function enrichment and KEGG pathway enrichment analysis using the DAVID database. An HF rat model was established, and serum AST and ALT activities were measured. The Hyp assay kit was utilized to detect the Hyp content in liver tissue. To the transcription levels of pro-inflammatory factors (IL-1β, TNF-α, IL-6) and anti-inflammatory factors (IL-10, TGF-β1, IL-4) in rat serum and liver.IL-1β, TNF-α, IL-10, and TGF-β1 were chosen for validation through ELISA. Western blotting and qRT-PCR were used to assess the expression of related proteins, namely NFKB1, NF-κBp65, NF-κBp50, α-SMA, and Col-1 in liver tissue. qRT-PCR was also employed to study the expression of ECM synthesis and proliferation-related genes, including Cyclin D1, TIMP1, COL1A1 in HSC-T6 cells and rat liver tissue, as well as the inhibition of the ECM-related gene MMP13 in HSC-T6 cells and rat liver tissue. A total of 16 valid compounds were predicted, with kaempferol, sitosterol, and isorhamnetin exhibiting high-degree values. KEGG enrichment analysis revealed that the target genes of SNSQ were enriched in multiple pathological pathways, with the NF-Kappa B signaling pathway being predominant. Molecular docking simulations indicated strong affinities between SNSQ’s primary components—kaempferol, sitosterol, isorhamnetin—and NFKB1. Experimental results demonstrated significant reductions in AST, ALT, and Hyp levels in the SNSQ group. Pro-inflammatory factors (IL-1β, TNF-ɑ) were markedly reduced, while anti-inflammatory factors (IL-10, TGF-β1) were substantially increased. The protein expression and transcription levels of α-SMA and Col-1 were significantly decreased, whereas those of NFKB1, NF-κBp65, and NF-κBp50 were notably elevated. mRNA expression levels of Cyclin D1, TIMP1, COL1A1 in HSC-T6 cells and rat liver tissue were significantly decreased, whereas MMP13 mRNA expression level was significantly increased. Treatment of HF with SNSQ involves multiple targets and pathways, with a close association with the overexpression of NFKB1 and activation of the NF-Kappa B signaling pathway. Its mechanism is closely linked to the activation of inflammatory responses, HSC activation, and proliferation.
... The pathological process of liver fibrosis encompasses activation of HSCs, inflammation, oxidative stress, etc., (Yang et al., 2023). With its hepatoprotective and anti-fibrotic effects, curcumin exerts its mechanisms through antioxidative and anti-inflammatory actions, inhibition of hepatic stellate cell activation, and the blockade of receptors and signaling pathways (Farzaei et al., 2018). ...
Article
Full-text available
Objective This meta-analysis aimed to determine the efficacy of curcumin in preventing liver fibrosis in animal models. Methods A systematic search was conducted on studies published from establishment to November 2023 in PubMed, Web of Science, Embase, Cochrane Library, and other databases. The methodological quality was assessed using Sycle’s RoB tool. An analysis of sensitivity and subgroups were performed when high heterogeneity was observed. A funnel plot was used to assess publication bias. Results This meta-analysis included 24 studies involving 440 animals with methodological quality scores ranging from 4 to 6. The results demonstrated that curcumin treatment significantly improved Aspartate aminotransferase (AST) [standard mean difference (SMD) = -3.90, 95% confidence interval (CI) (−4.96, −2.83), p < 0.01, I² = 85.9%], Alanine aminotransferase (ALT)[SMD = − 4.40, 95% CI (−5.40, −3.40), p < 0.01, I² = 81.2%]. Sensitivity analysis of AST and ALT confirmed the stability and reliability of the results obtained. However, the funnel plot exhibited asymmetry. Subgroup analysis based on species and animal models revealed statistically significant differences among subgroups. Furthermore, curcumin therapy improved fibrosis degree, oxidative stress level, inflammation level, and liver synthesis function in animal models of liver fibrosis. Conclusion Curcumin intervention not only mitigates liver fibrosis but also enhances liver function, while concurrently modulating inflammatory responses and antioxidant capacity in animal models. This result provided a strong basis for further large-scale animal studies as well as clinical trials in humans in the future. Systematic Review Registration: https://www.crd.york.ac.uk/prospero/, identifier CRD42024502671.
