Advances in cell sources of hepatocytes for bioartificial liver
2012-04-07
Hangzhou, China
Advances in cell sources of hepatocytes for bioartificial liver
Xiao-Ping Pan and Lan-Juan Li
Hangzhou, China
BACKGROUND:Orthotopic liver transplantation (OLT) is the most effective therapy for liver failure. However, OLT is severely limited by the shortage of liver donors. Bioartificial liver (BAL) shows great potential as an alternative therapy for liver failure. In recent years, progress has been made in BAL regarding genetically engineered cell lines, immortalized human hepatocytes, methods for preserving the phenotype of primary human hepatocytes, and other functional hepatocytes derived from stem cells.
DATA SOURCES:A systematic search of PubMed and ISI Web of Science was performed to identify relevant studies in English language literature using the key words such as liver failure, bioartificial liver, hepatocyte, stem cells, differentiation, and immortalization. More than 200 articles related to the cell sources of hepatocyte in BAL were systematically reviewed.
RESULTS:Methods for preserving the phenotype of primary human hepatocytes have been successfully developed. Many genetically engineered cell lines and immortalized human hepatocytes have also been established. Among these cell lines, the incorporation of BAL with GS-HepG2 cells or alginateencapsulated HepG2 cells could prolong the survival time and improve pathophysiological parameters in an animal model of liver failure. The cBAL111 cells were evaluated using the AMC-BAL bioreactor, which could eliminate ammonia and lidocaine, and produce albumin. Importantly, BAL loading with HepLi-4 cells could significantly improve the blood biochemical parameters, and prolong the survival time in pigs with liver failure. Other functional hepatocytes differentiated from stem cells, such as human liver progenitor cells, have been successfully achieved.
CONCLUSIONS:Aside from genetically modified liver cell lines and immortalized human hepatocytes, other functionalhepatocytes derived from stem cells show great potential as cell sources for BAL. BAL with safe and effective liver cells may be achieved for clinical liver failure in the near future.
(Hepatobiliary Pancreat Dis Int 2012;11:594-605)
bioartificial liver;liver failure; hepatocyte; liver cell source; stem cell; differentiation; immortalization
Introduction
Fulminant liver failure has very high mortality rates approaching 80%.[1,2]To date, orthotopic liver transplantation (OLT) is the most effective therapy for acute liver failure. However, OLT is severely limited by several factors, such as the shortage of liver organ donors, the risk of immune rejection, and the high costs of transplantation. The human liver has a remarkable capacity to regenerate after physical or toxic injury.[3-5]Thus, research has focused on alternative ways to bridge patients with liver failure until successful liver transplantation or the liver of the patient sufficiently regenerates to restore liver function.
Alternative therapies for the treatment of liver failure have been investigated and developed in recent years. The therapies include artificial liver support system,[6-8]bioartificial liver (BAL) support system,[9-11]and hepatocyte transplantation.[12-15]The artificial liver support system is purely a mechanical device or noncell based liver support device, which includes the molecular absorbent and recirculating system (MARS), prometheus system, single pass albumin dialysis (SPAD), selective plasma filtration, and so on. The system could provide detoxification function and improve the survival rate of patients with liver failure.[6-8,16,17]The artificial liver support system promises limited success because of the insufficient replacement of synthetic andmetabolic liver functions and the shortage of normal human plasma supply.[7,18-20]
BAL typically incorporates hepatocytes into bioreactors to provide the important liver functions such as oxidative detoxification, biotransformation, excretion and synthesis. Theoretically, BAL can compensate for the functions of an almost entire liver apart from the secretion of bile. To date, many BAL systems have been applied in clinical trials for liver failure.[21-28]The systems appear to be safe and result in an improvement in the clinical neurologic status and blood biochemical parameters of patients with liver failure. However, none of them has been approved by the medical authorities for routine clinical therapy of liver failure. Nevertheless, some substantial progress has been made in the isolation and preservation of primary human hepatocytes and the establishment of immortalized human hepatocytes and genetically engineered liver cell lines. Stem cells exhibit highly proliferative activity and multipotential ability of differentiation.[29,30]Stem cells are derived from human embryonic stem cells (hESCs),[31-33]human induced pluripotent stem cells (iPSCs),[34,35]human liver progenitor cells (HLPCs),[36]and human mesenchymal stem cells (MSCs),[37]possessing potential ability for differentiation of hepatocytes. They may provide functional hepatocytes for clinical use such as BAL and hepatocyte transplantation.[30,38-40]
Liver cells and bioreactors are the two key components of BAL. The progress of the optimization and modification of bioreactor configurations for BAL has been previously reviewed.[41]In this review, we focus on the advances in liver cell sources, the cellular component of BAL for potential application, including primary porcine hepatocytes, isolation and preservation of primary human hepatocytes, genetically engineered liver cell lines, immortalized human hepatocytes, hESCs, HLPCs, iPSCs, and MSCs.
