ArticlePDF AvailableLiterature Review

Mesenchymal stem cell therapies for liver cirrhosis: MSCs as “conducting cells” for improvement of liver fibrosis and regeneration

Authors:

Abstract and Figures

Mesenchymal stem cells (MSCs) can be cultured relatively easily and can be obtained not only from the bone marrow, but also from medical waste such as adipose tissue and umbilical cord tissue. Because of its low antigenicity, allogeneic MSC injection is safe. MSCs have been evaluated in more than 900 clinical trials in a variety of fields, with more than 50 clinical trials related to liver diseases. Experiments have suggested that MSCs function as “conducting cells” to affect various “effective cells” such as T cells, B cells, and macrophages. Recent clinical trials have focused on allogeneic MSCs. Thus, studies are needed to determine the most effective cell source, culture conditions, cell numbers, administration frequency, administration route, cost, safety, and liver disease treatments. Recently, the functions of exosomes have gained attention, and cell-free therapy may become possible as an alternative therapy for liver disease. In this review, we introduce general information, mechanism, representative clinical study data, recently started or planned clinical trials, and possibility of cell-free therapy of MSCs.
Content may be subject to copyright.
R E V I E W Open Access
Mesenchymal stem cell therapies for liver
cirrhosis: MSCs as conducting cellsfor
improvement of liver fibrosis and
regeneration
Atsunori Tsuchiya
*
, Suguru Takeuchi, Takayuki Watanabe, Tomoaki Yoshida, Shunsuke Nojiri, Masahiro Ogawa and
Shuji Terai
*
Abstract
Mesenchymal stem cells (MSCs) can be cultured relatively easily and can be obtained not only from the bone
marrow, but also from medical waste such as adipose tissue and umbilical cord tissue. Because of its low
antigenicity, allogeneic MSC injection is safe. MSCs have been evaluated in more than 900 clinical trials in a variety
of fields, with more than 50 clinical trials related to liver diseases. Experiments have suggested that MSCs function
as conducting cellsto affect various effective cellssuch as T cells, B cells, and macrophages. Recent clinical trials
have focused on allogeneic MSCs. Thus, studies are needed to determine the most effective cell source, culture
conditions, cell numbers, administration frequency, administration route, cost, safety, and liver disease treatments.
Recently, the functions of exosomes have gained attention, and cell-free therapy may become possible as an
alternative therapy for liver disease. In this review, we introduce general information, mechanism, representative
clinical study data, recently started or planned clinical trials, and possibility of cell-free therapy of MSCs.
Keywords: Liver cirrhosis, Acute on chronic, Mesenchymal stem cell, Cell therapy
Background
The liver, which is a vital organ, has many functions
such as protein, triglyceride, cholesterol, and glycogen
synthesis; detoxication; drug metabolism; and bile secre-
tion, and has a high regenerative potential. However, the
liver cannot withstand long-term chronic injury, severe
acute injury, and acute on chronic injury. Chronic liver
injuries such as those caused by hepatitis B virus (HBV)
and hepatitis C virus infections, non-alcoholic steatohe-
patitis (NASH), and alcoholic liver injury have been
widely studied. Because of the recent development of
anti-viral drugs, it is easy to control HBV [1] and eradi-
cate hepatitis C virus [2]. In contrast, the number of pa-
tients with NASH and alcoholic liver disease is
increasing and no effective drugs except conventional
alimentary therapy and exercise therapy are available;
thus, development of new therapies for these diseases is
becoming important [3]. Long-term damaged conditions
gradually result in the loss of liver function and accumu-
lation of extracellular matrix (ECM), finally leading to
liver cirrhosis. Particularly, the prognosis of patients with
decompensated liver cirrhosis is poor. Hepatic stellate cells
are central players in liver fibrosis and the major precursors of
activated myofibroblasts, which produce ECM during liver fi-
brosis [4,5]. A recent study reported that fibrosis, rather than
steatosis, determines the prognosis of patients [6,7]. In acute
liver injury such as hepatitis B, hepatitis A, and drug-induced
liver injury, patients with excessive damage cannot be suffi-
ciently treated by physicians. Additionally, acute on chronic
liver failure (ACLF), the definition of which slightly differs by
region [811], is also a recently focused disease condition.
Recently proposed diagnostic criteria for ACLF in Japan in-
clude patients with cirrhosis and a ChildPugh score of 59
should be diagnosed as having ACLF when a deterioration
of liver function (serum bilirubin level 5.0 mg/dL and pro-
thrombin time value 40% of the standardized values and/or
© The Author(s). 2019 Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0
International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and
reproduction in any medium, provided 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 Creative Commons Public Domain Dedication waiver
(http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.
* Correspondence: atsunori@med.niigata-u.ac.jp;terais@med.niigata-u.ac.jp
Division of Gastroenterology and Hepatology, Graduate School of Medical
and Dental Science, Niigata University, 1-757 Asahimachi-dori, Chuo-ku,
Niigata 951-8510, Japan
Inflammation and Regeneration
Tsuchiya et al. Inflammation and Regeneration (2019) 39:18
https://doi.org/10.1186/s41232-019-0107-z
international normalization rate 1.5) caused by severe liver
damage develops within 28 days after acute insults, such as
alcohol abuse, bacterial infection, gastrointestinal bleeding,
or the exacerbation of underlying liver diseases[11]. These
chronic, acute, and acute on chronic liver injuries can cause
death. Although liver transplantation can be conducted, the
shortage of donor organs is a serious problem. Cell therapies
maybeusefulfortreatingthesediseases.Wefirstlyshowed
that autologous bone marrow cell infusion (ABMi) therapy
was effective for the decompensation of liver cirrhosis pa-
tients [12]. Several clinical cases in this study [13]have
shown that bone marrow-derived cells can improve liver fi-
brosis and subsequently improve liver function. Therefore,
recently, we shifted our focus to cell therapies using mesen-
chymal stem cells. Mesenchymal stem cells (MSCs) have
been widely examined in clinical trials to evaluate their safety
and effectiveness in improving liver fibrosis and liver func-
tion. Positive results have been observed in numerous studies
of animal models [1417].
In this review, we describe the general information of
MSCs, mechanisms of MSC therapies (i.e., the conduct-
ing effect of MSCs), recently published outcomes of
MSC therapy, clinical trials that have recently started or
will begin soon, and recent research trends using extra-
cellular vesicles obtained from MSCs.
General information of MSCs
MSCs have been used in many fields to treat a variety of
diseases such as neural, heart, liver, intestinal, and lung
diseases. According to ClinicalTrials.Gov, more than 900
clinical trials have been registered in a variety of fields
and are increasing by nearly 100 trials each year. MSCs
can be obtained not only from the bone marrow, but
also from medical waste such as umbilical cord tissue,
adipose tissue, amniotic tissue, and dental pulp. These
cells are relatively easy to expand, maintain, and cryopre-
serve, while maintaining their viability. MSCs are positive
for the common markers CD73, CD90, and CD105 and
show differential potential towards adipocytes, osteoblasts,
and chondroblasts under appropriate conditions. The cells
are used to replace damaged cells or tissues mainly in the
orthopedic field; however, their main functions are deter-
mined by trophic factors including chemokines, cytokines,
growth factors, and exosomes, and MSCs exhibit anti-in-
flammation, anti-oxidant, angiogenesis, and anti-fibrosis ef-
fects [4,5]. Of these functions, MSCs are most commonly
applied to achieve anti-inflammation effects. MSCs produce
various factors such as nitric oxide/indoleamine 2,3-dioxy-
genase, interleukin (IL)-10, tumor necrosis factor-inducible
gene-6, and prostaglandin E2; inhibit T cell activation and
expansion; induce regulatory T cells; alter the polarity of
macrophages to the anti-inflammatory phenotype; and con-
trol the function of dendritic cells, B cells, and natural killer
cells [18,19]. Another important characteristic of MSCs is
that they generally have low immunogenicity. MSCs ex-
press low or modest levels of major histocompatibility com-
plex class I molecules and lack expression of major
histocompatibility complex class II and co-stimulatory mol-
ecules, such as CD40, CD80, and CD86 (B72), leading to
low immunogenicity, suggesting that MSCs can avoid im-
mune responses in recipients; thus, injection of autologous
or allogenic MSCs has been employed in clinical studies.