... Liver fibrosis is a common pathological occurrence and is initiated as a result of chronic liver injury due to alcohol, viral hepatitis, drugs, toxins, nonalcoholic steatohepatitis, autoimmune liver disease, and so on. The pathogenesis of hepatic fibrosis is associated with the progressive accumulation of activated hepatic stellate cells (aHSCs), which can transdifferentiate into myofibroblasts to produce an excess of the extracellular matrix (ECM) [3]. In the normal liver, HSCs are quiescent and contain retinoid (vitamin A) and numerous lipid droplets. ...
Article
Full-text available
BACKGROUND Liver fibrosis is a compensatory response during the tissue repair process in chronic liver injury, and finally leads to liver cirrhosis or even hepatocellular carcinoma. The pathogenesis of hepatic fibrosis is associated with the progressive accumulation of activated hepatic stellate cells (HSCs), which can transdifferentiate into myofibroblasts to produce an excess of the extracellular matrix (ECM). Myofibroblasts are the main source of the excessive ECM responsible for hepatic fibrosis. Therefore, activated hepatic stellate cells (aHSCs), the principal ECM producing cells in the injured liver, are a promising therapeutic target for the treatment of hepatic fibrosis. AIM To explore the effect of taurine on aHSC proliferation and the mechanisms involved. METHODS Human HSCs (LX-2) were randomly divided into five groups: Normal control group, platelet-derived growth factor-BB (PDGF-BB) (20 ng/mL) treated group, and low, medium, and high dosage of taurine (10 mmol/L, 50 mmol/L, and 100 mmol/L, respectively) with PDGF-BB (20 ng/mL) treated group. Cell Counting Kit-8 method was performed to evaluate the effect of taurine on the viability of aHSCs. Enzyme-linked immunosorbent assay was used to estimate the effect of taurine on the levels of reactive oxygen species (ROS), malondialdehyde, glutathione, and iron concentration. Transmission electron microscopy was applied to observe the effect of taurine on the autophagosomes and ferroptosis features in aHSCs. Quantitative real-time polymerase chain reaction and Western blot analysis were performed to detect the effect of taurine on the expression of α-SMA, Collagen I, Fibronectin 1, LC3B, ATG5, Beclin 1, PTGS2, SLC7A11, and p62. RESULTS Taurine promoted the death of aHSCs and reduced the deposition of the ECM. Treatment with taurine could alleviate autophagy in HSCs to inhibit their activation, by decreasing autophagosome formation, downregulating LC3B and Beclin 1 protein expression, and upregulating p62 protein expression. Meanwhile, treatment with taurine triggered ferroptosis and ferritinophagy to eliminate aHSCs characterized by iron overload, lipid ROS accumulation, glutathione depletion, and lipid peroxidation. Furthermore, bioinformatics analysis demonstrated that taurine had a direct targeting effect on nuclear receptor coactivator 4, exhibiting the best average binding affinity of -20.99 kcal/mol. CONCLUSION Taurine exerts therapeutic effects on liver fibrosis via mechanisms that involve inhibition of autophagy and trigger of ferroptosis and ferritinophagy in HSCs to eliminate aHSCs.
... In TME, endogenous MSCs can promote hepatic fibrosis by regulating the inflammatory microenvironment. However, in clinical trials, exogenous MSCs were used to treat hepatic fibrosis (69,70). ...
Article
Full-text available
Hepatocellular carcinoma (HCC) is a prevalent malignant cancer worldwide, characterized by high morbidity and mortality rates. Alpha-fetoprotein (AFP) is a glycoprotein synthesized by the liver and yolk sac during fetal development. However, the serum levels of AFP exhibit a significant correlation with the onset and progression of HCC in adults. Extensive research has demonstrated that the tumor microenvironment (TME) plays a crucial role in the malignant transformation of HCC, and AFP is a key factor in the TME, promoting HCC development. The objective of this review was to analyze the existing knowledge regarding the role of AFP in the TME. Specifically, this review focused on the effect of AFP on various cells in the TME, tumor immune evasion, and clinical application of AFP in the diagnosis and treatment of HCC. These findings offer valuable insights into the clinical treatment of HCC.