Primary porcine hepatocytes
Owing to their easy availability and high functional activities, primary porcine hepatocytes are most commonly used in BAL systems in clinical trials. A randomized controlled trial of BAL using primary porcine hepatocytes demonstrated that it is safe and effective in patients with fulminant/subfulminant liver failure.[27]However, primary porcine hepatocytes have a short lifespan and rapidly lose liver functionsin vitro, such as protein synthesis, ureagenesis, and cytochrome P450 activity. As previously reported, several immortalized porcine hepatocytes are established by transfer of SV40 LT into primary porcine hepatocytes.[42-44]This action enables primary porcine hepatocytes to pass and keep their key functions for a long time.
However, porcine endogenous retrovirus (PERV) poses a potential risk in pig-to-human xenozoonosis.[45-47]No evidence ofin vivoproductive infection in humans is currently available.[48-50]Specifically, no PERV infection has been found in long-term immunosuppressed patients who received porcine hepatocyte-based BAL after 8.7 years of follow-up.[51]However, PERV can still be transferred to the membranes of bioreactors, and PERV transmission after short-term direct contact of primary porcine liver cells supernatants with primary human cells could infect the human cells.[52,53]Because of crossspecies immunologic reactions and the potential risk of xenozoonosis, the use of primary porcine hepatocytes is severely hampered in BAL. Thus, the utilization of primary porcine hepatocytes for BAL is prohibited in several European countries.[54]
Isolation and preservation of primary human hepatocytes
Primary human hepatocytes are the most desirable option for BAL. However, liver donors for the isolation of primary human hepatoyctes, as well as for the entire liver transplantation, are scarce. Currently, human livers for hepatocyte isolation are commonly obtained from rejected livers for transplantation or non-transplantable liver organs, such as steatic liver and cirrhotic liver, which possess relatively lower cell yield and viability.[55-58]A standardized and cGMP-conformed method of human hepatocyte isolation has been developed to improve the cell viability for BAL.[59]
Primary human hepatocytes also have limited proliferation and easily lose liver functionin vitro. A number of approaches have been applied to preserve the phenotype of hepatocytes and improve liver-specific functions. Some of these approaches include improvement in cell media components, addition of extracellular matrix,[60,61]modification of biomaterials,[62]three-dimensional culture,[63,64]and co-culture of human hepatocytes and nonparenchymal liver cells[65]for long-time culture of hepatocytein vitro.
Among these methods, co-culture of hepatocytes and nonparenchymal liver cells is very important for preserving liver functions of primary human hepatocytes. Nonparenchymal liver cells include human umbilical vein endothelial cells,[66]endothelial vascular structures,[67]human hepatic stellate cells,[68]human biliary epithelial cells,[69]and bone marrow-derived MSCs.[70]
Importantly, the liver functions of primary human hepatocytes cultured on a bio-modified scaffold or biomaterial membrane are enhanced and maintained at high levels for a longer time, such as modified polyetheretherketone and polyurethane (PEEK-WCPU) membranes and NH3 plasma-grafted PEEK-WCPU membranes.[71,72]These results indicate that biomodified scaffolds are promising biomaterials for the potential use in BAL.
Human hepatoma cell line HepG2 is the most commonly used for BAL because of the advantages of the readily available and importantly secreting human proteins. The liver tumor-derived cell line C3A, a subclone of HepG2, has been used in the ELAD system for clinical trials. Improvement in either survival or biochemical parameters was not observed following the pilot-controlled clinical trial when C3A liver cells were applied to the ELAD system as the cell component of BAL.[21,73]Thus, the liver cell line C3A is of limited use because of its low liver functions.
