32
Accepted Manuscript Historical Perspectives and Advances in the Mesenchymal Stem Cell Research for the Treatment of Liver Diseases Chien-Wei Lee, Yu-Fan Chen, Hao-Hsiang Wu, Oscar K. Lee PII: S0016-5085(17)36280-7 DOI: 10.1053/j.gastro.2017.09.049 Reference: YGAST 61495 To appear in: Gastroenterology Accepted Date: 27 September 2017 Please cite this article as: Lee C-W, Chen Y-F, Wu H-H, Lee OK, Historical Perspectives and Advances in the Mesenchymal Stem Cell Research for the Treatment of Liver Diseases, Gastroenterology (2017), doi: 10.1053/j.gastro.2017.09.049. This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Historical Perspectives and Advances in Mesenchymal Stem ...download.xuebalib.com/4edru0NJdOQ6.pdf · reactivity, and tumorigenic potential. We review directions and advances in the

  • Upload
    others

  • View
    4

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Historical Perspectives and Advances in Mesenchymal Stem ...download.xuebalib.com/4edru0NJdOQ6.pdf · reactivity, and tumorigenic potential. We review directions and advances in the

Accepted Manuscript

Historical Perspectives and Advances in the Mesenchymal Stem Cell Research forthe Treatment of Liver Diseases

Chien-Wei Lee, Yu-Fan Chen, Hao-Hsiang Wu, Oscar K. Lee

PII: S0016-5085(17)36280-7DOI: 10.1053/j.gastro.2017.09.049Reference: YGAST 61495

To appear in: GastroenterologyAccepted Date: 27 September 2017

Please cite this article as: Lee C-W, Chen Y-F, Wu H-H, Lee OK, Historical Perspectives and Advancesin the Mesenchymal Stem Cell Research for the Treatment of Liver Diseases, Gastroenterology (2017),doi: 10.1053/j.gastro.2017.09.049.

This is a PDF file of an unedited manuscript that has been accepted for publication. As a service toour customers we are providing this early version of the manuscript. The manuscript will undergocopyediting, typesetting, and review of the resulting proof before it is published in its final form. Pleasenote that during the production process errors may be discovered which could affect the content, and alllegal disclaimers that apply to the journal pertain.

Page 2: Historical Perspectives and Advances in Mesenchymal Stem ...download.xuebalib.com/4edru0NJdOQ6.pdf · reactivity, and tumorigenic potential. We review directions and advances in the

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

1

Historical Perspectives and Advances in the Mesenchymal Stem Cell Research for the

Treatment of Liver Diseases

Short Title: Stem Cell Therapy for Liver Diseases

Chien-Wei Lee1,2,#

, Yu-Fan Chen2,3,#

, Hao-Hsiang Wu2,4,#

and Oscar K. Lee2,3,5,6,*

1Program in Molecular Medicine, National Yang-Ming University and Academia Sinica, Taipei,

Taiwan;

2Stem Cell Research Center, National Yang-Ming University, Taipei, Taiwan;

3Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan;

4Institute of Biophotonics, National Yang-Ming University, Taipei, Taiwan;

5Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan;

6Taipei City Hospital, Taipei, Taiwan.

#These authors contributed equally to this manuscript

Grant Support: The authors acknowledge financial support from the Ministry of Science and

Technology, Taiwan (MOST103-2314-B-010-053-MY3, MOST 106-2321-B-010-008, MOST

106-2911-I-010-502 and MOST 106-3114-B-010-002). This study was also supported by

Aiming for the Top University Plan, a grant from Ministry of Education.

Abbreviations: BAL, bioartificial liver system; GVHD, graft-versus-host disease; HESC, human

embryonic stem cell; HGF, hepatocyte growth factor; HLC, hepatocyte-like cell; HPSC, human

pluripotent stem cell; HSC, hepatic stellate cell; IDO, indoleamine 2,3-dioxygenase; iPSCs,

induced pluripotent stem cells; MSC, mesenchymal stem cell; PSC, pluripotent stem cell

*Correspondence: Oscar K. Lee, MD, PhD

Page 3: Historical Perspectives and Advances in Mesenchymal Stem ...download.xuebalib.com/4edru0NJdOQ6.pdf · reactivity, and tumorigenic potential. We review directions and advances in the

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

2

Taipei City Hospital, No. 145, Zhengzhou Road, Datong District, Taipei 10341, Taiwan

Tel.: +886-2-2559-6131; Fax: +886-2-2559-9051; E-mail: [email protected]

Conflicts of Interest: The authors disclose no conflicts.

Author Contributions: Chien-Wei Lee, Yu-Fan Chen, and Hao-Hsiang Wu wrote the

manuscript; Oscar K. Lee provided feedback and edited the manuscript; Oscar K. Lee

supervised the literature search.

Page 4: Historical Perspectives and Advances in Mesenchymal Stem ...download.xuebalib.com/4edru0NJdOQ6.pdf · reactivity, and tumorigenic potential. We review directions and advances in the

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

3

Abstract

Liver transplantation is the only effective therapy for patients with decompensated cirrhosis

and fulminant liver failure. However, due to a shortage of donor livers and complications

associated with immune suppression, there is an urgent need for new therapeutic strategies

for patients with end-stage liver diseases. Given their unique function in self-renewal and

differentiation potential, stem cells might be used to regenerate damaged liver tissue.

Recent studies have shown that stem cell-based therapies can improve liver function in a

mouse model of hepatic failure. Moreover, acellular liver scaffolds, seeded with hepatocytes,

produced functional bioengineered livers for organ transplantation in pre-clinical studies.

The therapeutic potential of stem cells or their differentiated progenies will depend on their

capacity to differentiate into mature and functional cell types after transplantation. It will

also be important to devise methods to overcome their genomic instability, immune

reactivity, and tumorigenic potential. We review directions and advances in the use of

mesenchymal stem cells and their derived hepatocytes for liver regeneration. We also

discuss the potential applications of hepatocytes derived from human pluripotent stem cells

and challenges to using these cells in treating end-stage liver disease.

Keywords: Stem Cell Therapy; Pluripotent Stem Cells; Hepatocytes; Regenerative Medicine

Page 5: Historical Perspectives and Advances in Mesenchymal Stem ...download.xuebalib.com/4edru0NJdOQ6.pdf · reactivity, and tumorigenic potential. We review directions and advances in the

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

4

Despite the success of transplantation of hematopoietic stem cells, few other cell-based

therapies have been successfully translated into clinical application. The dearth of cell-based

therapies is unfortunate as the potential and demand for such therapies are apparent. One

field with exciting developments in cell-based therapies is the field of liver diseases.

Acute liver failure, end-stage liver diseases, and inherited metabolic disorders usually

lead to morbidity and mortality, and are associated with a variety of etiologies (toxic injury,

viral infections, and autoimmune and genetic disorders). In the United States, approximately

30,000 deaths each year are caused by chronic liver disease; the incidence of such deaths

increases by 3 % each year.1 The only curative treatment for end-stage liver disease is liver

transplantation. However, there are more than 15,000 patients on the wait list for a liver in

the United States—approximately 50% of them will not receive a transplant. Liver disease is

the leading cause of premature death in the United Kingdom, resulting in the loss of a

greater number of life-years than many other causes.2 Liver transplantation, the best

treatment option, is limited by high cost and shortage of donor organs. Moreover,

long-term survival is hampered by the graft vs host disease (GVHD) and side effects of

life-long immunosuppression.3

Transplantation of hepatocytes has been considered as an alternative to

transplantation of organs, but faces multiple problems, including the shortage of high

quantity cell sources, challenges to expansion in vitro, concerns of allogeneic rejection and

xenozoonosis, and the fact that hepatocytes spontaneously and quickly lose hepatic

characteristics in vitro.4-6

Stem cells, including induced pluripotent stem cells (iPSCs),

mesenchymal stem cells (MSCs), fetal liver stem cells, fetal biliary tree stem cells,

chemically-induced liver progenitors, and bone marrow CD45+ cells, could provide

alternatives to organ and hepatocyte transplantation, and may offer solutions to the

above-mentioned problems.7, 8

Among them, MSCs have been investigated in detail and

hold great promise for clinical application, as a personalized cell therapy. This is because

Page 6: Historical Perspectives and Advances in Mesenchymal Stem ...download.xuebalib.com/4edru0NJdOQ6.pdf · reactivity, and tumorigenic potential. We review directions and advances in the

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

5

they can be conveniently isolated and expanded in culture, are immune modulatory, and

allogeneic cells to not pose a xenozoonosis risk. MSCs also provide trophic support, have the

potential to differentiate into hepatocyte-like cells (HLCs), and there are no ethical issues

are associated with their use.9-11

We review the role of MSCs in HLC generation and liver

regenerative medicine, and to point to new technologies for clinical application.