For example, Lalu et al. performed a meta-analysis of the
safety of MSCs in clinical trials and found that autologous
and allogenic MSC therapies are related to transient fever
but not related to infusion toxicity, organ system complica-
tions, infection, death, and malignancies [20]. Allogenic
MSC therapy has the potential to be applied in many
patients.
MSCs function as conducting cellsin liver
disease
The mechanisms of MSCs for treating liver diseases have
been evaluated from various perspectives in basic stud-
ies. MSCs have anti-inflammatory effects and reduce
damages to hepatocytes [21]. These anti-inflammatory
effects and decreases in hepatocyte damage reduce the
activation of hepatic stellate cells [22] and direct the ef-
fects of MSCs to reduce hepatic stellate cell activation
[23]. Additionally, we recently reported the effects
against macrophages. MSCs change the polarity of mac-
rophages towards an anti-inflammatory phenotype, in-
crease the production of matrix metalloproteinases to
reduce the ECM, and increase the ability of phagocytosis
of hepatocyte debris (during this process, macrophages
increase the levels of pro-regenerative factors) (Fig. 1)
[14]. When we administered bone marrow-derived
MSCs with macrophages produced by culturing bone
marrow cells for 7 days, host macrophages and neutro-
phils were also recruited to the liver.
The behaviors of macrophages after administration
showed differences between studies. Some studies re-
ported that administered MSCs can migrate to and func-
tion in the liver [24]. However, our recent studies of a
liver cirrhosis mouse model using two-photon excitation
microscopy revealed that when bone marrow-derived
macrophages (green fluorescent protein-labeled) and
MSCs (DsRed-labeled) were administered at the same
time via the tail vein, most MSCs migrated to the lung
and a low percentage of MSCs migrated to the liver,
which disappeared from both the lung and liver after 7
days. In contrast, macrophages migrated to both the
lung and liver, where they remained for 7 days. Particu-
larly, in the liver, administered macrophages migrated to
the damaged area where excess ECM and hepatocyte
debris were detected. Furthermore, we observed that the
administered GFP-positive macrophages phagocytosed
the debris in hepatocytes in the liver [14].
Tsuchiya et al. Inflammation and Regeneration (2019) 39:18 Page 2 of 6
While some aspects of the mechanisms of MSCs remain
unclear, these results revealed that MSCs function indir-
ectly as conducting cells,while macrophages, T cells, B
cells, and other cells function directly as effective cells.
Summary of MSC therapies from recently
published papers
Many reports have been published describing the results of
clinical trials using MSCs. Zhao et al. reported a meta-ana-
lysis of previously published papers up to June 2017. They
evaluated 23 reports of studies comparing MSC therapy to
conventional treatment. The authors concluded that MSC-
based therapy is relatively safe and improved liver function
during the first 6 months after administration. A single in-
jection administration via the hepatic artery and MSCs de-
rived from the bone marrow are optimal in terms of
improving liver function [25]. The analyzed cases were
quite heterogenous and included papers only written in
Chinese; thus, next, we show two representative studies.
Suk et al. reported a phase II study using bone marrow-de-
rived MSCs for treating alcoholic liver cirrhosis. An MSC
culture from 10 to 20 ml of bone marrow aspirated 1 month
before the first injection was administered one or two times
(5 × 10
7
cells/time)viathehepaticarteryandcomparedto
the control. Results of biopsy performed 6 months after cell
administration revealed 25% (one-time cell administration)
and 37% (two administrations) reductions in the fibrosis area.
Furthermore, the Child-Pugh scores of both the single and
double administration groups were improved significantly at
12 months after cell injection [26].
Lin et al. reported an open-label non-blinded random-
ized controlled study using 1.010 × 10
5
cells/kg of allo-
geneic bone marrow cells for treating patients with
HBV-related ACLF once per week for 4 weeks; these
subjects were followed for 24 weeks. The clinical labora-
tory results showed that serum total bilirubin and model
for end-stage liver disease scores were improved com-
pared to those obtained after standard medical therapy.
The authors further observed that the incidences of se-
vere infection and mortality from multiple organ failure
were reduced after cell administration [27].
Two representative cases of cirrhosis and ACLF of MSC
therapy were shown above, both of which were treated
safely and showed some favorable effects. Based on these
studies, additional clinical studies were designed. The next
section describes recently designed clinical trials.
Recently started or planned clinical trials
To describe recent trends in clinical trials using MSCs, we
evaluated clinical studies which began or will begin after 1
January 2017 according to ClinicalTrials.gov. Thirteen clin-
ical trials were registered; 1 case was a follow-up study of a
clinical trial, and thus, we excluded this case and analyzed
the other 12 cases. As shown in Tables 1, 8 of 12 (66.7%)
cases were from China and 1 case (8.3%) each occurred in
Germany, Japan, Taiwan, and Singapore (Fig. 2a). Four of
12 cases (33.3%) were ACLF, 7 of 12 cases (58.3%) were cir-
rhosis in which each studys etiology of cirrhosis slightly dif-
fered, and 1 case (8.3%) was a target for primary biliary
cholangitis (Fig. 2b). Five of 12 (41.6%) cases were treated
with allogeneic MSCs, and 2 of 12 cases were treated with
autologous MSCs (16.7%). In 5 of 12 cases, it was unclear
whether allogeneic or autologous cells were used; however,
based on the study design and disease condition, allogeneic
MSCs were administered in most cases, suggesting that
allogeneic cases were increased compared to previously
Fig. 1 Mechanisms of MSCs for liver disease. MSCs have various effects including the reduction of hepatocyte injury and inflammation.
Additionally, MSCs affect macrophages and increase matrix metalloproteinase expression and phagocytosis, promoting the regenerative process
Tsuchiya et al. Inflammation and Regeneration (2019) 39:18 Page 3 of 6
Table 1 Recently started or planned clinical trials
Country Conditions Auto/
Allo
Origin Cell number Cell injection
times
Route Phase Study design
1 China ACLF N/A N/A 110 × 10
5
/kg 4 Peripheral
vein
I/II Randomized/open label
2 China ACLF N/A N/A 0.11×10
6
/kg 3 Peripheral
vein
N/A Randomized/double
blind
3 Germany ACLF Allo Skin (ABCB5+
cells)
2×10
6
/kg 3 Peripheral
vein
I/II Non-randomized/open
label
4 China Cirrhosis N/A N/A N/A N/A N/A I/II Non-randomized/open
label
5 China Cirrhosis Allo UC 1.5 × 10
6
/kg 2~4 Peripheral
vein
II Non-randomized/open
label
6 China Cirrhosis (HBV, HCV) N/A N/A 1 × 10
6
/kg 4 Peripheral
vein
N/A Randomized/single
blind
7 Japan Cirrhosis (NASH,
HCV)
Allo AD N/A 1 Peripheral
vein
I/II Non-randomized/open
label
8 Taiwan ACLF Allo AD 0.52×10
6
/kg N/A Peripheral
vein
I Non-randomized/open
label
9 Singapore Cirrhosis Auto BM 0.51×10
6
/kg N/A Peripheral
vein
I/II Non-randomized/open
label
10 China Alcoholic liver
cirrhosis
Auto BM 5 × 10
7
cells/10
ml
1 Hepatic
artery
I Non-randomized/open
label
11 China Cirrhosis (HBV) Allo UC 6 × 10
7
(30 ml) N/A Peripheral
vein
I Non-randomized/open
label
12 China PBC N/A N/A 0.11×10
6
/kg 3 Peripheral
vein
N/A Randomized/double
blind
Fig. 2 Recent trends in clinical trials using MSCs. Proportion of country (a), disease conditions (b), autologous or allogeneic (c), and tissue origin
of MSCs (d) in recent clinical trials
Tsuchiya et al. Inflammation and Regeneration (2019) 39:18 Page 4 of 6
reported frequencies (allogeneic, 53%; autologous, 45%)
(Fig. 2c). Regarding the cell origin, 1 case (8.3%) occurred
in the skin, while the other cases occurred in the bone
marrow (2 cases; 16.7%), adipose tissue (2 cases; 16.7%),
and umbilical cord tissue (2 cases; 16.7%). The cases of
bone marrow origin were all autologous cases (Fig. 2d).