... The safety and efficacy of MSCs have been shown in a range of clinical settings, including autoimmune diseases, organ failure, GvHD and COVID-19, while also being reported to ameliorate liver injury in the context of both acute and chronic liver diseases. [9][10][11] Tellingly, several clinical data have shown that MSCs can safely improve clinical outcomes of patients with alcohol-associated cirrhosis. [12] The pleiotropic effects of MSCs represent a potential advantage over pharmacological therapies and principally not only focus on their limited differentiation potentials but, more importantly, on their role in immunomodulation, resulting in a favorable immune microenvironment and releasing growth factors to activate endogenous tissue repair. ...
Article
Full-text available
Background Alcohol-associated liver disease (ALD) is a major health care challenge worldwide with limited therapeutic options. Although mesenchymal stem/stromal cells (MSCs) represent a newly emerging therapeutic approach to treat ALD, thus far, there have been extensive efforts to try and enhance their efficacy, including genetically engineering MSCs. FGF21, an endocrine stress-responsive hormone, has been shown to regulate energy balance, glucose, and lipid metabolism and to enhance the homing of MSCs toward injured sites. Therefore, the purpose of this study was to investigate whether MSCs that overexpress FGF21 (FGF21-MSCs) improve the therapeutic effect of MSCs in treating ALD. Methods Human umbilical cord-derived MSCs served as the gene delivery vehicle for the FGF21 gene. Human umbilical cord-derived MSCs were transduced with the FGF21 gene using lentiviral vectors to mediate FGF21 overexpression. We utilized both chronic Lieber-DeCarli and Gao-binge models of ethanol-induced liver injury to observe the therapeutic effect of FGF21-MSCs. Liver injury was phenotypically evaluated by performing biochemical methods, histology, and inflammatory cytokine levels. Results Compared with MSCs alone, administration of MSCs overexpressing FGF21(FGF21-MSCs) treatment significantly enhanced the therapeutic effect of ALD in mice, as indicated by the alleviation of liver injury with reduced steatosis, inflammatory infiltration, oxidative stress, and hepatic apoptosis, and the promotion of liver regeneration. Mechanistically, FGF21 could facilitate the immunomodulatory function of MSCs on macrophages by setting metabolic commitment for oxidative phosphorylation, which enables macrophages to exhibit anti-inflammatory inclination. Conclusions Our data elucidate that MSC modification by FGF21 could enhance their therapeutic effect in ALD and may help in the exploration of effective MSCs-based cell therapies for the treatment of ALD.
... However, some important parameters were not fully explored in this study. In a recently published review [35], Yang et al. placed the factors influencing the efficacy of stem cell therapy into two main categories, which were cells factors (the sources, cell preparation procedure, dosages, engraftment routes) and patients factors (liver function, inflammation, age, etc.). We could notice that there are many factors affecting the therapeutic effect. ...
Article
Full-text available
Background Stem cell transplantation shows great potential to improve the long-term survival of cirrhosis patients. However, therapeutic effects may not be homogeneous across the whole study population. This study constructed an easy-to-use nomogram to improve prognostic prediction and aid in treatment decision making for cirrhotic patients. Methods From August 2005 to April 2019, 315 patients with decompensated cirrhosis receiving autologous peripheral blood stem cell (PBSC) transplantation were enrolled in this study. They were randomly classified into training (2/3) and validation (1/3) groups. A predictive model was developed using Cox proportional hazard models and subsequently validated. The predictive performance of the model was evaluated and also compared with other prognostic models. Results Age, creatinine, neutrophil-to-lymphocyte ratio, and Child–Turcotte–Pugh class were included in the nomogram as prognostic variables. The nomogram showed high discrimination power concerning the area under receiver operating characteristic curves (3/5-year AUC: 0.742/0.698) and good consistency suggested by calibration plots. Patients could be accurately stratified into poor- and good-outcome groups regarding liver-transplantation free survival after receiving PBSC therapy (P < 0.001). Compared with poor-outcome group, the liver function of patients listed for liver transplantation in the good-outcome group was significantly improved (P < 0.001). Besides, our nomogram achieved a higher C-index (0.685, 95% CI 0.633–0.738) and better clinical utility compared with other conventional prognostic models. Conclusions The proposed nomogram facilitated an accurate prognostic prediction for patients with decompensated cirrhosis receiving PBSC transplantation. Moreover, it also held the promise to stratify patients in clinical trials or practice to implement optimal treatment regimens for individuals.