In Treatment of Liver Disease

MSCs were first isolated from the bone marrow in 1968, by Friedenstein et al.12

These cells

can be also isolated from other adult somatic tissues, such as the adipose tissue and

umbilical cord blood. Given the self-renewal abilities of MSC, and their therapeutic potential

in tissue engineering and regenerative medicine, MSCs are considered as an ideal cell source

for transplantation. The safety and feasibility of MSC-based therapies have been

demonstrated in clinical trials for many diseases, including GVHD, heart diseases,

osteoarthritis, bone and cartilage injuries, diabetes and its complications, cancer, spinal cord

injury, multiple sclerosis, liver cirrhosis, respiratory disorders, Crohn’s disease, autoimmune

diseases, and others.13

MSCs can localize to damaged liver and eliminate liver fibrosis. Migration of MSCs is

facilitated by release of several molecules from the damaged liver that interact with

different receptors expressed by the MSCs.14-16

MSCs might be used to treat different types

of liver disease because they trans-differentiate in vitro and MSC-derived HLCs can

substitute for injured hepatocytes to restore liver function, have immunosuppressive and

anti-inflammatory effects, promote survival of resident hepatocytes, and direct

differentiation in vivo (Figure 1).

Hepatocyte Differentiation

Transplanted MSC-derived HLCs increase hepatic function at the engraftment site in models

Page 7: Historical Perspectives and Advances in Mesenchymal Stem ...download.xuebalib.com/4edru0NJdOQ6.pdf · reactivity, and tumorigenic potential. We review directions and advances in the

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

6

of acute and chronic liver failure. In 2004, Shu et al17

demonstrated the differentiation of rat

bone marrow MSCs into albumin- and cytokeratin 18-expressing HLCs. In the same year, we

reported a 2-step protocol for the induction of HLCs from human bone marrow MSCs—these

cells have hepatocyte-specific gene expression patterns and hepatocyte-specific functions,

including urea and glycogen production.18

Based on these findings, successful differentiation

of MSCs from other tissue origin into HLCs was also reported. HLCs have been derived from

umbilical cord blood and bone marrow-derived MSCs.19, 20

Furthermore, adipose

tissue-derived MSCs were also successfully induced to differentiate into HLCs.21

In the last decade, different groups established several hepatic differentiation protocols

for MSCs (summarized in Table 1). MSCs are more efficiently induced into HLCs in mice via a

2-step hepatic differentiation protocol18,21

than by the hepatic differentiation protocol

described by Shu et al.17

Transplantation of MSC-derived hepatocytes can restore liver

function to mice with carbon tetrachloride (CCl4)-induced liver failure.22

Co-culture of MSCs

with primary liver cells induces differentiation of MSCs into HLCs 23

. MSC-derived HLCs,

formed by co-culture with primary hepatocytes, integrate into injured liver and reduce

fibrosis 24

. Incubation of MSCs with injured liver tissue was reported to increase expression

of hepatocyte-specific genes in MSCs.25

These observations provide evidence for alternative

methods of inducing differentiation of MSCs into HLCs, via interactions with liver cells, and

the possibility of tissue integration after stem cell transplantation.

Small Inhibitors in Hepatocyte Differentiation

Differentiation of MSCs into hepatocytes involves in the mesenchymal to epithelial transition.

An inhibitor of RAC1 (NSC23766) maintains the epithelial morphology of MSCs, accelerates

upregulation of hepatocyte marker genes, and induces hepatocyte cell functions, by altering

actin polymerization during differentiation.26

Chromatin-remodeling agents, such as valproic

acid, 5-aza-2'-deoxycytidine, and trichostatin A, promote differentiation of MSCs into

Page 8: Historical Perspectives and Advances in Mesenchymal Stem ...download.xuebalib.com/4edru0NJdOQ6.pdf · reactivity, and tumorigenic potential. We review directions and advances in the

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

7

hepatocytes, indicating epigenetic regulation of this transition. hepatocytes.27-29

Manipulating hepatocyte differentiation of MSCs, via intracellular and extracellular factors,

might promote hepatic maturation, accelerate cell differentiation, or increase proliferation.

Mechanical Forces and Microenvironment Architecture

Mechanical forces and the architecture of the microenvironment affect differentiation

of MSCs into hepatocytes. Continuous flow of fluid over microfluidic chips increases the

ability of MSCs to uptake low-density lipoprotein as they differentiate into hepatocytes 30

.

Low shear stress increases differentiation of MSCs into hepatocytes.31

To mimic the

3-dimensional (3D) microenvironment and architecture of the liver, some groups fabricated

artificial 3D scaffolds and investigated whether they induce differentiation of MSCs more

efficiently than 2D culture. MSCs were reported to increase differentiation into hepatocytes

on poly-ε-caprolactone/collagen/polyethersulfone nanofiber scaffolds 32

. Poly-L-lactic acid

nanofiber scaffolds were also reported to increase the expression of hepatocyte-specific

genes by differentiating MSCs.33

Interestingly, during the differentiation of MSCs into

hepatocytes on artificial 3D scaffolds, MSCs form hepatic spheroids that express markers of

hepatocytes and undergo glycogen storage, urea production, albumin secretion, and

cytochrome P450 activity.34

Despite reports indicating the potential of MSCs to differentiate into hepatocytes,

MSC-derived HLCs are similar to fetal hepatocytes and do not reach full maturity in vitro 33

.

Moreover, different types of stem cell sources for the induction of HLCs have been reported;

these were generated using a modified differentiation protocol.35

Therefore, the maturation

process of MSCs during hepatic differentiation requires improvement.

Anti-inflammatory Effects

The liver is an immunologically complex organ. It contains specialized immune cells,

Page 9: Historical Perspectives and Advances in Mesenchymal Stem ...download.xuebalib.com/4edru0NJdOQ6.pdf · reactivity, and tumorigenic potential. We review directions and advances in the

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

8

including Kupffer cells; antigen-presenting liver sinusoidal endothelial cells and dendritic

cells; and liver-resident lymphocytes.36

In addition, the hepatocytes and hepatic stellate cells

(HSCs) affect the immune response in the liver by activating T cells.37-39

The liver

microenvironment and immune response maintain liver homeostasis.

Inflammation is one of the most characteristic features of different liver diseases and is

present at all disease stages.40

MSCs modulate innate and adaptive immune responses.41

MSC-derived prostaglandin E2 and indoleamine 2,3-dioxygenase (IDO) reduces proliferation,

cytolytic activity, and cytokine secretion of natural killer cells.42

MSCs also repress the

activity of Kupffer cells, reducing secretion of the inflammatory cytokine tumor necrosis

factor;43

they also convert M1 macrophages (inflammatory) to M2 macrophages

(anti-inflammatory), by secreting prostaglandin E2.44

MSC-derived human leukocyte antigen

G inactivates dendritic cells;45

MSC-derived interleukin 6 (IL6) and hepatocyte growth factor

(HGF) inhibit the maturation of monocytes into dendritic cells, reducing their inflammatory

potential and changing activated CD4+ T cells from T-helper 1 to T-helper 2 phenotypes.