The cell numbers employed in the trials slightly differed;
approximately 0.11.0 × 10
6
cells/kg were injected 14
times, cell administration in 10 of the 12 cases (83.3%)
was performed via the peripheral vein, and 1 case (8.3%)
involved autologous bone marrow case from the hepatic
artery, suggesting that the recent trend in administration
is the peripheral vein. However, further studies of direct
infusion of MSCs are necessary to achieve efficient effects.
All clinical trials are still in phase I or/and II.
Recent research trend of using extracellular
vesicles obtained from MSCs
Although most MSCs were trapped in the lung, they
showed therapeutic effects. To explain this phenomenon,
extracellular vesicles (EVs) have been evaluated [2834].
EVs include apoptotic bodies (504000 nm in diameter),
microvesicles (MVs; 1001000 nm in diameter), and exo-
somes (40100 nm in diameter), among which exosomes
are the most widely studied [31]. Exosomes are lipid vesi-
cles produced by multivesicular bodies prior to extracellular
secretion. They can be sedimented by ultra-centrifugation
and include endosome-derived components as well as
many bioactive molecules such as proteins, lipids, mRNAs,
microRNAs (miRNAs), long non-coding RNAs, transfer
RNA, genomic DNA, cDNA, and mitochondrial DNA [35].
Exosome membranes are enriched in cholesterol, sphingo-
myelin, ceramide, and lipid raft proteins. Exosomes are cell
type-specific; however, they contain evolutionarily con-
served sets of proteins including tetraspanins (CD81, CD63,
and CD9), heat shock proteins (HSP60, HSP70, and
HSP90), AlIX, and tumor susceptibility gene 101 and have
been reported to have multiple functions including angio-
genesis, cell proliferation, and collagen reduction. MSCs are
relatively easy to expand and well-known to produce abun-
dant exosomes and thus are theoretically ideal tools for de-
veloping cell-free therapies [28,3032,34]. Haga et al.
reported that mouse bone marrow-derived MSC EVs have
therapeutic effects for hepatic failure induced by D-galact-
osamine (D-gal) and tumor necrosis factor α. In this experi-
ment, high levels of EVs were observed after 6 h in the liver
and spleen. Jiang et al. reported that exosomes derived from
human umbilical cord-derived MSCs alleviate acute liver
failure (lipopolysaccharide/D-galactosamine-induced liver
injury model) [36]. They reported that MSC exosomes re-
duced the activation of the NLRP3 inflammasome, IL-1β,
and IL-6 in macrophages. Borrelli et al. reported the use of
drug-loaded EVs for treating hepatocellular carcinoma.
Exosomes contain diverse and numerous miRNAs, and
thus, determining the roles of miRNA is very difficult. Fer-
guson et al. reported that most previous studies used a can-
didate approach with specific miRNAs to assess their
therapeutic effects; however, this approach may not fully
capture the various biological effects induced by miRNAs
in the MSC exosomes of recipient cells [29]. System-level
studies are thus needed.
Conclusions
MSCs are attractive cell therapies that function as con-
ducting cellsagainst many types of immune cells and in-
duce a variety of therapeutic effects. However, several
years have passed since the first MSC theories were postu-
lated, and thus, these clinical studies must be evaluated to
determine whether MSC therapies are indeed effective in
human liver diseases. In recent clinical trials, the trend of
MSC therapy appears to have shifted from administration
of autologous cells towards allogeneic cells. This research
area is very attractive for developing effective anti-fibrotic
therapies. Studies are needed to determine the most ef-
fective cell source, culture condition, cell number, admin-
istration frequency, and administration route, at low cost
for treating specific liver diseases. Cell-free therapy using
exosomes is an attractive approach.
Abbreviations
ACLF: Acute on chronic liver failure; ECM: Extracellular matrix; EV: Extracellular
vesicle; MSC: Mesenchymal stem cell
Acknowledgements
The authors thank Dr. Yusuke Watanabe and Dr. Yuichi Kojima for their
cooperation.
Authorscontributions
AT, ST, TW, TY, SN, and MO analyzed and interpreted the previous literature
and clinical study patient. AT and ST were the major contributors in writing
the manuscript. All authors read and approved the final manuscript.
Funding
This work was supported by a Grant-in-Aid for Scientific Research (C) (18
K07903) from the Ministry of Education, Science, Technology, Sports, and Cul-
ture of Japan and by the Research Program on Hepatitis from AMED to Shuji
Terai (17fk0210101h0001).
Availability of data and materials
All data generated or analyzed during this study are included in this
published article.
Ethics approval and consent to participate
Not applicable.
Consent for publication
Not applicable.
Competing interests
The authors declare that they have no competing interests.
Received: 5 April 2019 Accepted: 8 August 2019
References
1. Tang LSY, Covert E, Wilson E, et al. Chronic hepatitis B infection: a review.
JAMA. 2018;319:180213.
Tsuchiya et al. Inflammation and Regeneration (2019) 39:18 Page 5 of 6
2. Kieslich K, Ahn J, Badano G, et al. Public participation in decision-making on the
coverage of new antivirals for hepatitis C. J Health Organ Manag. 2016;30:76985.
3. Tateishi R, Uchino K, Fujiwara N, et al. A nationwide survey on non-B, non-C
hepatocellular carcinoma in Japan: 2011-2015 update. J Gastroenterol. 2019;54:36776.
4. Tsuchiya A, Kojima Y, Ikarashi S, et al. Clinical trials using mesenchymal stem cells in
liver diseases and inflammatory bowel diseases. Inflamm Regen. 2017;37:16.
5. Terai S, Tsuchiya A. Status of and candidates for cell therapy in liver
cirrhosis: overcoming the point of no returnin advanced liver cirrhosis. J
Gastroenterol. 2017;52:12940.
6. Angulo P, Kleiner DE, Dam-Larsen S, et al. Liver fibrosis, but no other
histologic features, is associated with long-term outcomes of patients with
nonalcoholic fatty liver disease. Gastroenterology. 2015;149:38997 e10.
7. Hagstrom H, Nasr P, Ekstedt M, et al. Fibrosis stage but not NASH predicts
mortality and time to development of severe liver disease in biopsy-proven
NAFLD. J Hepatol. 2017;67:126573.
8. Sarin SK, Kumar A, Angus PW, et al. Diagnosis and management of acute
variceal bleeding: Asian Pacific Association for Study of the Liver
recommendations. Hepatol Int. 2011;5:60724.
9. Olson JC, Wendon JA, Kramer DJ, et al. Intensive care of the patient with
cirrhosis. Hepatology. 2011;54:186472.
10. Jalan R, Gines P, Olson JC, et al. Acute-on chronic liver failure. J Hepatol.
2012;57:133648.
11. Mochida S, Nakayama N, Ido A, et al. Proposed diagnostic criteria for acute-
on-chronic liver failure in Japan. Hepatol Res. 2018;48:21924.
12. Terai S, Ishikawa T, Omori K, et al. Improved liver function in patients with
liver cirrhosis after autologous bone marrow cell infusion therapy. Stem
Cells. 2006;24:22928.
13. Terai S, Sakaida I. Current status of autologous bone marrow cell infusion
therapy for liver cirrhosis patients. Hepatol Res. 2008;38(Suppl 1):S725.