Article
Aims Neovascularization plays a crucial role in liver fibrosis (LF), and blocking vascular endothelial growth factor receptors (VEGFR) has been shown to improve fibrosis. The aim of our study was to investigate the role of dual neovascularization targets, VEGFR, and platelet‐derived growth factor receptor (PDGFR), in ameliorating fibrosis. Methods In vitro, we observed the effects of apatinib (APA) (a VEGFR inhibitor) and donafenib (DON) (a VEGFR and PDGFR inhibitor) on the activation, proliferation, and apoptosis of hepatic stellate cells (HSCs) from rats and humans. In vivo, we established a thioacetamide (TAA)‐induced liver fibrosis rat model to explore the antifibrosis effect of APA and DON. We used the method of random table to randomly divide the rats into 4 groups. We detected the expression of angiogenesis‐related proteins using Western blot and immunohistochemistry. Results APA and DON inhibited the proliferation and activation of HSCs, promoted apoptosis of HSCs, and arrested the S phase of the cell cycle in vitro. We also found that DON had a stronger inhibitory effect on HSCs. In vivo, APA and DON ameliorated liver fibrosis, reduced collagen deposition and α‐SMA expression in rats, and DON had a stronger improvement effect. APA and DON downregulated the expression of VEGFR2 while inhibiting the phosphorylation of Akt and ERK1/2. DON can act through both VEGF and PDGF pathways, whereas APA can only act through the VEGF pathway. Conclusion Antiangiogenesis is a promising approach for the treatment of fibrosis. Compared with a single‐target drug (APA), the dual‐target drug (DON) can achieve better therapeutic effects.
Article
Full-text available
Liver fibrosis results from chronic liver injury and is characterized by excessive deposition of extracellular matrix proteins including collagen. It can progress to cirrhosis and liver failure. Multiple cellular signaling pathways drive hepatic stellate cell activation and fibrogenesis. Advances in biomarkers, imaging modalities, and omics platforms enable noninvasive diagnosis and staging of liver fibrosis. Emerging antifibrotic approaches include medications like pirfenidone, obeticholic acid, and monoclonal antibodies targeting pro-fibrotic mediators. Cell therapies using mesenchymal stem cells demonstrate antifibrotic potential through paracrine immunosuppression. Tissue-engineered liver grafts and biomaterial carriers for localized drug delivery are promising technologies. Microfluidic liver-on-a-chip platforms with patient-derived cells provide unprecedented models to study human liver fibrosis and test drug candidates. Significant progress has elucidated mechanisms underlying liver fibrogenesis and uncovered novel therapeutic targets. Ongoing challenges include translating preclinical findings, improving antifibrotic efficacy, and enabling personalized precision medicine approaches. Further research into combinatorial therapies, biomarkers, and tissue engineering technologies will advance the treatment of liver fibrosis from all causes.
Article
Full-text available
Inflammatory cytokine storms can trigger disease exacerbation and even death and have reached a consensus in the clinical treatment of acute organ failure. However, the existing strategies remain a great challenge to efficiently suppress inflammatory cytokine storms for promoting organ repair and regeneration. Herein, fully human umbilical cord (UC)-derived adhesive materials (UCAM) that integrate decellularized extracellular matrix (ECM) nanofiber hydrogel and homologous mesenchymal stem cells (MSCs) are demonstrated to greatly suppress inflammatory cytokine storms, demonstrating high efficacy in treating acute liver failure (ALF) in rats with 90% hepatectomy. The UC-derived adhesive materials have the capacity to secrete a significant quantity of cytokines by MSCs to recruit activated immune cells to migrate into their ECM nanofiber networks, segregating them away from the infection area and thereby greatly suppressing the inflammatory cytokine storms. As expected, the UC-derived adhesive materials can significantly promote hepatocyte proliferation to achieve functional recovery and regeneration of the liver, significantly improving the survival rate in rats. Our fully human UC-derived adhesive materials provide a new avenue in suppressing inflammatory cytokine storms for promoting organ regeneration that would be really utility in clinical organ transplantation-related treatment.