46

MSCs also inhibit proliferation of CD8+ T cells and increase the rate of conversion of CD4+ T

cells from T-helper 1 to T-helper 2 phenotypes by secreting heme oxygenase 1 and IDO.47-50

Furthermore, MSCs induce the development of regulatory T cells to modulate CD4+ T cell

polarization.51

MSCs directly inhibit B cell proliferation and terminal differentiation through soluble

factors and cell contacts.52

MSCs also act as immunosuppressors following liver

transplantation in mice.53

Only a few studies reported the use of immunosuppression during

liver transplantation but they indicated that liver transplantation with MSC infusion might

prevent acute rejection by regulating T-cell expansion and increasing serum levels of

IL10.54-57

MSC-derived IDO increases survival of patients with refractory chronic GVHD by

stimulating CD5+ regulatory B cells to produce IL10; this is associated with reduced

inflammatory cytokine production by T cells.58

Collectively, MSCs convert the overall

Page 10: Historical Perspectives and Advances in Mesenchymal Stem ...download.xuebalib.com/4edru0NJdOQ6.pdf · reactivity, and tumorigenic potential. We review directions and advances in the

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

9

immune response to less inflammatory (or more tolerant) response by altering the

secretome of the immune cells.59

Most of the immunomodulatory features of MSCs are attributed to their secretome and

proteome. However, the spectrum of secretory factors of MSCs differs from that of

MSCs-derived hepatocyte-like cells and hepatocytes.27, 60

Although immunoregulation by

MSCs participates in liver repair and liver transplantation43, 54, 61

, the immunomodulatory

features of MSCs are typically reported in other organs rather than in the liver. Therefore,

further investigations are indispensable to determine whether immunomodulation by MSCs

is crucial in liver regeneration.

Anti-fibrotic Effects

Clinical chronic liver inflammation caused by viral infection, alcoholic liver disease,

nonalcoholic steatohepatitis, and autoimmune diseases results in liver fibrosis and

cirrhosis.62

Hepatic inflammation activates HSCs to produce extracellular matrix; this

precedes and promotes progression of liver fibrosis. MSCs reduce or reverse liver fibrosis via

their immunomodulatory and anti-inflammatory effects.63, 64

On the other hand, MSCs

directly inactivate HSCs by secreting IL10 and tumor necrosis factor, and induce HSC

apoptosis through HGF, nerve growth factor signaling via p75, and Fas–FasL pathway.65

MSCs

might therefore be used in treatment of hepatic fibrosis.65

MSCs can degrade ECM directly,

by secreting matrix metalloproteinases,66

or indirectly, by stimulating the immune cells to

increase the production of these enzymes.67

MSCs were shown to reduce liver fibrosis in

mice by secreting cytokines and matrix metalloproteinases.68-72

DLK1 contributes to HSC

activation; MSC transplantation reduced activation of HSCs by DLK1 and liver fibrosis.73

Liver Regeneration and Direct MSC Differentiation In Vivo

The immunomodulatory activities and anti-fibrotic effects of MSCs are important for liver

Page 11: Historical Perspectives and Advances in Mesenchymal Stem ...download.xuebalib.com/4edru0NJdOQ6.pdf · reactivity, and tumorigenic potential. We review directions and advances in the

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

10

regeneration. MSCs are also directly involved in maintaining the hepatocyte pool, by

repressing hepatocyte apoptosis and stimulating hepatocyte proliferation, and by secreting

factors such as HGF that maintain liver function.74

Christ et al. reported an in vitro

differentiation protocol for generating hepatocytes from bone marrow and adipose tissue

MSC.75

They showed that MSC-derived hepatocyte-like cells engraft more efficiently than

undifferentiated cells.76

In addition, transplantation of MSCs, MSC-derived brown adipocytes,

or MSC lysates into obese mice reduced fat accumulation and hepatocyte lipotoxicity,

restoring liver functions.77, 78

Although rare, transplanted MSCs can spontaneously

differentiate into hepatocytes in rat79

and this may also, at least partially, contribute to liver

regeneration.

MSC-based Treatments

MSC-derived microvesicles

Microvesicles are extracellular vesicles that contain exosomes and microparticles. Exosomes

are small extracellular vesicles (30–120 nm in diameter) derived from budding membranes

of multi-vesicular bodies; microparticles are large cell-derived vesicles (100 nm to 1 µm)

derived from budding of the cell membrane. Microvesicles can interact with local or distal

cells and transfer molecules such as RNAs, microRNAs (miRNAs), and proteins, for

intercellular communication through a paracrine effect.

Stem cell-derived exosomes and micro-vesicles have been tested in models of liver

diseases.80-84

Injection of MSC-derived exosomes reduced fibrosis in livers of mice following

CCl4 injections, by suppressing expression of collagen and tumor growth factor beta 1.85

MSC-derived exosomes upregulate expression of priming-phase genes during liver

regeneration, leading to increased number of proliferating cell nuclear antigen-positive cells

and levels of cyclin D1 in livers of mice with CCl4-induced fibrosis. MSC-derived exosomes

also inhibit acetaminophen- and H2O2-induced apoptosis of hepatocytes.86

Page 12: Historical Perspectives and Advances in Mesenchymal Stem ...download.xuebalib.com/4edru0NJdOQ6.pdf · reactivity, and tumorigenic potential. We review directions and advances in the

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

11

Exosome miRNAs contribute to development of liver disease in animal models. 87

Exosome miRNA125b, derived from MSCs, reduces activation of the hedgehog pathway in

HSCs, by reducing levels of Smo expression and subsequent CCl4-induced liver fibrosis in

rat.88

MSC-derived exosomes also increase expression of anti-inflammatory cytokines and

the proportion of T-regulatory cells in mice with concanavalin A-induced liver injury,

supporting their immunosuppressive effects.89

MSC-derived exosomes might therefore be

developed for treatment of liver diseases, but additional studies are needed to elucidate

mechanisms of MSC-derived exosomes in liver regeneration. Studies with large animal

models are necessary to translate the findings into clinical practice.

Bioartificial liver (BAL) systems

In 1956, researchers 90

demonstrated that a fresh bovine liver homogenate metabolizes

salicylic acids and ketone bodies, and produces urea from ammonium chloride. In 1967,

some case reports indicated that cross-circulation and exchange transfusion supported livers

of patients in acute comas.91

Accordingly, BAL was developed as a therapy of acute liver

failures.

BAL systems are extracorporeal systems consisting of a bioreactor and biocomponents

(hepatocytes), connected to patient circulation and supporting liver functions. Many studies

indicate that BAL systems improve the survival in animal models.92-94

Matsumura et al

reported clinical application of a BAL system in 1987.95

Eleven BAL device systems are now

available. Among them, the Extracorporeal Liver Assist Device and the HepatAssist are being

tested in clinical trials.96, 97

Since human hepatocyte sources are limited, stem cell-derived HLCs might be used as

substitutes in the BAL system. A BAL system with human fetal fibroblast-derived HLCs was

reported to prolong survival of pigs with acute liver failure in 2016.98

Highly functional HLCs

have been generated from human pluripotent stem cells .99, 100

Human pluripotent stem cell

Page 13: Historical Perspectives and Advances in Mesenchymal Stem ...download.xuebalib.com/4edru0NJdOQ6.pdf · reactivity, and tumorigenic potential. We review directions and advances in the

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

12

(HPSC)-derived HLCs are an ideal cell source for a clinical-grade BAL system. BAL systems

might be developed as alternative strategies for patients with acute liver failure. The use of

BAL systems could also extend the wait time for a suitable liver donor for patients with

end-stage liver disease.

Organ bioengineering

Organ bioengineering involving decellularized organs has recently enabled the development

of a bone fide ECM architecture for stem cell differentiation. Researchers have induced

hepatic differentiation of MSCs on ECM sheets derived from decellularized liver, and

demonstrated efficient transplantation of these cells into mice with CCl4-induced liver

failure101

. Direct infusion of MSCs into de-cellularized liver could be more efficient in

induction of hepatic differentiation.102

Loss of biliary structure or impaired blood supply in

donor livers suppresses liver regeneration after transplantation 103

, so re-cellularization of

liver tissues with a combination of liver sinusoidal endothelial cells, bile duct cells, and

hepatocytes is essential. This strategy might be used in transplantation.