14. Watanabe Y, Tsuchiya A, Seino S, et al. Mesenchymal stem cells and
induced bone marrow-derived macrophages synergistically improve liver
fibrosis in mice. Stem Cells Transl Med. 2019;8:27184.
15. Iwamoto T, Terai S, Hisanaga T, et al. Bone-marrow-derived cells cultured in
serum-free medium reduce liver fibrosis and improve liver function in
carbon-tetrachloride-treated cirrhotic mice. Cell Tissue Res. 2013;351:48795.
16. Lee CW, Hsiao WT, Lee OK. Mesenchymal stromal cell-based therapies
reduce obesity and metabolic syndromes induced by a high-fat diet. Transl
Res. 2017;182:6174 e8.
17. Chen L, Zhang C, Chen L, et al. Human menstrual blood-derived stem cells
ameliorate liver fibrosis in mice by targeting hepatic stellate cells via
paracrine mediators. Stem Cells Transl Med. 2017;6:27284.
18. Kawata Y, Tsuchiya A, Seino S, et al. Early injection of human adipose tissue-
derived mesenchymal stem cell after inflammation ameliorates dextran
sulfate sodium-induced colitis in mice through the induction of M2
macrophages and regulatory T cells. Cell Tissue Res. 2019;376(2):25771.
https://doi.org/10.1007/s00441-018-02981-w.
19. Volarevic V, Gazdic M, Simovic Markovic B, et al. Mesenchymal stem cell-
derived factors: immuno-modulatory effects and therapeutic potential.
Biofactors. 2017;43:63344.
20. Lalu MM, McIntyre L, Pugliese C, et al. Safety of cell therapy with
mesenchymal stromal cells (SafeCell): a systematic review and meta-analysis
of clinical trials. PLoS One. 2012;7:e47559.
21. Forbes SJ, Newsome PN. New horizons for stem cell therapy in liver disease.
J Hepatol. 2012;56:4969.
22. Wang L, Li J, Liu H, et al. Pilot study of umbilical cord-derived mesenchymal
stem cell transfusion in patients with primary biliary cirrhosis. J
Gastroenterol Hepatol. 2013;28(Suppl 1):8592.
23. An SY, Jang YJ, Lim HJ, et al. Milk fat globule-EGF factor 8, secreted by
mesenchymal stem cells, protects against liver fibrosis in mice.
Gastroenterology. 2017;152:117486.
24. Sohni A, Verfaillie CM. Mesenchymal stem cells migration homing and
tracking. Stem Cells Int. 2013;2013:130763.
25. Zhao L, Chen S, Shi X, et al. A pooled analysis of mesenchymal stem cell-
based therapy for liver disease. Stem Cell Res Ther. 2018;9:72.
26. Suk KT, Yoon JH, Kim MY, et al. Transplantation with autologous bone
marrow-derived mesenchymal stem cells for alcoholic cirrhosis: phase 2
trial. Hepatology. 2016;64:218597.
27. Lin BL, Chen JF, Qiu WH, et al. Allogeneic bone marrow-derived
mesenchymal stromal cells for hepatitis B virus-related acute-on-chronic
liver failure: a randomized controlled trial. Hepatology. 2017;66:20919.
28. Jiang L, Zhang S, Hu H, et al. Exosomes derived from human umbilical cord
mesenchymal stem cells alleviate acute liver failure by reducing the activity
of the NLRP3 inflammasome in macrophages. Biochem Biophys Res
Commun. 2019;508:73541.
29. Ferguson SW, Wang J, Lee CJ, et al. The microRNA regulatory landscape of
MSC-derived exosomes: a systems view. Sci Rep. 2018;8:1419.
30. Phinney DG, Pittenger MF. Concise review: MSC-derived exosomes for cell-
free therapy. Stem Cells. 2017;35:8518.
31. Lou G, Chen Z, Zheng M, et al. Mesenchymal stem cell-derived exosomes as
a new therapeutic strategy for liver diseases. Exp Mol Med. 2017;49:e346.
32. Damania A, Jaiman D, Teotia AK, et al. Mesenchymal stromal cell-derived
exosome-rich fractionated secretome confers a hepatoprotective effect in
liver injury. Stem Cell Res Ther. 2018;9:31.
33. Borrelli DA, Yankson K, Shukla N, et al. Extracellular vesicle therapeutics for
liver disease. J Control Release. 2018;273:8698.
34. Qiu G, Zheng G, Ge M, et al. Mesenchymal stem cell-derived extracellular
vesicles affect disease outcomes via transfer of microRNAs. Stem Cell Res
Ther. 2018;9:320.
35. Cheng L, Zhang K, Wu S, et al. Focus on mesenchymal stem cell-derived
exosomes: opportunities and challenges in cell-free therapy. Stem Cells Int.
2017;2017:6305295.
36. Haga H, Yan IK, Takahashi K, et al. Extracellular vesicles from bone marrow-
derived mesenchymal stem cells improve survival from lethal hepatic failure
in mice. Stem Cells Transl Med. 2017;6:126272.
PublishersNote
Springer Nature remains neutral with regard to jurisdictional claims in
published maps and institutional affiliations.
Tsuchiya et al. Inflammation and Regeneration (2019) 39:18 Page 6 of 6
... Most acute liver injury is caused by short-term exposure and is commonly reversible by fibrotic remodeling [3]. On the other hand, chronic injury, severe acute injury, and acute-on-chronic injury are the one that pose serious problems for the liver as they may cause prolonged inflammation, imbalanced fibrotic remodeling, altered chemokines and cytokines, and could progress to liver fibrosis [3,4]. Long-term damaged conditions will cause liver function loss and accumulation of the ECM, progressing into liver cirrhosis [4]. ...
... On the other hand, chronic injury, severe acute injury, and acute-on-chronic injury are the one that pose serious problems for the liver as they may cause prolonged inflammation, imbalanced fibrotic remodeling, altered chemokines and cytokines, and could progress to liver fibrosis [3,4]. Long-term damaged conditions will cause liver function loss and accumulation of the ECM, progressing into liver cirrhosis [4]. Chronic liver injuries are one of the highest leading causes of death, especially in developing countries [5]. ...
... Chronic liver injury such as viral infection, nonalcoholic steatohepatitis (NASH), nonalcoholic fatty liver disease (NAFLD), and alcohol-related liver diseases (ALD) and its associated complications are responsible for approximately 1 million deaths annually [6]. Currently, the available and effective treatment plans for liver injury, liver fibrosis, liver cirrhosis, NAFLD, NASH, and its complications are limited, implicating the need for innovative treatment approaches [4][5][6]. ...
Article
Full-text available
Chronic liver injuries and their complications are leading causes of death, especially in developing countries (Sharma and Nagalli in Sex/Gender-Specific Medicine in the Gastrointestinal Diseases, StatPearls Publishing, 2023). The available and effective treatment plans are limited, implicating the need for innovative treatment approaches (Tsuchiya et al. in Inflamm Regener, 2019;Sharma and Nagalli in Sex/Gender-Specific Medicine in the Gastrointestinal Diseases, StatPearls Publishing, 2023;Younossi et al. in Clin Gastroenterol Hepatol 21:1978–1991, 2023;). This paper aims to summarize the effects and mechanisms of hUC-MSC-exo on liver injuries and its complications; it also suggests future directions for future research. The outcomes of interest are the morphology and histology of the liver, pathology score, liver function enzyme, glucose and lipid metabolism, and the effect hUC-MSC-exo had on gene regulation regarding liver diseases. A comprehensive review of nineteen studies was conducted to assess the effectiveness of the implementation of the hUC-MSC-Exo, instilling confidence in the validity of the findings. Regarding the morphology and histology of the liver and pathology score, hUC-MSC-exo treatment resulted in improved liver morphology post-treatment, as indicated by the reduction in pathology scores. However, these observed improvements in the liver surface are not directly attributed to the hUC-MSC-Exo itself but to the overall healing processes stimulated by the treatment. In physiological outcomes, hUC-MSC-exo also improves glucose and lipid metabolism, especially in diet-induced liver injury and its complications. In gene regulation, one interesting gene in this intervention is the fat mass and obesity-associated (FTO), in which hUC-MSC-exo combined with miRNAs can suppress FTO. HUC-MSC-Exo can improve by utilizing several possible pathways, targeting pinpoints in the pathogenesis of liver disease or glucose and lipid metabolism. This study presents hUC-MSC-exo better in all outcomes of interest compared to the control or sham group. Further specification of indications of the hUC-MSC-exo method may be beneficial and essential to be analyzed in future reviews to better understand the effectiveness of each hUC-MSC-exo dose, duration, and medium.