More importantly, co-culture of HepG2 cells with nonparenchymal liver cells may also improve liver-specific functions.[74-77]Cultivation of C3A cells with alginate beads effectively improves albumin production and easily forms microvilli-like structures characteristic of normal hepatocytes, thereby improving mass transfers and enhancing treatment efficacy.[78,79]The liver differentiation function of FLC-4, a novel hepatoma cell line, can be significantly enhanced when it is cultured in three dimensional cell shape and lactose-silk fibroin conjugate sponges.[80,81]
Genetically engineered tumor liver cell lines
Due to HepG2 cells lacking some key liver functions, some genes such as glutamine synthetase (GS) and hepatocyte nuclear factor (HNF)-4 genes were transferred into HepG2 cells to enhance their weak or lost liver functions. Thus, several novel liver cell lines including HepG2-GS,[82,83]HepG2-GS-3A4,[84,85]HepG2-Bcl2,[86]HepG2/(hArgI+hOTC)4,[87]cBAL119,[88]and HepG2/HNF-4[89]were developed through genetic engineering. The cBAL119 was generated by transduction of HepG2 cells with the human pregnane X receptor (PXR) gene. All these genetically engineered HepG2 can improve the liver-specific functions of hepatocytes for potential application in bioartificial liver. To address intrinsic tumorigenesis, thymidine kinase (TK) was transferred into HepG2 cells (i.e., HepG2/ tk). Moreover, intrasplenic injection of HepG2/tk into rats with liver failure significantly improved blood biochemical parameters and significantly prolonged the survival of rats.[90]These findings indicate that HepG2/ tk is a potential cell source for BAL in the future.
Immortalized human hepatocyte cell lines
To address the issue that primary human hepatocytes easily lose liver functions and do not proliferate for a long-termin vitroculture, several liver cell lines have been developed by gene transfer of the simian virus 40 large antigen gene (SV40 LT) or human telomerase reverse transcriptase such as immortalized human hepatocytes[91-93]and immortalized human fetal hepatocytes.[93-95]
To avoid the risk of SV40 LT tumorigensis, several safer immortalized human hepatocytes have been established, such as the SV40 LT temperature-sensitive cell line[96]and OUMS-29/TK.[97]Moreover, researchers have adopted a reversible immortalization system that uses the Cre-loxP site-specific recombination reaction to remove the targeting SV40 LT gene in reversibly immortalized human hepatocytes, such as NKNT-3, 16-T3, and HepLi4 cells.[98-101]
Immortalized human hepatocytes have been cocultured with human stellate cells or microencapsulated for large-scale culture using roller bottles to improve the differentiation grade and liver functions of immortalized human hepatocytes, which could be candidates for use in BAL.[102,103]
Researchers have already focused on deriving functional hepatocytes from stem cells to address the issue of cell sources of hepatocytes for application in BAL. Stem cells could be differentiated into functional hepatocytes, such as hESCs, HLPCs, iPSCs, and human MSCs.
hESCs
hESCs are isolated from the inner cell mass of blastocysts of fertilized human embryos, having powerful self-renewal and pluripotency.[104,105]hESCs have powerful potential to produce functional hepatocytes as a source of hepatocytes for cellbased replacement therapies and transplantation.[106]Rambhatla et al[107]reported that hESCs could be differentiated into hepatocyte-like cells.
Several protocols for hepatic differentiation of hESCs were developed to provide a source of functional hepatocytes. HNF4α transduction could enhance the efficient generation of functional hepatocytes from hESCs.[108]A relatively homogenous population of hepatocytes was generated from hESCs, which appear to have complete metabolic function of primary hepatocytes.[109]hESCs could be differentiated efficientlyinto functional hepatocytes under a three-stage process, in which hESCs are firstly enabled toward definitive endoderm with activin A and sodium butyrate, followed by dimethyl sulfoxide (DMSO), hepatocyte growth factor, and oncostatin M. hESC-derived functional hepatocytes express several members of cytochrome P450 isozymes, which are capable of converting the substrates to metabolites.[110]Functional hepatocytes could also be generated from hESCs under chemically defined conditions, including a combination of activin, fibroblast growth factor 2, bone morphogenetic protein 4, phosphoinositide 3-kinase inhibition, fibroblast growth factor 10, retinoic acid, and an inhibitor of activin/nodal receptor.[111]
A 3D co-culture system, 3D collagen scaffolds, and ultraweb nanofibers could promote hepatic differentiation of hESCs.[112-116]Specifically, functional hepatocytes differentiated from hESCs could exhibit the potential of liver function of hepatocytes after their transplantation into an animal model.[31]Among these differentiation techniques, the 3D co-culture system of hESCs in combination with tissue engineering may provide a source of functional hepatocytes for potential application in novel BAL.