Although the use of MSC-based therapy and recellularized liver tissue is a more

cost-and time-effective option than whole-organ transplantation, the recelluarization

protocol should be optimized and the limitations need to be addressed before future clinical

applications.104, 105

These include, the route of administration or recellularization (e.g., direct

injection or continuous perfusion at a physiological pressure), potential immunogenicity, lack

of hepatic non-parenchymal cells, the possibility of small-for-size syndrome, and differences

in the vascular infrastructure of recellularized and normal liver tissues.106-108

Trials of MSC-Based Therapies

Anti-inflammatory and anti-fibrotic effects of MSCs have been observed in animal models,109,

110so several clinical trials of human MSCs had been conducted in patients with liver

Page 14: Historical Perspectives and Advances in Mesenchymal Stem ...download.xuebalib.com/4edru0NJdOQ6.pdf · reactivity, and tumorigenic potential. We review directions and advances in the

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

13

fibrosis.7, 111-114

To evaluate MSC transplantation in patients, serum creatinine and bilirubin

are used as a therapeutic index in patients with advanced liver fibrosis.115, 116

Evaluation of

the endpoints in these studies revealed the safety and efficacy of human MSC

transplantation. Jang et al reported a reduction of collagen deposition in the histological

analyses of the liver from patients after MSC transplantation in 2014 114

. In 2016, MSC

transplantation was tested in a phase 2 trial of patients with alcoholic cirrhosis.117

MSC

transplantation was reported to reduce collagen deposition and model for end-stage liver

disease scores and increased liver function in these patients. Based on the results of these

studies, we anticipate phase 3 clinical trials in the near future. Although these studies

indicate the therapeutic potential of MSC transplantation for patients with liver fibrosis,

studies are needed to determine the appropriate therapeutic window, MSC dosage, and

mechanisms of fibrosis reduction.

HPSC Administration

Advantages

In 1998, Thomson et al118

reported isolation of human embryonic stem cells (HESCs) from

blastocysts and confirmed that these cells maintained the potential to differentiate into all 3

embryonic germ layers. These cells became powerful tools used in developmental biology,

drug discovery, and regenerative medicine because of their ability to self-renew indefinitely

and their ability to differentiate into any cell lineage. Despite their great medical potential,

there was controversy over use of HESCs, because generation of HESCs involves the

destruction of a human embryo. Another important issue is the immunogenicity of HESCs.

In 2006, Takahashi et al119

reported reprogramming of fibroblasts into iPSCs by

expression of 4 transcription factors (OCT3/4, SOX2, KLF4, and c-Myc). The generation of

human iPSCs was reported by same group in 2007.120

iPSC generation does not involve

destruction of human embryos, and these cells can be generated in an autologous,

Page 15: Historical Perspectives and Advances in Mesenchymal Stem ...download.xuebalib.com/4edru0NJdOQ6.pdf · reactivity, and tumorigenic potential. We review directions and advances in the

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

14

patient-specific manner, and differentiated into various cell types. The unlimited supply of

these cells provides the patients with an alternative autologous, rather than allogeneic,

transplantation option.

Patients with acute liver failure or chronic liver disease are ideal candidates for

cell-based transplantation. Patients with acute liver failure require an immediate restoration

of the liver function by liver or hepatocyte transplantation, but only 10% or fewer of

required liver transplantations are performed.121

As an immediately available and

inexhaustible cell source, PSC-derived hepatocytes might be used to treat these patients.

Transplantation of PSC-derived hepatocytes by a direct splenic injection improves liver

function and prolongs survival of mice with CCl4-induced liver injury.122-124

MSCs are available from the human bone marrow, fat, umbilical cord, dental pulp, and

PSCs. Among these alternatives, human PSCs with extended in vitro culture are the best

sources for MSC production.125

In addition, PSC-derived MSCs have higher telomerase

activity,125

are less sensitive to inflammatory cytokines such as interferon gamma,126

and

have stronger immunomodulation than bone marrow-derived MSCs.127

PSC-derived MSCs

therefore provide an ideal source for clinical application. Researchers have generated

decellularized liver tissues with 3D anatomical structures, including a microvascular network,

that is difficult to manufacture in vitro.128

HPSC-derived hepatic cells could therefore provide

an ideal cell source to develop a BAL system, because of their similarity to primary human

hepatocytes. By studying the liver development, scientists might identify the factors

required for direct differentiation of HPSCs into hepatocytes.129

Challenges

It is important to maintain the genomic stability of PSCs to be used in cell-based therapies.

Cultured PSCs and reprogrammed cells acquire genomic abnormalities, such as large

chromosomal aberrations,130, 131

variations in gene copy numbers,132-134

and somatic

Page 16: Historical Perspectives and Advances in Mesenchymal Stem ...download.xuebalib.com/4edru0NJdOQ6.pdf · reactivity, and tumorigenic potential. We review directions and advances in the

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

15

mutations in coding regions.135, 136

For example, shortly after derivation, 10% of PSCs contain

at least 1 large chromosomal aberration, indicating that large chromosomal aberrations

occur early in the PSC culture process. Interestingly, levels of serum response factor, a

transcription factor, were reported to be reduced in diploid HPSCs. So reduced expression of

serum response factor could contribute to chromosomal instability in these cells.137

A similar

mechanism might operate during the initiation of chromosome instability in diploid cells.

Similarly, DNA analysis studies indicated that iPSC cell lines already contain copy number

variations during early stages of passage, so copy number variations might be induced by the

reprogramming process. Genomic instability is a feature of cancer, so the maintenance of

PSC genomic stability is important.

Some challenges of using HPSCs for therapeutic purposes are linked with the availability

of a homogeneous supply of mature PSC-derived cells. Alarmingly, Zhao et al138

showed that

cells derived from iPSCs induced immune response in syngeneic mice. Providing

HPSC-derived cells for use in clinical therapies requires further, substantial development.

Nonetheless, the utility of PSC-derived cells has been demonstrated in treatment of diabetes,

liver diseases, retinal diseases, muscular dystrophies, and heart diseases.139

Future Directions

An unlimited supply of hepatocytes is needed for cell-based, regenerative treatments for

patients with liver disease. HPSCs are more suitable candidates for generation of functional

hepatocytes than other types of stem cells. Nevertheless, genomic aberrations and the need

for rapid generation of mature and functional hepatocytes challenge their clinical application.

MSCs can suppress activated immune cells, and have anti-inflammatory and anti-fibrotic

effects, so they have been studied for their effects during immune rejection following

allogeneic transplantation and liver regeneration.

Page 17: Historical Perspectives and Advances in Mesenchymal Stem ...download.xuebalib.com/4edru0NJdOQ6.pdf · reactivity, and tumorigenic potential. We review directions and advances in the

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

16

MSCs might also protect the liver from injury, by producing trophic factors (IL10 and

HGF).110

MSC transplantation has been tested, with promising results, in several clinical trials,

and is a possible alternative approach for treatment of patients with liver diseases. MSCs can

reduce collagen deposition in patients with alcoholic liver cirrhosis.114

Although studies have

found MSC transplantation to be safe in preclinical and clinical trials,140, 141

some

chromosomal aberrations have been reported in cultured cells.142-144

Long-term culture

expansion may cause genomic instability and lead to transformation of mouse MSCs,

although this is rarely observed in human MSCs.145-148

Stem cell therapies hold a great

promise for liver regenerative medicine. Mechanistic and clinical studies should foster a

closer collaboration between the academic and industry researchers, to make the most of

the therapeutic potential of MSCs.

Page 18: Historical Perspectives and Advances in Mesenchymal Stem ...download.xuebalib.com/4edru0NJdOQ6.pdf · reactivity, and tumorigenic potential. We review directions and advances in the

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

17

References :

1. Murphy SL, Xu J, Kochanek KD. Deaths: final data for 2010. Natl Vital Stat Rep

2013;61:1-117.

2. Owen A, Newsome PN. Mesenchymal stromal cell therapy in liver disease:

opportunities and lessons to be learnt? Am J Physiol Gastrointest Liver Physiol

2015;309:G791-800.

3. Londono MC, Rimola A, O'Grady J, et al. Immunosuppression minimization vs.

complete drug withdrawal in liver transplantation. J Hepatol 2013;59:872-9.

4. Chen Y, Wong PP, Sjeklocha L, et al. Mature hepatocytes exhibit unexpected plasticity

by direct dedifferentiation into liver progenitor cells in culture. Hepatology

2012;55:563-74.

5. Huebert RC, Rakela J. Cellular therapy for liver disease. Mayo Clin Proc

2014;89:414-24.

6. Ferrer JR, Chokechanachaisakul A, Wertheim JA. New Tools in Experimental Cellular

Therapy for the Treatment of Liver Diseases. Curr Transplant Rep 2015;2:202-210.

7. Nicolas CT, Wang Y, Nyberg SL. Cell therapy in chronic liver disease. Curr Opin

Gastroenterol 2016;32:189-94.

8. Katsuda T, Kawamata M, Hagiwara K, et al. Conversion of Terminally Committed

Hepatocytes to Culturable Bipotent Progenitor Cells with Regenerative Capacity. Cell

Stem Cell 2017;20:41-55.