... Mesenchymal stem cells (MSCs), as the main stem cells for cell-based therapy, provide another option for liver organ or cell transplantation. Various stem cells from human bone marrow (BMSCs), umbilical cord (UCSCs) and adipose tissue (ADSCs) have been used in the treatment of liver diseases [7][8][9][10] . Clinical and experimental studies have shown that MSCs could effectively protect hepatocytes through transdifferentiating into functional hepatocytes, secreting a variety of immunosuppressive factors and nutritional factors, and secrete exosomes through autocrine or paracrine, thus playing an anti-fibrotic role and promoting liver regeneration 8,11 . ...
... Various stem cells from human bone marrow (BMSCs), umbilical cord (UCSCs) and adipose tissue (ADSCs) have been used in the treatment of liver diseases [7][8][9][10] . Clinical and experimental studies have shown that MSCs could effectively protect hepatocytes through transdifferentiating into functional hepatocytes, secreting a variety of immunosuppressive factors and nutritional factors, and secrete exosomes through autocrine or paracrine, thus playing an anti-fibrotic role and promoting liver regeneration 8,11 . In addition, there are several advantages of adipose mesenchymal stem cells 12 . ...
Article
Full-text available
Liver fibrosis is a chronic liver disease with progressive wound healing reaction caused by liver injury. Currently, there is no FDA approved drugs for liver fibrosis. Human adipose mesenchymal stem cells (hADSCs) have shown remarkable therapeutic effects in liver diseases. However, few studies have evaluated the therapeutic role of hADSCs in liver fibrosis, and the detailed mechanism of action is unknown. Here, we investigated the in vitro and in vivo anti-fibrosis efficacy of hADSCs and identified important metabolic changes and detailed mechanisms through transcriptomic and metabolomic analyses. We found that hADSCs could inhibit the proliferation of activated hepatic stellate cells (HSCs), promote their apoptosis, and effectively inhibit the expression of pro-fibrotic protein. It can significantly reduce collagen deposition and liver injury, improve liver function and alleviate liver inflammation in cirrhotic mouse models. In addition, transcriptome analysis revealed that the key mechanism of hADSCs against liver fibrosis is the regulation of AGE-RAGE signaling pathway. Metabolic analysis showed that hADSCs influenced changes of metabolites in lipid metabolism. Therefore, our study shows that hADSCs could reduce the activation of hepatic stellate cells and inhibit the progression of liver fibrosis, which has important potential in the treatment of liver fibrosis as well as other refractory chronic liver diseases.
... Recently, their multi-potentiality and peculiar ability to self-renew have given them a bright approach to being involved in the treatment of different diseases. Several clinical trials have been performed to assess the potency of MSCs in ameliorating several diseases (Tsuchiya et al. 2019;Shin et al. 2020). ...
Article
Full-text available
High fructose diet (HFrD) has been approved to be involved in the pathogenesis of insulin resistance. Mesenchymal stem cells have a vital role in the treatment of various diseases including metabolic disturbances. We investigated the effect of Adipose-derived mesenchymal stem cells (ADMSCs) against HFrD-induced metabolic disorders and the molecular mechanisms for this effect. Rats were divided into 3 groups; control, HFrD, and combined HFrD with ADMSCs. We assessed liver functions, gut microbiota activity, oxidative stress, adiponectin, and IL10 levels. Also, we measured SREBP-1, IRS-1 expression using Western blot, and Malat1 expression using rt-PCR. ADMSCs antagonized metabolic abnormalities induced by HFrD in the form of improvement of liver functions and alleviation of oxidative stress. In addition, ADMSCs ameliorated gut microbiota activity besides the elevation of adiponectin and IL10 levels. ADMSCs attenuated insulin resistance through upregulation of IRS1 and downregulation of SREBP-1 and Malat1. ADMSCs can protect against HFrD-induced metabolic hazards.
... The increase in IL-10 levels and the reduction of TNF-α and PGE2 expression indicate the antiinflammatory properties of HS-MSCs, which play a pivotal role in supporting wound healing and tissue repair [36,37] Alongside the rise in IL-10 levels, HS-MSCs showed a significant decrease in the expression of pro-inflammatory cytokines TNF-α and PGE2. This decrease in cytokines suggests the secretome's crucial role in dampening the inflammatory environment within the burn wound area, creating conditions favorable for the wound healing process [9,10,38,39] The inhibition of PGE2 leads to immunosuppressive effects, hindering T-cell proliferation, thus reducing inflammation and facilitating wound healing. PGE2 exhibits evident anti-inflammatory and pro-angiogenic effects by shifting macrophages from the M1 to the M2 phenotype at injured sites. ...
Article
Full-text available
Background: Severe third-degree burns pose a notable obstacle in clinical settings, frequently leading to hindered healing of wounds and the development of scars. In this research, we explored how the secretome of hypoxic mesenchymal stem cells (HS-MSCs) could be improved the healing of burn wounds. Methods: In this study, we used 20 male Wistar rats that induced third degree wound by metal 80 exposure for 10 s. These rats divided into 4 groups: Healthy, control, HS-MSCs 100 µL, and HS-MSCs 200 µL groups. We injected the HS-MSCs, which carry various beneficial molecules like IL-10, TGF-β, PDGF, and VEGF, directly into the skin in a third-degree burn rat model on day 1 after burn induction, and harvest on day 14. Results: Our research discovered that using HS-MSCs resulted in a dosage-related rise in IL-10, an anti-inflammatory cytokine, alongside a notable decrease in TNF-α and PGE2, pro-inflammatory cytokines. These effects quickened the healing process by improving re-epithelialization and minimizing scar formation. Conclusions: Our findings indicate that HS-MSCs show promise as a treatment for severe burns by potentially controlling inflammation and fostering a regenerative healing process. More investigation is needed to understand the exact mechanisms and to explore how HS-MSCs could be applied in clinical treatments. HIGHLIGHTS HS-MSCs suppress inflammation and promote regenerative healing in severe burns HS-MSCs accelerated re-epithelialization and reduced scarring GRAPHICAL ABSTRACT
... Moreover, EV miR-411-5p from M2 macrophages suppresses HSC activation by targeting calmodulin-regulated spectrin-associated protein 1 (CAMSAP1), which is one of the plausible aims of MASH treatment [115]. Second, the potential therapeutic effects of MSCderived EVs (MSC-EXos) in liver diseases have been confirmed [116]. MCSs are pluripotent stem cells that originate from early embryonic development, are widely used for regulating inflammation and are easily isolated from diverse tissues, including bone marrow, muscles, the umbilical cord, adipose tissue, and tendons. ...
Article
Full-text available
Liver fibrosis, a chronic and long-term disease, can develop into hepatocellular carcinoma (HCC) and ultimately lead to liver failure. Early diagnosis and effective treatment still face significant challenges. Liver inflammation leads to liver fibrosis through continuous activation of hepatic stellate cells (HSCs) and the accumulation of immune cells. Intracellular communication among various immune cells is important for mediating the inflammatory response during fibrogenesis. Extracellular vesicles (EVs), which are lipid bilayer membrane-enclosed particles naturally secreted by cells, make great contributions to cell-cell communication and the transport of bioactive molecules. Nearly all the cells that participate in liver fibrosis release EVs loaded with lipids, proteins, and nucleic acids. EVs from hepatocytes, immune cells and stem cells are involved in mediating the inflammatory microenvironment of liver fibrosis. Recently, an increasing number of extracellular vesicle-based clinical applications have emerged, providing promising cell-free diagnostic and therapeutic tools for liver fibrosis because of their crucial role in immunomodulation during pathogenesis. The advantages of extracellular vesicle-based therapies include stability, biocompatibility, low cytotoxicity, and minimal immunogenicity, which highlight their great potential for drug delivery and specific treatments for liver fibrosis. In this review, we summarize the complex biological functions of EVs in the inflammatory response in the pathogenesis of liver fibrosis and evaluate the potential of EVs in the diagnosis and treatment of liver fibrosis.