Article
Full-text available
Acute-on-chronic liver failure is a distinct clinical syndrome characterized by a dysregulated immune response and extensive hepatocyte death without satisfactory therapies. As a cytoplasmic degradative and quality-control process, autophagy was implicated in maintaining intracellular homeostasis, and decreased hepatic autophagy was found in many liver diseases and contributes to disease pathogenesis. Previously, we identified the therapeutic potential of mesenchymal stem cells (MSCs) in ACLF patients; however, the intrinsic mechanisms are incompletely understood. Herein, we showed that MSCs restored the impaired autophagic flux and alleviated liver injuries in ACLF mice, but these effects were abolished when autophago-lysosomal maturation was inhibited by leupeptin (leu), suggesting that MSCs exerted their hepatoprotective function in a pro-autophagic dependent manner. Moreover, we described a connection between transcription factor EB (TFEB) and autophagic activity in this context, as evidenced by increased nuclei translocation of TFEB elicited by MSCs were capable of promoting liver autophagy. Mechanistically, we confirmed that let-7a-5p enriched in MSCs derived exosomes (MSC-Exo) could activate autophagy by targeting MAP4K3 to reduce TFEB phosphorylation, and MAP4K3 knockdown partially attenuates the effect of anti-let-7a-5p oligonucleotide via decreasing the inflammatory response, in addition, inducing autophagy. Altogether, these findings revealed that the hepatoprotective effect of MSCs may partially profit from its exosomal let-7a-5p mediating autophagy repairment, which may provide new insights for the therapeutic target of ACLF treatment.
Article
Full-text available
Mesenchymal stromal cell (MSC) therapy has seen increased attention as a possible option to treat a number of inflammatory conditions including COVID-19 acute respiratory distress syndrome (ARDS). As rates of obesity and metabolic disease continue to rise worldwide, increasing proportions of patients treated with MSC therapy will be living with obesity. The obese environment poses critical challenges for immunomodulatory therapies that should be accounted for during development and testing of MSCs. In this review, we look to cancer immunotherapy as a model for the challenges MSCs may face in obese environments. We then outline current evidence that obesity alters MSC immunomodulatory function, drastically modifies the host immune system, and therefore reshapes interactions between MSCs and immune cells. Finally, we argue that obese environments may alter essential features of allogeneic MSCs and offer potential strategies for licensing of MSCs to enhance their efficacy in the obese microenvironment. Our aim is to combine insights from basic research in MSC biology and clinical trials to inform new strategies to ensure MSC therapy is effective for a broad range of patients.
Article
Full-text available
Activated hepatic stellate cells (HSCs) are significant in liver fibrosis. Our past investigations have shown that human umbilical cord mesenchymal stem cells (hucMSCs) and their secreted exosomes (MSC-ex) could alleviate liver fibrosis via restraining HSCs activation. However, the mechanisms underlying the efficacy were not clear. Ferroptosis is a regulatory cell death caused by excessive lipid peroxidation, and it plays a vital role in the occurrence and development of liver fibrosis. In the present study, we aimed to study the proferroptosis effect and mechanism of MSC-ex in HSCs. MSC-ex were collected and purified from human umbilical cord MSCs. Proferroptosis effect of MSC-ex was examined in HSCs line LX-2 and CCl4 induced liver fibrosis in mice. Gene knockdown or overexpression approaches were used to investigate the biofactors in MSC-ex-mediated ferroptosis regulation. Results: MSC-ex could trigger HSCs ferroptosis by promoting ferroptosis-like cell death, ROS formation, mitochondrial dysfunction, Fe ² ⁺ release, and lipid peroxidation in human HSCs line LX-2. Glutathione peroxidase 4 (GPX4) is a crucial regulator of ferroptosis. We found that intravenous injection of MSC-ex significantly decreased glutathione peroxidase 4 (GPX4) expression in activated HSCs and collagen deposition in experimental mouse fibrotic livers. Mechanistically, MSC-ex derived BECN1 promoted HSCs ferroptosis by suppressing xCT-driven GPX4 expression. In addition, ferritinophagy and necroptosis might also play a role in MSC-ex-promoted LX-2 cell death. Knockdown of BECN1 in MSC diminished proferroptosis and anti-fibrosis effects of MSC-ex in LX-2 and fibrotic livers. MSC-ex may promote xCT/GPX4 mediated HSCs ferroptosis through the delivery of BECN1 and highlights BECN1 as a potential biofactor for alleviating liver fibrosis.