9. Pittenger MF, Mackay AM, Beck SC, et al. Multilineage potential of adult human

mesenchymal stem cells. Science 1999;284:143-7.

10. Makino S, Fukuda K, Miyoshi S, et al. Cardiomyocytes can be generated from marrow

stromal cells in vitro. J Clin Invest 1999;103:697-705.

11. Wislet-Gendebien S, Hans G, Leprince P, et al. Plasticity of cultured mesenchymal

stem cells: switch from nestin-positive to excitable neuron-like phenotype. Stem Cells

2005;23:392-402.

12. Friedenstein AJ, Petrakova KV, Kurolesova AI, et al. Heterotopic of bone marrow.

Analysis of precursor cells for osteogenic and hematopoietic tissues. Transplantation

1968;6:230-47.

13. Campbell A, Brieva T, Raviv L, et al. Concise Review: Process Development

Considerations for Cell Therapy. Stem Cells Transl Med 2015;4:1155-63.

14. Chen J, Li Y, Katakowski M, et al. Intravenous bone marrow stromal cell therapy

reduces apoptosis and promotes endogenous cell proliferation after stroke in female

rat. J Neurosci Res 2003;73:778-86.

15. Ruster B, Gottig S, Ludwig RJ, et al. Mesenchymal stem cells display coordinated

rolling and adhesion behavior on endothelial cells. Blood 2006;108:3938-44.

16. Ma HC, Shi XL, Ren HZ, et al. Targeted migration of mesenchymal stem cells modified

Page 19: Historical Perspectives and Advances in Mesenchymal Stem ...download.xuebalib.com/4edru0NJdOQ6.pdf · reactivity, and tumorigenic potential. We review directions and advances in the

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

18

with CXCR4 to acute failing liver improves liver regeneration. World J Gastroenterol

2014;20:14884-94.

17. Shu SN, Wei L, Wang JH, et al. Hepatic differentiation capability of rat bone

marrow-derived mesenchymal stem cells and hematopoietic stem cells. World J

Gastroenterol 2004;10:2818-22.

18. Lee KD, Kuo TK, Whang-Peng J, et al. In vitro hepatic differentiation of human

mesenchymal stem cells. Hepatology 2004;40:1275-84.

19. Lee OK, Kuo TK, Chen WM, et al. Isolation of multipotent mesenchymal stem cells

from umbilical cord blood. Blood 2004;103:1669-75.

20. Hong SH, Gang EJ, Jeong JA, et al. In vitro differentiation of human umbilical cord

blood-derived mesenchymal stem cells into hepatocyte-like cells. Biochem Biophys

Res Commun 2005;330:1153-61.

21. Banas A, Teratani T, Yamamoto Y, et al. Adipose tissue-derived mesenchymal stem

cells as a source of human hepatocytes. Hepatology 2007;46:219-28.

22. Kuo TK, Hung SP, Chuang CH, et al. Stem cell therapy for liver disease: parameters

governing the success of using bone marrow mesenchymal stem cells.

Gastroenterology 2008;134:2111-21, 2121.e1-3.

23. Qihao Z, Xigu C, Guanghui C, et al. Spheroid formation and differentiation into

hepatocyte-like cells of rat mesenchymal stem cell induced by co-culture with liver

cells. DNA Cell Biol 2007;26:497-503.

24. Li TZ, Kim JH, Cho HH, et al. Therapeutic potential of bone-marrow-derived

mesenchymal stem cells differentiated with growth-factor-free coculture method in

liver-injured rats. Tissue Eng Part A 2010;16:2649-59.

25. Mohsin S, Shams S, Ali Nasir G, et al. Enhanced hepatic differentiation of

mesenchymal stem cells after pretreatment with injured liver tissue. Differentiation

2011;81:42-8.

26. Teng NY, Liu YS, Wu HH, et al. Promotion of mesenchymal-to-epithelial transition by

Rac1 inhibition with small molecules accelerates hepatic differentiation of

mesenchymal stromal cells. Tissue Eng Part A 2015;21:1444-54.

27. Lee CW, Huang WC, Huang HD, et al. DNA Methyltransferases Modulate Hepatogenic

Lineage Plasticity of Mesenchymal Stromal Cells. Stem Cell Reports 2017;9:247-263.

28. An SY, Han J, Lim HJ, et al. Valproic acid promotes differentiation of hepatocyte-like

cells from whole human umbilical cord-derived mesenchymal stem cells. Tissue Cell

2014;46:127-35.

29. Ye D, Li T, Heraud P, et al. Effect of Chromatin-Remodeling Agents in Hepatic

Differentiation of Rat Bone Marrow-Derived Mesenchymal Stem Cells In Vitro and In

Vivo. Stem Cells Int 2016;2016:3038764.

30. Ju X, Li D, Gao N, et al. Hepatogenic differentiation of mesenchymal stem cells using

Page 20: Historical Perspectives and Advances in Mesenchymal Stem ...download.xuebalib.com/4edru0NJdOQ6.pdf · reactivity, and tumorigenic potential. We review directions and advances in the

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

19

microfluidic chips. Biotechnol J 2008;3:383-91.

31. Yen MH, Wu YY, Liu YS, et al. Efficient generation of hepatic cells from mesenchymal

stromal cells by an innovative bio-microfluidic cell culture device. Stem Cell Res Ther

2016;7:120.

32. Kazemnejad S, Allameh A, Soleimani M, et al. Biochemical and molecular

characterization of hepatocyte-like cells derived from human bone marrow

mesenchymal stem cells on a novel three-dimensional biocompatible nanofibrous

scaffold. J Gastroenterol Hepatol 2009;24:278-87.

33. Ghaedi M, Soleimani M, Shabani I, et al. Hepatic differentiation from human

mesenchymal stem cells on a novel nanofiber scaffold. Cell Mol Biol Lett

2012;17:89-106.

34. Chitrangi S, Nair P, Khanna A. Three-dimensional polymer scaffolds for enhanced

differentiation of human mesenchymal stem cells to hepatocyte-like cells: a

comparative study. J Tissue Eng Regen Med 2016.

35. Lee HJ, Jung J, Cho KJ, et al. Comparison of in vitro hepatogenic differentiation

potential between various placenta-derived stem cells and other adult stem cells as

an alternative source of functional hepatocytes. Differentiation 2012;84:223-31.

36. Jenne CN, Kubes P. Immune surveillance by the liver. Nat Immunol

2013;14:996-1006.

37. Winau F, Hegasy G, Weiskirchen R, et al. Ito cells are liver-resident antigen-presenting

cells for activating T cell responses. Immunity 2007;26:117-29.

38. Vinas O, Bataller R, Sancho-Bru P, et al. Human hepatic stellate cells show features of

antigen-presenting cells and stimulate lymphocyte proliferation. Hepatology

2003;38:919-29.

39. Gao D, Li J, Orosz CG, et al. Different costimulation signals used by CD4(+) and CD8(+)

cells that independently initiate rejection of allogenic hepatocytes in mice.

Hepatology 2000;32:1018-28.

40. Seki E, Schwabe RF. Hepatic inflammation and fibrosis: functional links and key

pathways. Hepatology 2015;61:1066-79.

41. Prockop DJ, Oh JY. Mesenchymal stem/stromal cells (MSCs): role as guardians of

inflammation. Mol Ther 2012;20:14-20.

42. Spaggiari GM, Capobianco A, Abdelrazik H, et al. Mesenchymal stem cells inhibit

natural killer-cell proliferation, cytotoxicity, and cytokine production: role of

indoleamine 2,3-dioxygenase and prostaglandin E2. Blood 2008;111:1327-33.

43. Hong IH, Han SY, Ki MR, et al. Inhibition of kupffer cell activity improves

transplantation of human adipose-derived stem cells and liver functions. Cell

Transplant 2013;22:447-59.

44. Prockop DJ. Concise review: two negative feedback loops place mesenchymal

Page 21: Historical Perspectives and Advances in Mesenchymal Stem ...download.xuebalib.com/4edru0NJdOQ6.pdf · reactivity, and tumorigenic potential. We review directions and advances in the

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

20

stem/stromal cells at the center of early regulators of inflammation. Stem Cells

2013;31:2042-6.

45. Naji A, Rouas-Freiss N, Durrbach A, et al. Concise review: combining human

leukocyte antigen G and mesenchymal stem cells for immunosuppressant biotherapy.