... 13,14 This supports the emerging evidences on the critical role of MSCs in liver repair and fibrosis regression which is mediated via modulation of macrophage phenotype. 15,16 Evaluating the therapeutic potential of MSC-derived EVs in liver fibrosis has also shown their ability to alleviate liver damage and promote tissue regeneration by reducing the secretion of proinflammatory cytokines, while promoting the proliferation of anti-inflammatory cells, such as M2 macrophages and Tregs both in vitro and in vivo. [17][18][19] Compared to MSCs derived from BM or AD, WJ-MSCs have shown better safety and compatibility, higher proliferation and secretion capacity. ...
Article
Full-text available
The potential of extracellular vesicles (EVs) isolated from mesenchymal stromal cells in guiding macrophages toward anti‐inflammatory immunophenotypes, has been reported in several studies. In our study, we provided experimental evidence of a distinctive effect played by Wharton Jelly mesenchymal stromal cell‐derived EVs (WJ‐EVs) on human macrophages. We particularly analyzed their anti‐inflammatory effects on macrophages by evaluating their interactions with stellate cells, and their protective role in liver fibrosis. A three‐step gradient method was used to isolate monocytes from umbilical cord blood (UCB). Two subpopulations of WJ‐EVs were isolated by high‐speed (20,000 g) and differential ultracentrifugation (110,000 g). Further to their characterization, they were designated as EV20K and EV110K and incubated at different concentrations with UCB‐derived monocytes for 7 days. Their anti‐fibrotic effect was assessed by studying the differentiation and functional levels of generated macrophages and their potential to modulate the survival and activity of LX2 stellate cells. The EV20K triggers the polarization of UCB‐derived monocytes towards a peculiar M2‐like functional phenotype more effectively than the M‐CSF positive control. The EV20K treated macrophages were characterized by a higher expression of scavenger receptors, increased phagocytic capacity and production level of interleukin‐10 and transforming growth factor‐β. Conditioned medium from those polarized macrophages attenuated the proliferation, contractility and activation of LX2 stellate cells. Our data show that EV20K derived from WJ‐MSCs induces activated macrophages to suppress immune responses and potentially play a protective role in the pathogenesis of liver fibrosis by directly inhibiting HSC’s activation.
... Numerous animal model studies have yielded positive results [24][25][26][27]. MSCs have anti-inflammatory, antioxidant, angiogenic, and anti-fibrotic properties, and they are mostly used to replace damaged cells or tissues; however, trophic factors, such as chemokines, cytokines, growth factors, and exosomes, dictate their main roles [28,29]. The fibrocyte appears to be a modest contributing cell type, at least in the kidney and liver, although reports suggest that a subpopulation of BM-derived cells known as fibrocytes directly lays down a fibrous matrix [30][31][32][33]. ...
Article
Full-text available
Aim This study explores the possible therapeutic role of rats and mice bone marrow-derived mesenchymal stem cells (BM-MSCs) on renal damage and toxicity brought on by carbon tetrachloride (CCl4) in Wistar rats. Methods Following an intraperitoneal injection of CCl4 (0.5 mL/kg b.w. twice weekly) for eight weeks, male Wistar rats were intravenously treated with rats and mice BM-MSCs (1 × 10⁶ cells in 0.2 mL Dulbecco’s Modified Eagle Medium (DMEM)/rat/week) a week for four weeks. Kidney functions were evaluated and kidney samples were examined using hematoxylin and eosin (H&E), Masson’s trichrome (MT) staining techniques, and electron microscopy analysis. Kidney cyclooxygenase-2 (COX-2), protein 53 (p53), and tumor necrosis factor-α (TNF-α) were detected by immunohistochemical staining techniques. Additionally, bioindicators of oxidative stress and antioxidant defense systems were identified in kidney tissue. Results In CCl4-injected rats, serum creatinine, urea, and uric acid levels significantly increased, as did renal lipid peroxidation (LPO), while superoxide dismutase, glutathione peroxidase (GPx), glutathione (GSH) transferase, and GSH levels significantly dropped in the kidneys. Histologically, the kidneys displayed a wide range of structural abnormalities, such as glomerular shrinkage, tubular dilations, inflammatory leukocytic infiltration, fibroblast proliferation, and elevated collagen content. Inflammatory cytokines like COX-2 and TNF-α as well as the pro-apoptotic mediator p53 were considerably upregulated. Treatment of BM-MSCs from mice and rats with CCl4-injected rats considerably reduced the previously noted abnormalities. Conclusions By boosting antioxidant defense and reducing apoptosis and inflammation, BM-MSCs from mice and rats were able to enhance kidney function and histological integrity in rats that had received CCl4 injections.
... Besides, MSCs have the ability to move forward damaged areas under the signals released by the lesion, which makes it possible to achieve transplantation through many different ways, including intravenous, intraperitoneal, intrahepatic, or portalvenous injection [57]. MSCs can also secrete trophic factors to improve the restoration and regeneration of impaired liver [58]. Vanessa et al. found that ATP7B overexpression provided a selection advantage to MSCs in high copper microenvironments and might represent novel cell transplants for therapy of WD [59]. ...
Article
The human central nervous system (CNS) has a limited capacity for regeneration and repair,as many other organs do. Partly as a result, neurological diseases are the leading cause of medical burden globally. Most neurological disorders cannot be cured, and primary treatments focus on managing their symptoms and slowing down their progression. Cell therapy for neurological disorders offers several therapeutic potentials and provides hope for many patients. Here we provide a general over- view of cell therapy in neurological disorders such as Parkinson’s disease (PD), Alzheimer’s disease(AD), Amyotrophic Lateral Sclerosis (ALS), Wilson’s Disease (WD), stroke and traumatic brain injury (TBI), involving many forms of stem cells, including embryonic stem cells and induced pluripotent stem cells. We also address the current concerns and perspectives for the future. Most studies for celltherapy in neurological diseases are in the pre-clinical stage, and there is still a great need for further research to translate neural replacement and regenerative therapies into clinical settings.
Article
Full-text available
Introduction Liver fibrosis is a crucial pathological factor in the persistence and progression of chronic liver disease. Increasing evidence has demonstrated the significant potential of extracellular vesicles (EVs) secreted by bone marrow mesenchymal stem cells (BMSCs) in the clinical treatment of liver fibrosis. This study aimed to mechanistically investigate the impact of BMSC-derived EVs (BMSC-EVs) containing miR-7045-5p on the autophagy of activated hepatic stellate cells (HSCs) during liver fibrosis. Method BMSCs were isolated from the bilateral femurs and tibiae of mice. Their identity was confirmed via immunofluorescence staining for the BMSC marker CD44. EVs were harvested from BMSC culture medium at passages 3–5 and then DiR-labeled. Labeled BMSC-EVs were co-cultured with the HSC-T6 cell line to determine their uptake and sub-cellular localization in HSCs. Various methods, such as western blotting, qRT-PCR, and ELISA, were employed to assess the effects of BMSC-EVs on the fibrotic activation (marked by COL1-A1 and α-SMA expression) and autophagy (p62, Atg16L1, Beclin-1, and LC3 expression) of HSC-T6 cells. Additionally, the BMSC-EV–induced changes in autophagy-related signaling pathways (PI3K, AKT, and mTOR pathways) in these cells were evaluated. Finally, the gene-chip detection technology was utilized to predict the involvement of BMSC-EV–derived miRNAs (BMSC-EV–miRs) in the observed effects, with a focus on miR-7045-5p, and our findings were validated in HSCs transfected with a miR-7045-5p mimic. Result The gene-chip detection results indicated that miR-7045-5p was enriched in BMSC-EVs compared with BMSCs and targeted Akt. In the CCl4-induced mouse model of liver fibrosis, BMSC-EV–miR-7045-5p ameliorated the fibrosis and enhanced liver function by suppressing the PI3K/Akt/mTOR signaling pathway. Additionally, miR-7045-5p inhibited TGF-β1–induced fibrotic activation of HSC-T6 cells. Conclusion BMSC-EVs promote autophagy in HSC-T6 cells and alleviate liver fibrosis by inhibiting the PI3K/Akt/mTOR signaling pathway at least in part by delivering anti-fibrotic miRNAs, such as miR-7045-5p.