HLPCs
During the development of human liver, HLPCs found in the early fetal liver bud are termed hepatoblasts, and hepatic oval cells are the bipotent progenitors capable of generating hepatocytes and bile duct cells in adult liver.[117-119]Several human fetal liver progenitor cells from the human fetal liver have been isolated and characterized.[120,121]Several liver progenitor cells were successfully isolated and characterized from normal adult human liver and ischemic liver tissue.[122-124]Adult liver progenitor cells and human fetal hepatic progenitor cells could be differentiated into hepatocyte-like cellsin vitro.[36,122]Importantly, liver progenitor cells could be differentiated into mature hepatocyte-like cells when cultured in a rotary BAL.[125]The HepaRG cell line, a human bipotent liver progenitor cell line, could be differentiated into hepatocyte clusters at a high density with or without DMSO. HepaRG cells are very suitable for BAL application.[126,127]
Human iPSCs
iPSCs are defined as reprogrammed somatic cells that share features of embryonic stem cells, including morphology, expression of key pluripotency genes, and unlimited self-renewal.[128-131]Human iPSCs are generated from somatic cells by retroviral induction of four transcription factors, namely, Oct3/4, Sox2, c-Myc, and Klf4.[130-132]Direct reprogramming of somatic cells is effectively promoted by the maternal transcription factor Glis1 or overexpression of the homeobox gene HEX.[133,134]Human iPSCs could be effectively induced into functional hepatocyte-like cells.[34,35,135]HNF4α transduction and 3D co-culture system could enhance the generation of functional hepatocytes from human iPSCs.[108,112]Mature hepatocyte-like cells could be generated from human iPSCs using an efficient three-step protocol.[136]Hepatocyte-like cells also could be efficiently generated from human ESCs and iPSCs via sequential transduction of SOX17 and HEX.[137]Moreover, the transplantation of mature hepatocyte-like cells derived from human iPSCs into a carbon tetrachloride-injured mouse significantly improves the general condition of a liver-injured mouse after transplantation.[138]
Human MSCs
Human MSCs include human bone marrow-derived MSCs (hBMSCs), human adipose-derived stem cells (hADSCs), human placenta-derived MSCs (hPDMSCs), and so on.[40,139,140]
hBMSCs
Bone marrow is a reservoir of hematopoietic stem cells and MSCs. hBMSCs are suitable as therapeutic tools with relatively easy availability and expansionin vitro. The generation of hepatocyte-like cells from hBMSCs has become a real alternative to the isolation of primary hepatocytes.[141]BMSCs could be effectively induced into functional hepatocytes.[142,143]
Alginate scaffold could enhance the differentiation of BMSCs into hepatocyte-like cells.[144]Hepatic differentiation of hBMSCs could be achieved using tetracycline-regulated HNF3β.[145]A novel 3D biocompatible nanofibrous scaffold could produce functional hepatocyte-like cells derived from hBMSCs.[146-148]The functional hepatocytes derived from hBMSCs could exhibit the functional features of hepatocytes and engraft in the host liver parenchyma of immunocompromised mice.[149]
hADSCs
hADSCs not only share similar characteristics with BMSCs having the potential to differentiate into hepatocytes, but also have a less invasive surgical procedure for harvesting them.[150,151]
Human adipose tissue-derived MSCs gain functional hepatocytes under conditions favoring hepatocyte differentiationin vitro,[37,152,153]which could promote hepatic integrationin vivo.[37]Functional hepatocytes can be produced in large numbers for clinical therapy owing to liver failure.[153]
hADSCs have a similar potential for hepatic differentiation into BMSCs, but have a longer culture period and higher proliferation capacity, indicating that adipose tissue may be an ideal source of large amounts of stem cells for hepatocyte transplantation and BAL.[154]Supplementation of five developmental transcription factors may enhance the differentiation of hADSCs into hepatocytes.[155]hADSCs can differentiate into hepatic lineagein vitro, and hADSCs transplanted into a CCl4-injured SCID mouse model can be differentiated into hepatocytesin vivo.[156]In vitroandin vivostudies also showed that poly-lactide-co-glycolide scaffolds could enhance the hepatogenesis of hADSCs.[157]
hPMSCs
Placenta tissue, a biological waste after delivery, has become a potential source of MSCs for basic and clinical application because of its phenotypic plasticity, easy accessibility, and lack of ethical concerns.[158,159]
Owing to the complex structure of the placenta, the fetal adnexa is composed of the placenta, fetal membranes, and umbilical cord. The cell populations isolated from the placenta consist of human amniotic epithelial cells (hAEC), human amniotic MSCs (hAMSCs), human chorionic MSCs (hCMSCs), and human chorionic trophoblastic cells (hCTC). Amniotic epithelial cells, human amniotic membrane MSCs, and umbilical cord blood MSCs will be the focus.