Stem Cells 2013;31:2296-303.

46. Deng Y, Zhang Y, Ye L, et al. Umbilical Cord-derived Mesenchymal Stem Cells Instruct

Monocytes Towards an IL10-producing Phenotype by Secreting IL6 and HGF. Sci Rep

2016;6:37566.

47. Meisel R, Zibert A, Laryea M, et al. Human bone marrow stromal cells inhibit

allogeneic T-cell responses by indoleamine 2,3-dioxygenase-mediated tryptophan

degradation. Blood 2004;103:4619-21.

48. Ezquer F, Ezquer M, Contador D, et al. The antidiabetic effect of mesenchymal stem

cells is unrelated to their transdifferentiation potential but to their capability to

restore Th1/Th2 balance and to modify the pancreatic microenvironment. Stem Cells

2012;30:1664-74.

49. Woo J, Iyer S, Cornejo MC, et al. Stress protein-induced immunosuppression:

inhibition of cellular immune effector functions following overexpression of haem

oxygenase (HSP 32). Transpl Immunol 1998;6:84-93.

50. Soleymaninejadian E, Pramanik K, Samadian E. Immunomodulatory properties of

mesenchymal stem cells: cytokines and factors. Am J Reprod Immunol 2012;67:1-8.

51. Sun L, Akiyama K, Zhang H, et al. Mesenchymal stem cell transplantation reverses

multiorgan dysfunction in systemic lupus erythematosus mice and humans. Stem

Cells 2009;27:1421-32.

52. Augello A, Tasso R, Negrini SM, et al. Bone marrow mesenchymal progenitor cells

inhibit lymphocyte proliferation by activation of the programmed death 1 pathway.

Eur J Immunol 2005;35:1482-90.

53. Vandermeulen M, Gregoire C, Briquet A, et al. Rationale for the potential use of

mesenchymal stromal cells in liver transplantation. World J Gastroenterol

2014;20:16418-32.

54. Wan CD, Cheng R, Wang HB, et al. Immunomodulatory effects of mesenchymal stem

cells derived from adipose tissues in a rat orthotopic liver transplantation model.

Hepatobiliary Pancreat Dis Int 2008;7:29-33.

55. Wang JW, Liu YB, Xu B, et al. [The study on immunomodulation of donor

mesenchymal stem cells on discordant liver xenotransplantation]. Zhonghua Wai Ke

Za Zhi 2005;43:1254-8.

56. Wang Y, Zhang A, Ye Z, et al. Bone marrow-derived mesenchymal stem cells inhibit

acute rejection of rat liver allografts in association with regulatory T-cell expansion.

Transplant Proc 2009;41:4352-6.

Page 22: Historical Perspectives and Advances in Mesenchymal Stem ...download.xuebalib.com/4edru0NJdOQ6.pdf · reactivity, and tumorigenic potential. We review directions and advances in the

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

21

57. Sun Z, Li T, Wen H, et al. Immunological effect induced by mesenchymal stem cells in

a rat liver transplantation model. Exp Ther Med 2015;10:401-406.

58. Peng Y, Chen X, Liu Q, et al. Mesenchymal stromal cells infusions improve refractory

chronic graft versus host disease through an increase of CD5+ regulatory B cells

producing interleukin 10. Leukemia 2015;29:636-46.

59. Aggarwal S, Pittenger MF. Human mesenchymal stem cells modulate allogeneic

immune cell responses. Blood 2005;105:1815-22.

60. 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;152:1174-1186.

61. Ryu KH, Kim SY, Kim YR, et al. Tonsil-derived mesenchymal stem cells alleviate

concanavalin A-induced acute liver injury. Exp Cell Res 2014;326:143-54.

62. Koyama Y, Brenner DA. Liver inflammation and fibrosis. The Journal of Clinical

Investigation 2017;127:55-64.

63. Huang B, Cheng X, Wang H, et al. Mesenchymal stem cells and their secreted

molecules predominantly ameliorate fulminant hepatic failure and chronic liver

fibrosis in mice respectively. J Transl Med 2016;14:45.

64. Li Q, Zhou X, Shi Y, et al. In vivo tracking and comparison of the therapeutic effects of

MSCs and HSCs for liver injury. PLoS One 2013;8:e62363.

65. Akiyama K, Chen C, Wang D, et al. Mesenchymal-stem-cell-induced

immunoregulation involves FAS-ligand-/FAS-mediated T cell apoptosis. Cell Stem Cell

2012;10:544-55.

66. Higashiyama R, Inagaki Y, Hong YY, et al. Bone marrow-derived cells express matrix

metalloproteinases and contribute to regression of liver fibrosis in mice. Hepatology

2007;45:213-22.

67. Haldar D, Henderson NC, Hirschfield G, et al. Mesenchymal stromal cells and liver

fibrosis: a complicated relationship. FASEB J 2016;30:3905-3928.

68. Huang CK, Lee SO, Lai KP, et al. Targeting androgen receptor in bone marrow

mesenchymal stem cells leads to better transplantation therapy efficacy in liver

cirrhosis. Hepatology 2013;57:1550-63.

69. 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;351:487-95.

70. Li B, Shao Q, Ji D, et al. Mesenchymal stem cells mitigate cirrhosis through BMP7. Cell

Physiol Biochem 2015;35:433-40.

71. Meier RP, Mahou R, Morel P, et al. Microencapsulated human mesenchymal stem

cells decrease liver fibrosis in mice. J Hepatol 2015;62:634-41.

72. Chen X, Gan Y, Li W, et al. The interaction between mesenchymal stem cells and

Page 23: Historical Perspectives and Advances in Mesenchymal Stem ...download.xuebalib.com/4edru0NJdOQ6.pdf · reactivity, and tumorigenic potential. We review directions and advances in the

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

22

steroids during inflammation. Cell Death Dis 2014;5:e1009.

73. Pan RL, Wang P, Xiang LX, et al. Delta-like 1 serves as a new target and contributor to

liver fibrosis down-regulated by mesenchymal stem cell transplantation. J Biol Chem

2011;286:12340-8.

74. van Poll D, Parekkadan B, Cho CH, et al. Mesenchymal stem cell-derived molecules

directly modulate hepatocellular death and regeneration in vitro and in vivo.

Hepatology 2008;47:1634-43.

75. Aurich I, Mueller LP, Aurich H, et al. Functional integration of hepatocytes derived

from human mesenchymal stem cells into mouse livers. Gut 2007;56:405-15.

76. Aurich H, Sgodda M, Kaltwasser P, et al. Hepatocyte differentiation of mesenchymal

stem cells from human adipose tissue in vitro promotes hepatic integration in vivo.

Gut 2009;58:570-81.

77. Lee CW, Hsiao WT, Lee OK. Mesenchymal stromal cell-based therapies reduce obesity

and metabolic syndromes induced by a high-fat diet. Transl Res 2016.

78. Ji AT, Chang YC, Fu YJ, et al. Niche-dependent regulations of metabolic balance in

high-fat diet-induced diabetic mice by mesenchymal stromal cells. Diabetes

2015;64:926-36.

79. Oh SH, Witek RP, Bae SH, et al. Bone marrow-derived hepatic oval cells differentiate

into hepatocytes in 2-acetylaminofluorene/partial hepatectomy-induced liver

regeneration. Gastroenterology 2007;132:1077-87.

80. Biancone L, Bruno S, Deregibus MC, et al. Therapeutic potential of mesenchymal

stem cell-derived microvesicles. Nephrol Dial Transplant 2012;27:3037-42.

81. Lai RC, Yeo RWY, Lim SK. Mesenchymal stem cell exosomes. Seminars in Cell &

Developmental Biology 2015;40:82-88.

82. Sato K, Meng F, Glaser S, et al. Exosomes in liver pathology. J Hepatol

2016;65:213-21.

83. Hirsova P, Ibrahim SH, Verma VK, et al. Extracellular vesicles in liver pathobiology:

Small particles with big impact. Hepatology 2016;64:2219-2233.

84. Han C, Sun X, Liu L, et al. Exosomes and Their Therapeutic Potentials of Stem Cells.

Stem Cells Int 2016;2016:7653489.

85. Li T, Yan Y, Wang B, et al. Exosomes derived from human umbilical cord mesenchymal

stem cells alleviate liver fibrosis. Stem Cells Dev 2013;22:845-54.