Article
Viscoelastic mechanical characteristics of the liver play a crucial role in fibrosis and cancer progression. Despite the evolution of liver fibrosis, as a pivotal step toward cirrhosis and hepatoma, originates from molecular dysfunctions and further extends to cellular and tissue levels, current clinical assessment is still largely based on tissue level stiffness. This study introduces a novel multiscale viscoelastic signature-based machine learning model for liver fibrosis evaluation. Utilizing a hybrid hierarchical theory-microrheology approach, we unveil a universal two-stage power-law rheology capturing dynamic mechanical variations in diverse liver conditions. Our analysis of multiscale viscoelastic disparities through a self-similar hierarchical theoretical framework enhances our understanding of fibrosis evolution. Distinct mechanical signatures observed across liver states provide valuable insights for assessing fibrotic individuals and treatment responses at different spatial scales. Furthermore, we propose a series of threshold values for each marker in the diagnosis of liver fibrosis. Notably, based on these new viscoelastic signatures, we eventually propose a Light Gradient Boosting Machine (LightGBM) diagnostic model that outperforms conventional stiffness-based classification, offering superior diagnostic precision for fibrotic treatment. This research contributes to the growing knowledge of viscoelastic characteristics in soft tissues and holds promise for innovative diagnostic strategies in various diseases and cancers.
Article
Full-text available
Inflammatory bowel diseases (IBDs) are sometimes refractory to current therapy or associated with severe adverse events during immunosuppressive therapy; thus, new therapies are urgently needed. Recently, mesenchymal stem cells (MSCs) have attracted attention based on their multitude of functions including anti-inflammatory effects. However, proper timing of MSC therapy and the mechanisms underlying the therapeutic effects of MSCs on colitis are not fully elucidated. Human adipose tissue-derived mesenchymal stem cells (hAdMSCs; 1 × 10⁶) were administrated via the tail vein on day 3 (early) or 11 (delayed) using a 7-day dextran sulfate sodium (DSS)-induced mouse model of colitis. The effects were evaluated based on colon length, disease activity index (DAI) and histological score. Cytokine-encoding mRNA levels T cells and macrophages were evaluated by real-time PCR and flow cytometry. Regarding the timing of administration, early (day 3) injection significantly ameliorated DSS-induced colitis in terms of both DAI and histological score, compared to those parameters with delayed (day 11) injection. With early cell injection, the tissue mRNA levels of anti-inflammatory cytokine genes (Il10, Tgfb) increased, whereas those of inflammatory cytokine genes (Il6, Tnfa and Il17a) decreased significantly. Regarding the associated mechanism, hAdMSCs suppressed T cell proliferation and activation in vitro, increased the number of regulatory T cells in vivo and changed the polarity of macrophages (into the anti-inflammatory M2 phenotype) in vitro. Timing of injection is critical for the effective therapeutic effects of hAdMSCs. Furthermore, part of the associated mechanism includes T cell activation and expansion and altered macrophage polarization.
Article
Full-text available
Background We previously reported that the incidence of hepatocellular carcinoma (HCC) with non-viral etiologies increased rapidly between 1991 and 2010 in Japan. Methods To update this investigation, we enrolled patients who were initially diagnosed as having non-B, non-C HCC at participating hospitals between 2011 and 2015. In addition to the patient characteristics investigated in the previous report, we also investigated the duration of alcohol consumption. The overall survival rate was analyzed using the Kaplan–Meier method, and the hazard function against the body mass index (BMI) was plotted using cubic splines. Results A total of 2087 patients were enrolled. The proportion of patients with non-viral etiologies has continued to increase from 10.0% in 1991 to 32.5% in 2015. Patients were also older (median ages, 70–73 years) and more obese (median BMIs, 23.9–24.2 kg/m²), and the proportions of patients with diabetes mellitus (46.1% to 51.6%), hypertension (42.7% to 58.6%), dyslipidemia (14.6% to 22.9%), and fatty liver (24.0% to 28.8%) had all increased significantly. There was a significant inverse relationship between the duration and the amount of daily alcohol consumption. The improvement in the overall survival was relatively small, with a decreased proportion of patients under surveillance (41.3% to 31.6%). A hazard function plot showed a curve similar to that in our previous report, with a lowest hazard of ~ 26 kg/m². Conclusions The proportion of HCC patients with non-viral etiologies continues to increase in Japan. Lifetime total amount of alcohol consumption may be a risk factor.
Article
Full-text available
Abstract Mesenchymal stem cells (MSCs) are adult stromal cells with the capacity to differentiate into multiple types of cells. MSCs represent an attractive option in regenerative medicine due to their multifaceted abilities for tissue repair, immunosuppression, and anti-inflammation. Recent studies demonstrate that MSCs exert their effects via paracrine activity, which is at least partially mediated by extracellular vesicles (EVs). MSC-derived EVs (MSC-EVs) could mimic the function of parental MSCs by transferring their components such as DNA, proteins/peptides, mRNA, microRNA (miRNA), lipids, and organelles to recipient cells. In this review, we aim to summarize the mechanism and role of miRNA transfer in mediating the effects of MSC-EVs in the models of human diseases. The first three sections of the review discuss the sorting of miRNAs into EVs, uptake of EVs by target cells, and functional transfer of miRNAs via EVs. Then, we describe the composition of miRNAs in MSC-EVs. Next, we provide the existing evidence that MSC-EVs affect the outcomes of renal, liver, heart, and brain diseases by transferring their miRNA contents. In conclusion, EV-mediated miRNA transfer plays an important role in disease-modulating capacity of MSCs.
Article
Full-text available
We describe a novel therapeutic approach for cirrhosis using mesenchymal stem cells (MSCs) and colony-stimulating factor-1-induced bone marrow-derived macrophages (id-BMMs) and analyze the mechanisms underlying fibrosis improvement and regeneration. Mouse MSCs and id-BMMs were cultured from mouse bone marrow and their interactions analyzed in vitro. MSCs, id-BMMs, and a combination therapy using MSCs and id-BMMs were administered to mice with CCl4 -induced cirrhosis. Fibrosis regression, liver regeneration, and liver-migrating host cells were evaluated. Administered cell behavior was also tracked by intravital imaging. In coculture, MSCs induced switching of id-BMMs toward the M2 phenotype with high phagocytic activity. In vivo, the combination therapy reduced liver fibrosis (associated with increased matrix metalloproteinases expression), increased hepatocyte proliferation (associated with increased hepatocyte growth factor, vascular endothelial growth factor, and oncostatin M in the liver), and reduced blood levels of liver enzymes, more effectively than MSCs or id-BMMs monotherapy. Intravital imaging showed that after combination cell administration, a large number of id-BMMs, which phagocytosed hepatocyte debris and were retained in the liver for more than 7 days, along with a few MSCs, the majority of which were trapped in the lung, migrated to the fibrotic area in the liver. Host macrophages and neutrophils infiltrated after combination therapy and contributed to liver fibrosis regression and promoted regeneration along with administered cells. Indirect effector MSCs and direct effector id-BMMs synergistically improved cirrhosis along with host cells in mice. These studies pave the way for new treatments for cirrhosis. Stem Cells Translational Medicine 2018.