Article
Full-text available
Mesenchymal stem cells (MSCs) have attracted interest for their potential to alleviate liver injury. Here, the protective effect of MSCs on carbon tetrachloride (CCl 4 )-induced acute liver injury (ALI) was investigated. In this study, we illustrated a novel mechanism that ferroptosis, a newly recognized form of regulated cell death, contributed to CCl 4 -induced ALI. Subsequently, based on the in vitro and in vivo evidence that MSCs and MSC-derived exosomes (MSC-Exo) treatment achieved pathological remission and inhibited the production of lipid peroxidation, we proposed an MSC-based therapy for CCl 4 -induced ALI. More intriguingly, treatment with MSCs and MSC-Exo downregulated the mRNA level of prostaglandin-endoperoxide synthase 2 ( Ptgs2 ) and lipoxygenases ( LOXs ) while it restored the protein level of SLC7A11 in primary hepatocytes and mouse liver, indicating that the inhibition of ferroptosis partly accounted for the protective effect of MSCs and MSC-Exo on ALI. We further revealed that MSC-Exo-induced expression of SLC7A11 protein was accompanied by increasing of CD44 and OTUB1. The aberrant expression of ubiquitinated SLC7A11 triggered by CCl 4 could be rescued with OTUB1-mediated deubiquitination, thus strengthening SLC7A11 stability and thereby leading to the activation of system X C ⁻ to prevent CCl 4 -induced hepatocyte ferroptosis. In conclusion, we showed that MSC-Exo had a protective role against ferroptosis by maintaining SLC7A11 function, thus proposing a novel therapeutic strategy for ferroptosis-induced ALI.
Article
Full-text available
Hair follicle-derived mesenchymal stem cells (HF-MSCs) show considerable therapeutic potential for liver cirrhosis (LC). To improve the effectiveness of naïve HF-MSC treatments on LC, we used bioinformatic tools to identify an exogenous gene targeting HSCs among the differentially expressed genes (DEGs) in LC to modify HF-MSCs. Extracellular matrix protein 1 (ECM1) was identified as a DEG that was significantly downregulated in the cirrhotic liver. Then, ECM1-overexpressing HF-MSCs (ECM1-HF-MSCs) were transplanted into mice with LC to explore the effectiveness and correlated mechanism of gene-overexpressing HF-MSCs on LC. The results showed that ECM1-HF-MSCs significantly improved liver function and liver pathological injury in LC after cell therapy relative to the other treatment groups. Moreover, we found that ECM1-HF-MSCs homed to the injured liver and expressed the hepatocyte-specific surface markers ALB, CK18, and AFP. In addition, hepatic stellate cell (HSC) activation was significantly inhibited in the cell treatment groups in vivo and in vitro, especially in the ECM1-HF-MSC group. Additionally, TGF-β/Smad signal inhibition was the most significant in the ECM1-HF-MSC group in vivo and in vitro. The findings indicate that the genetic modification of HF-MSCs with bioinformatic tools may provide a broad perspective for precision treatment of LC.
Article
Full-text available
Matrix remodeling could be an important mode of action of multipotent mesenchymal stromal cells (MSC) in extracellular matrix (ECM) disease, but knowledge is limited in this respect. As MSC are well-known to adapt their behavior to their environment, we aimed to investigate if their mode of action would change in response to healthy versus pathologically altered ECM. Human MSC-derived ECM was produced under different culture conditions, including standard culture, culture on Matrigel-coated dishes, and stimulation with the pro-fibrotic transforming growth factor-β1 (TGFβ1). The MSC-ECM was decellularized, characterized by histochemistry, and used as MSC culture substrate reflecting different ECM conditions. MSC were cultured on the different ECM substrates or in control conditions for 2 days. Culture on ECM increased the presence of surface molecules with ECM receptor function in the MSC, demonstrating an interaction between MSC and ECM. In MSC cultured on Matrigel-ECM and TGFβ1-ECM, which displayed a fibrosis-like morphology, gene expression of collagens and decorin, as well as total matrix metalloproteinase (MMP) activity in the supernatant were decreased as compared with control conditions. These results demonstrated that MSC adapt to their ECM environment, which may include pathological adaptations that could compromise therapeutic efficacy.