86. Tan CY, Lai RC, Wong W, et al. Mesenchymal stem cell-derived exosomes promote

hepatic regeneration in drug-induced liver injury models. Stem Cell Res Ther

2014;5:76.

87. 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.

88. Hyun J, Wang S, Kim J, et al. MicroRNA125b-mediated Hedgehog signaling influences

Page 24: Historical Perspectives and Advances in Mesenchymal Stem ...download.xuebalib.com/4edru0NJdOQ6.pdf · reactivity, and tumorigenic potential. We review directions and advances in the

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

23

liver regeneration by chorionic plate-derived mesenchymal stem cells. Sci Rep

2015;5:14135.

89. Tamura R, Uemoto S, Tabata Y. Immunosuppressive effect of mesenchymal stem

cell-derived exosomes on a concanavalin A-induced liver injury model. Inflammation

and Regeneration 2016;36:26.

90. F. S. Prime ricerche per la realizzazione di un fegato artificiale. Chir Patol Sperim.

1956;4:1401-1404.

91. Burnell JM, Dawborn JK, Epstein RB, et al. Acute hepatic coma treated by

cross-circulation or exchange transfusion. N Engl J Med 1967;276:935-43.

92. Suh KS, Lilja H, Kamohara Y, et al. Bioartificial liver treatment in rats with fulminant

hepatic failure: effect on DNA-binding activity of liver-enriched and

growth-associated transcription factors. J Surg Res 1999;85:243-50.

93. Flendrig LM, Calise F, Di Florio E, et al. Significantly improved survival time in pigs

with complete liver ischemia treated with a novel bioartificial liver. Int J Artif Organs

1999;22:701-9.

94. Sosef MN, Abrahamse LS, van de Kerkhove MP, et al. Assessment of the

AMC-bioartificial liver in the anhepatic pig. Transplantation 2002;73:204-9.

95. Matsumura KN, Guevara GR, Huston H, et al. Hybrid bioartificial liver in hepatic

failure: preliminary clinical report. Surgery 1987;101:99-103.

96. van de Kerkhove MP, Hoekstra R, Chamuleau RA, et al. Clinical application of

bioartificial liver support systems. Ann Surg 2004;240:216-30.

97. Nicolas CT, Hickey RD, Chen HS, et al. Concise Review: Liver Regenerative Medicine:

From Hepatocyte Transplantation to Bioartificial Livers and Bioengineered Grafts.

Stem Cells 2017;35:42-50.

98. Shi XL, Gao Y, Yan Y, et al. Improved survival of porcine acute liver failure by a

bioartificial liver device implanted with induced human functional hepatocytes. Cell

Res 2016;26:206-16.

99. Hannoun Z, Steichen C, Dianat N, et al. The potential of induced pluripotent stem cell

derived hepatocytes. J Hepatol 2016;65:182-199.

100. Sakiyama R, Blau BJ, Miki T. Clinical translation of bioartificial liver support systems

with human pluripotent stem cell-derived hepatic cells. World J Gastroenterol

2017;23:1974-1979.

101. Ji R, Zhang N, You N, et al. The differentiation of MSCs into functional hepatocyte-like

cells in a liver biomatrix scaffold and their transplantation into liver-fibrotic mice.

Biomaterials 2012;33:8995-9008.

102. Jiang WC, Cheng YH, Yen MH, et al. Cryo-chemical decellularization of the whole liver

for mesenchymal stem cells-based functional hepatic tissue engineering.

Biomaterials 2014;35:3607-17.

Page 25: Historical Perspectives and Advances in Mesenchymal Stem ...download.xuebalib.com/4edru0NJdOQ6.pdf · reactivity, and tumorigenic potential. We review directions and advances in the

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

24

103. op den Dries S, Westerkamp AC, Karimian N, et al. Injury to peribiliary glands and

vascular plexus before liver transplantation predicts formation of non-anastomotic

biliary strictures. J Hepatol 2014;60:1172-9.

104. Pan XN, Zheng LQ, Lai XH. Bone marrow-derived mesenchymal stem cell therapy for

decompensated liver cirrhosis: a meta-analysis. World J Gastroenterol

2014;20:14051-7.

105. Kim G, Eom YW, Baik SK, et al. Therapeutic Effects of Mesenchymal Stem Cells for

Patients with Chronic Liver Diseases: Systematic Review and Meta-analysis. J Korean

Med Sci 2015;30:1405-15.

106. Franco D. Towards a bioengineered liver. J Hepatol 2014;60:455-6.

107. Collin de l'Hortet A, Takeishi K, Guzman-Lepe J, et al. Liver-Regenerative

Transplantation: Regrow and Reset. Am J Transplant 2016;16:1688-96.

108. Bao J, Shi Y, Sun H, et al. Construction of a portal implantable functional

tissue-engineered liver using perfusion-decellularized matrix and hepatocytes in rats.

Cell Transplant 2011;20:753-66.

109. Christ B, Bruckner S, Winkler S. The Therapeutic Promise of Mesenchymal Stem Cells

for Liver Restoration. Trends Mol Med 2015;21:673-86.

110. Berardis S, Dwisthi Sattwika P, Najimi M, et al. Use of mesenchymal stem cells to

treat liver fibrosis: current situation and future prospects. World J Gastroenterol

2015;21:742-58.

111. Eom YW, Shim KY, Baik SK. Mesenchymal stem cell therapy for liver fibrosis. Korean J

Intern Med 2015;30:580-9.

112. Zhang Z, Lin H, Shi M, et al. Human umbilical cord mesenchymal stem cells improve

liver function and ascites in decompensated liver cirrhosis patients. J Gastroenterol

Hepatol 2012;27 Suppl 2:112-20.

113. Amin MA, Sabry D, Rashed LA, et al. Short-term evaluation of autologous

transplantation of bone marrow-derived mesenchymal stem cells in patients with

cirrhosis: Egyptian study. Clin Transplant 2013;27:607-12.

114. Jang YO, Kim YJ, Baik SK, et al. Histological improvement following administration of

autologous bone marrow-derived mesenchymal stem cells for alcoholic cirrhosis: a

pilot study. Liver Int 2014;34:33-41.

115. Kamath PS, Wiesner RH, Malinchoc M, et al. A model to predict survival in patients

with end-stage liver disease. Hepatology 2001;33:464-70.

116. Than NN, Tomlinson CL, Haldar D, et al. Clinical effectiveness of cell therapies in

patients with chronic liver disease and acute-on-chronic liver failure: a systematic

review protocol. Syst Rev 2016;5:100.

117. Suk KT, Yoon JH, Kim MY, et al. Transplantation with autologous bone marrow-derived

mesenchymal stem cells for alcoholic cirrhosis: Phase 2 trial. Hepatology

Page 26: Historical Perspectives and Advances in Mesenchymal Stem ...download.xuebalib.com/4edru0NJdOQ6.pdf · reactivity, and tumorigenic potential. We review directions and advances in the

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

25

2016;64:2185-2197.

118. Thomson JA, Itskovitz-Eldor J, Shapiro SS, et al. Embryonic stem cell lines derived

from human blastocysts. Science 1998;282:1145-7.

119. Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic

and adult fibroblast cultures by defined factors. Cell 2006;126:663-76.

120. Takahashi K, Tanabe K, Ohnuki M, et al. Induction of pluripotent stem cells from adult

human fibroblasts by defined factors. Cell 2007;131:861-72.

121. Muller SA, Mehrabi A, Schmied BM, et al. Partial liver transplantation-living donor

liver transplantation and split liver transplantation. Nephrol Dial Transplant 2007;22

Suppl 8:viii13-viii22.

122. Chen YF, Tseng CY, Wang HW, et al. Rapid generation of mature hepatocyte-like cells

from human induced pluripotent stem cells by an efficient three-step protocol.

Hepatology 2012;55:1193-203.

123. Vosough M, Omidinia E, Kadivar M, et al. Generation of functional hepatocyte-like

cells from human pluripotent stem cells in a scalable suspension culture. Stem Cells

Dev 2013;22:2693-705.

124. Woo DH, Kim SK, Lim HJ, et al. Direct and indirect contribution of human embryonic

stem cell-derived hepatocyte-like cells to liver repair in mice. Gastroenterology

2012;142:602-11.