Article
Full-text available
Background: Liver disease is a major cause of death and disability. Mesenchymal stem cells (MSCs) show promise for the treatment of liver disease. However, whether MSC-based therapy is more effective than conventional treatment is unclear, as are the optimal MSC source, the administration frequency, and the most effective MSC delivery route. We therefore undertook a systematic review and meta-analysis of the therapeutic efficacy of MSCs against liver disease and the related factors. Methods: We systematically searched Medline (PubMed), Cochrane Library, EMBASE, ClinicalTrials.gov, and SinoMed CBM to identify studies published up to June 2017 involving liver disease patients receiving MSC-based therapy and which reported estimates of liver function during the follow-up period. Results: Thirty-nine studies were selected from 672 publications. According to a meta-analysis of 23 controlled studies, compared with conventional treatment MSC therapy significantly improves liver function in patients with liver disease in terms of the model of end-stage liver disease score, albumin, alanine aminotransferase, and total bilirubin levels, and prothrombin time, up to 6 months after administration. However, it has no beneficial effects in terms of prothrombin activity, international normalized ratio, or cholinesterase level. Considerable heterogeneity was identified at most time points. Subgroup analyses showed that a single MSC injection was more effective than multiple injections, MSC administration was more effective via the hepatic artery than the peripheral vein, and MSCs derived from bone marrow were more effective than those derived from the umbilical cord. Conclusions: MSC-based therapy is relatively safe and improves liver function during the first 6 months after administration. A single injection administration via the hepatic artery and MSCs derived from bone marrow are optimal in terms of improving liver function. However the significant heterogeneity among studies and discontinuous results of the subgroup meta-analysis should be addressed; moreover the long-term efficacy of MSC therapy warrants further investigation.
Article
Full-text available
Background: Mesenchymal stromal cells (MSCs) are an attractive therapeutic agent in regenerative medicine. Recently, there has been a paradigm shift from differentiation of MSCs to their paracrine effects at the injury site. Several reports elucidate the role of trophic factors secreted by MSCs toward the repair of injured tissues. We hypothesize that fractionating the MSC secretome will enrich exosomes containing soluble bioactive molecules, improving its therapeutic potential for liver failure. Methods: Rat bone marrow MSCs were isolated and the conditioned media filtered, concentrated and ultracentrifuged to generate fractionated secretome. This secretome was characterized for the presence of exosomes and recovery from liver injury assessed in in-vitro liver injury models. The results were further validated in vivo. Results: Studies on in-vitro liver injury models using acetaminophen and hydrogen peroxide show better cell recovery and reduced cytotoxicity in the presence of fractionated as opposed to unfractionated secretome. Further, the cells showed reduced oxidative stress in the presence of fractionated secretome, suggesting a potential antioxidative effect. These results were further validated in vivo in liver failure models, wherein improved liver regeneration in the presence of fractionated secretome (0.819 ± 0.035) was observed as compared to unfractionated secretome (0.718 ± 0.042). Conclusions: The work presented is a proof of concept that fractionating the secretome enriches certain bioactive molecules involved in the repair and recovery of injured liver tissue. Exosome enriched mesenchymal stromal cell-derived fractionated secretome potentiates recovery upon injection in injured liver.
Article
Human umbilical cord mesenchymal stem cell-derived exosomes (hUCMSC-EXOs) play an important role in the regulation of the immune system and inflammatory responses; however, their role in acute liver failure (ALF) and related pathological conditions is unclear. In this study, we found that hUCMSC-EXOs can reduce the expression of the NLRP3 inflammasome and downstream inflammatory factors in acute liver failure. Western blot and ELISA results showed that hUCMSC-EXOs decreased the expression of NLRP3, caspase-1, IL-1β and IL-6 in LPS-stimulated RAW 264.7 macrophages. In vivo, the hUCMSC-EXOs repaired damaged liver tissue and decreased the expression of the NLRP3 inflammasome and the levels of ALT and AST in a mouse ALF model. The results of this study provide a new strategy for the application of human umbilical cord mesenchymal stem cell-derived exosomes in the treatment of ALF.
Article
Importance More than 240 million individuals worldwide are infected with chronic hepatitis B virus (HBV). Among individuals with chronic HBV infection who are untreated, 15% to 40% progress to cirrhosis, which may lead to liver failure and liver cancer. Observations Pegylated interferon and nucleos(t)ide analogues (lamivudine, adefovir, entecavir, tenofovir disoproxil, and tenofovir alafenamide) suppress HBV DNA replication and improve liver inflammation and fibrosis. Long-term viral suppression is associated with regression of liver fibrosis and reduced risk of hepatocellular carcinoma in cohort studies. The cure (defined as hepatitis B surface antigen loss with undetectable HBV DNA) rates after treatment remain low (3%-7% with pegylated interferon and 1%-12% with nucleos[t]ide analogue therapy). Pegylated interferon therapy can be completed in 48 weeks and is not associated with the development of resistance; however, its use is limited by poor tolerability and adverse effects such as bone marrow suppression and exacerbation of existing neuropsychiatric symptoms such as depression. Newer agents (entecavir, tenofovir disoproxil, and tenofovir alafenamide) may be associated with a significantly reduced risk of drug resistance compared with older agents (lamivudine and adefovir) and should be considered as the first-line treatment. Conclusions and Relevance Antiviral treatment with either pegylated interferon or a nucleos(t)ide analogue (lamivudine, adefovir, entecavir, tenofovir disoproxil, or tenofovir alafenamide) should be offered to patients with chronic HBV infection and liver inflammation in an effort to reduce progression of liver disease. Nucleos(t)ide analogues should be considered as first-line therapy. Because cure rates are low, most patients will require therapy indefinitely.
Article
Extracellular vesicles (EVs) are endogenous nanoparticles that play important roles in intercellular communication. Unmodified and engineered EVs can be utilized for therapeutic purposes. For instance, mesenchymal stem cell (MSC)-derived EVs have shown promise for tissue repair, while drug-loaded EVs have the potential to be used for cancer treatment. The liver is an ideal target for EV therapy due to the intrinsic regenerative capacity of hepatic tissue and the tropism of systemically injected nanovesicles for this organ. This review will give an overview of the potential of EV therapeutics in liver disease. Specifically, the mechanisms by which MSC-EVs induce liver repair will be covered. Moreover, the use of drug-loaded EVs for the treatment of hepatocellular carcinoma will also be discussed. Although there are several challenges associated with the clinical translation of EVs, these biological nanoparticles represent a promising new therapeutic modality for liver disease.
Article
To establish diagnostic criteria for acute-on-chronic liver failure (ACLF) in Japan, the Intractable Hepato-Biliary Disease Study Group of Japan performed a multicenter pilot survey for patients fulfilling the APASL, EASL-Clif Consortium, or Chinese Medical Association (CMA) diagnostic criteria for ACLF and found that the APASL criteria were suitable for screening Japanese patients with ACLF when patients whose conditions were triggered by gastrointestinal bleeding were included within the disease entity, while the EASL-Clif Consortium criteria were useful for classifying the severity of the patients’ conditions. Based on these observations, the Study Group proposed the following diagnostic criteria for ACLF in Japan: patients with cirrhosis and a Child-Pugh score of 5-9 should be diagnosed as having ACLF when a deterioration of liver function (serum bilirubin level of 5.0 mg/dL or more and prothrombin time value of 40% or less of the standardized values and/or international normalization rates [INRs] of 1.5 or more) caused by severe liver damage develops within 28 days after acute insults, such as alcohol abuse, bacterial infection, gastrointestinal bleeding, or the exacerbation of underlying liver diseases. The severities of the patients can be classified into 4 grades depending on the extent of the deterioration in organ functions, including kidney, cerebral, blood coagulation, circulatory and respiratory functions as well as liver function. The usefulness of these novel criteria should be validated prospectively in a largescale cohort in the future.