Article
Wound healing is the response of tissue to injury that results in scar formation. Tissue regeneration would be a more ideal response. Previously, we have isolated a population of cells from avian, rodent, and rabbit skeletal muscle capable of differentiating into multiple mesodermal phenotypes. The present experiments were designed to determine whether a similar population of cells exist in human skeletal muscle. Separate cell preparations from skeletal muscle on an amputated leg of a 75-year-old female and the pectoralis muscle of a 27-year-old male were enzymatically dissociated and cultured to confluence in Eagle's minimal essential medium with 10 per cent preselected horse serum, then trypsinized, filtered, and slowly frozen in 7.5 per cent dimethylsulfoxide to -80° C. The cells were thawed and plated with the same media plus dexamethasone (a nonspecific differentiation agent) at 10– 10– -10– ⁶ M concentrations for up to 6 weeks. Immunological and histochemical staining assays were performed. Phenotypes observed included stem cells with typical stellate morphology (control), skeletal myotubes (anti-myosin), smooth muscle (anti-a-actin), bone (von Kossa stain), cartilage (Alcec blue), and fat (Sudan black B). These experiments establish the existence of a population of mesenchymal stem cells in human skeletal muscle capable of differentiating into multiple mesodermal phenotypes. The possibility exists of manipulating the mesenchymal stem cells to achieve appropriate regeneration of mesenchymal tissues in the injured patient.
Article
Mesenchymal stem/stromal cells (MSCs) exist in almost all tissues and participate in tissue regeneration and homeostasis. In-vitro-expanded MSCs are employed as therapeutics for autoimmune diseases, organ failures, and many other chronic disorders. Remarkably, the reparative and homeostatic maintenance functions of MSCs rely on their interaction with the inflammatory microenvironment. Here, we discuss the characteristics and functions of MSCs under different pathophysiological conditions and highlight how the immunomodulatory functions of MSCs are altered in accordance with the inflammatory cues. We hope to provide new insights into the diverse immunoregulatory properties of MSCs during tissue regeneration and therapy.
Article
Background: Liver inflammation is the main cause of severe liver diseases, including liver fibrosis, steatohepatitis, cirrhosis and hepatocellular carcinoma. Cell therapy topics are receiving increasingly more attention. The therapeutic applications of mesenchymal stem cells (MSC) have become one of the most discussed issues. While other stem cells have therapeutic effects, they have only one or two clinical applications. MSCs are responsible for repairing a variety of tissue injuries. Moreover, MSCs could be derived from several sources, including adipose tissue. MSCs are usually more abundant and easier to obtain compared to other stem cells. Methods: To prove the concept that MSCs have homing ability to the injured tissue and assist in tissue repair, we examined the effects of intravenous injected adipose-derived mesenchymal stem cells (ADSCs) in a N-nitrosodiethylamine (DEN)-induced liver injury rat model. Results: The significant repairing ability of ADSCs was observed. The levels of fibrosis, apoptosis, and tumorigenesis in the DEN-injured liver tissues all decreased after ADSC treatment. Furthermore, to enhance the therapeutic effects of ADSCs, we pretreated them with L-theanine, which promotes the hepatocyte growth factor secretion of ADSC, and therefore improved the healing effects on injured liver tissue. Conclusion: ADSCs, especially L-theanine-pretreated ADSCs, have anti-inflammation, anti-apoptosis, and anti-tumorigenesis effects on the N-nitrosodiethylamine-induced liver injury rat model.
Article
Fibrosis is caused by extensive deposition of extracellular matrix (ECM) components, which play a crucial role in injury repair. Fibrosis attributes to ~45% of all deaths worldwide. The molecular pathology of different fibrotic diseases varies, and a number of bioactive factors are involved in the pathogenic process. Mesenchymal stem cells (MSCs) are a type of multipotent stem cells that have promising therapeutic effects in the treatment of different diseases. Current updates of fibrotic pathogenesis reveal that residential MSCs may differentiate into myofibroblasts which lead to the fibrosis development. However, preclinical and clinical trials with autologous or allogeneic MSCs infusion demonstrate that MSCs can relieve the fibrotic diseases by modulating inflammation, regenerating damaged tissues, remodeling the ECMs, and modulating the death of stressed cells after implantation. A variety of animal models were developed to study the mechanisms behind different fibrotic tissues and test the preclinical efficacy of MSC therapy in these diseases. Furthermore, MSCs have been used for treating liver cirrhosis and pulmonary fibrosis patients in several clinical trials, leading to satisfactory clinical efficacy without severe adverse events. This review discusses the two opposite roles of residential MSCs and external MSCs in fibrotic diseases, and summarizes the current perspective of therapeutic mechanism of MSCs in fibrosis, through both laboratory study and clinical trials.