125. Lian Q, Zhang Y, Zhang J, et al. Functional mesenchymal stem cells derived from

human induced pluripotent stem cells attenuate limb ischemia in mice. Circulation

2010;121:1113-23.

126. Sun YQ, Zhang Y, Li X, et al. Insensitivity of Human iPS Cells-Derived Mesenchymal

Stem Cells to Interferon-gamma-induced HLA Expression Potentiates Repair

Efficiency of Hind Limb Ischemia in Immune Humanized NOD Scid Gamma Mice.

Stem Cells 2015;33:3452-67.

127. Zhang Y, Liao S, Yang M, et al. Improved cell survival and paracrine capacity of human

embryonic stem cell-derived mesenchymal stem cells promote therapeutic potential

for pulmonary arterial hypertension. Cell Transplant 2012;21:2225-39.

128. Uygun BE, Soto-Gutierrez A, Yagi H, et al. Organ reengineering through development

of a transplantable recellularized liver graft using decellularized liver matrix. Nat Med

2010;16:814-20.

129. Gerbal-Chaloin S, Funakoshi N, Caillaud A, et al. Human induced pluripotent stem

cells in hepatology: beyond the proof of concept. Am J Pathol 2014;184:332-47.

130. Ben-David U, Kopper O, Benvenisty N. Expanding the boundaries of embryonic stem

cells. Cell Stem Cell 2012;10:666-77.

131. Lund RJ, Narva E, Lahesmaa R. Genetic and epigenetic stability of human pluripotent

stem cells. Nat Rev Genet 2012;13:732-44.

Page 27: Historical Perspectives and Advances in Mesenchymal Stem ...download.xuebalib.com/4edru0NJdOQ6.pdf · reactivity, and tumorigenic potential. We review directions and advances in the

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

26

132. Hussein SM, Batada NN, Vuoristo S, et al. Copy number variation and selection during

reprogramming to pluripotency. Nature 2011;471:58-62.

133. Laurent LC, Ulitsky I, Slavin I, et al. Dynamic changes in the copy number of

pluripotency and cell proliferation genes in human ESCs and iPSCs during

reprogramming and time in culture. Cell Stem Cell 2011;8:106-18.

134. Martins-Taylor K, Nisler BS, Taapken SM, et al. Recurrent copy number variations in

human induced pluripotent stem cells. Nat Biotechnol 2011;29:488-91.

135. Gore A, Li Z, Fung HL, et al. Somatic coding mutations in human induced pluripotent

stem cells. Nature 2011;471:63-7.

136. Abyzov A, Mariani J, Palejev D, et al. Somatic copy number mosaicism in human skin

revealed by induced pluripotent stem cells. Nature 2012;492:438-42.

137. Lamm N, Ben-David U, Golan-Lev T, et al. Genomic Instability in Human Pluripotent

Stem Cells Arises from Replicative Stress and Chromosome Condensation Defects.

Cell Stem Cell 2016;18:253-261.

138. Zhao T, Zhang ZN, Rong Z, et al. Immunogenicity of induced pluripotent stem cells.

Nature 2011;474:212-5.

139. Fox IJ, Daley GQ, Goldman SA, et al. Stem cell therapy. Use of differentiated

pluripotent stem cells as replacement therapy for treating disease. Science

2014;345:1247391.

140. Wei X, Yang X, Han ZP, et al. Mesenchymal stem cells: a new trend for cell therapy.

Acta Pharmacol Sin 2013;34:747-54.

141. Keating A. Mesenchymal stromal cells: new directions. Cell Stem Cell

2012;10:709-16.

142. Barkholt L, Flory E, Jekerle V, et al. Risk of tumorigenicity in mesenchymal stromal

cell-based therapies--bridging scientific observations and regulatory viewpoints.

Cytotherapy 2013;15:753-9.

143. Grigorian AS, Kruglyakov PV, Taminkina UA, et al. Alterations of cytological and

karyological profile of human mesenchymal stem cells during in vitro culturing. Bull

Exp Biol Med 2010;150:125-30.

144. Ben-David U, Mayshar Y, Benvenisty N. Large-scale analysis reveals acquisition of

lineage-specific chromosomal aberrations in human adult stem cells. Cell Stem Cell

2011;9:97-102.

145. Josse C, Schoemans R, Niessen NA, et al. Systematic chromosomal aberrations found

in murine bone marrow-derived mesenchymal stem cells. Stem Cells Dev

2010;19:1167-73.

146. Miura M, Miura Y, Padilla-Nash HM, et al. Accumulated chromosomal instability in

murine bone marrow mesenchymal stem cells leads to malignant transformation.

Stem Cells 2006;24:1095-103.

Page 28: Historical Perspectives and Advances in Mesenchymal Stem ...download.xuebalib.com/4edru0NJdOQ6.pdf · reactivity, and tumorigenic potential. We review directions and advances in the

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

27

147. Foudah D, Redaelli S, Donzelli E, et al. Monitoring the genomic stability of in vitro

cultured rat bone-marrow-derived mesenchymal stem cells. Chromosome Res

2009;17:1025-39.

148. Rosland GV, Svendsen A, Torsvik A, et al. Long-term cultures of bone marrow-derived

human mesenchymal stem cells frequently undergo spontaneous malignant

transformation. Cancer Res 2009;69:5331-9.

Page 29: Historical Perspectives and Advances in Mesenchymal Stem ...download.xuebalib.com/4edru0NJdOQ6.pdf · reactivity, and tumorigenic potential. We review directions and advances in the

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

Table 1. Current MSC hepatic differentiation protocols

Cell Source Differentiation Protocol Cell Characteristics Reference

Rat bone marrow

DMEM-LG and MCDB-201

(6:4) supplemented with 0.5%

FBS, 1 µM dexamethasone,

10 ng/mL EGF, and ITS+

(Insulin-Transferrin-Selenium)

premix

Round cells expressing

CK18 and albumin [17]

Human bone marrow

Induction: IMDM

supplemented with 20 ng/mL

HGF, 10 ng/mL FGF2, and

0.61 g/L nicotinamide

Maturation: IMDM

supplemented with 20 ng/mL

oncostatin M, 1 µM

dexamethasone, and 50

mg/mL ITS+ premix

Cuboidal cells with

abundant intracellular

granules, expressing

liver-specific genes;

glycogen storage, urea

production, and LDL uptake

[18]

Human umbilical cord blood

Induction: IMDM

supplemented with 10% FBS,

0.5 µM dexamethasone, 50

mg/mL ITS+ premix, and 50

ng/mL HGF

Maturation: IMDM

supplemented with 10% FBS,

0.5 µM dexamethasone, 50

mg/mL ITS+ premix, and 50

ng/mL oncostatin M

Round cells expressing

liver-specific genes; LDL

uptake

[19]

Human adipose tissue

Hepatic commitment:

HCM-modified William’s E

medium supplemented with

150 ng/mL HGF, 300 ng/mL

FGF1, and 25 ng/mL FGF4

Maturation: HCM-modified

William’s E medium

supplemented with 30 ng/mL

oncostatin M and 0.2 µM

dexamethasone

Round cells expressing

liver-specific genes;

glycogen storage, urea

production, and LDL uptake

[21]

Page 30: Historical Perspectives and Advances in Mesenchymal Stem ...download.xuebalib.com/4edru0NJdOQ6.pdf · reactivity, and tumorigenic potential. We review directions and advances in the

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

Rat bone marrow Co-cultured with primary liver

cells and injured liver tissue

Spheroids expressing

liver-specific genes;

glycogen storage

[23,24,25]

Page 31: Historical Perspectives and Advances in Mesenchymal Stem ...download.xuebalib.com/4edru0NJdOQ6.pdf · reactivity, and tumorigenic potential. We review directions and advances in the

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

Page 32: Historical Perspectives and Advances in Mesenchymal Stem ...download.xuebalib.com/4edru0NJdOQ6.pdf · reactivity, and tumorigenic potential. We review directions and advances in the

本文献由“学霸图书馆-文献云下载”收集自网络,仅供学习交流使用。

学霸图书馆(www.xuebalib.com)是一个“整合众多图书馆数据库资源,

提供一站式文献检索和下载服务”的24 小时在线不限IP

图书馆。

图书馆致力于便利、促进学习与科研,提供最强文献下载服务。

图书馆导航:

图书馆首页 文献云下载 图书馆入口 外文数据库大全 疑难文献辅助工具