hAECs as stem cell markers are capable of differentiation toward all the three germ layers. hAECs exhibit hepatocyte-like characteristics and functions, and maintain proliferation.[160,161]hAECs could be differentiated into functional hepatocytes bothin vitroandin vivo,[162]indicating that amniotic epithelial cells are a useful and noncontroversial source of cells for clinical application.
Human amniotic membrane MSCs isolated from the placenta, being immune-privileged and advantageous as therapeutic cells, could be induced into cells of functional hepatocytes under appropriate conditions.[163-166]This finding indicates that amniotic membrane MSCs are a uniquely useful and noncontroversial stem cell source.
hPDMSCs could be manufactured by their isolation and expansion from the human placenta.[167]They also can be expanded for large amounts of hPDMSCs using suspension culture bioreactors. In addition, these stem cells are able to maintain their antigenic phenotype.[168]It was reported that transplantation of human amniotic membrane MSCs could ameliorate carbon tetrachlorideinduced liver cirrhosis in mouse.[169,170]Encouragingly, our team has recently reported that hPDMSCs could not only efficiently differentiate into functional hepatocytes, but also prolong the survival time of ALF pigs through portal vein transplantation.[171]
Human umbilical cord blood mesenchymal stem cells (hUCB-MSCs) can be obtained from the source of cells by different methods.[172-174]hUCB-MSCs can be differentiated into functional hepatocyte-like cells with low immunogenicityin vitroorin vivo.[175-180]
More importantly, hUCB-MSCs can enhance the recovery of CCl4-injured mouse liver, thus they could be used for the treatment of liver dysfunction or liver injury.[181,182]Transplantation of hUCB-MSCs can significantly improve the survival of rats with acute hepatic necrosis or acute liver failure.[183,184]
Ongoing preclinical assessment orin vitrotest
In recent years, genetically engineered cell lines, immortalized human hepatocytes, methods for preserving the phenotype of primary human hepatocytes, and other functional hepatocytes derived from stem cells have been developed for preclinical assessment orin vitrotest.
Modified bioreactors mimicking the liver architecture can support the maintenance of primary human hepatocytes, preserving their liver-specific functions for BAL applications, such as 3D tissue re-structuring bioreactors,[185,186]modified polyetheretherketone and polyethersulfone, and crossed hollow fiber membrane bioreactors.[187]Tissue-like structured bioreactors provide a 3D network of interwoven capillary membranes with integrated oxygenation and decentralized mass exchange.[188,189]
The alginate-encapsulated HepG2 cells or C3A cells in a fluidized bed bioreactor can maintain viability, as well as metabolic, synthetic, and detoxification activities in human plasma after liver failure. Therefore, the system can be improved to form the cellular component of a BAL.[190,191]The BAL system with alginate-encapsulated HepG2 cells could improve systemic parameters of liver failure in a rabbit model, indicating that the encapsulated cells have the potential to develop the BAL for acute liver failure.[192]Moreover, BAL treatment with GS-HepG2 cells could prolong the survival time and improve pathophysiological parameters, such as blood ammonia level and coagulation indices in pigs with ischemic liver failure.[193,194]Another study reported that BAL with FLC-4 cells can prevent irreversible brain damage from hepatic encephalopathy in mini-pigs with acute liver failure.[195]
As previously reported, the immortalized human fetal hepatocyte line cBAL111 could eliminate ammonia, galactose, and lidocaine, and produce albumin when cultured inside the AMC-BAL bioreactor.[196]Importantly, the incorporation of BAL with the reversibly immortalized human hepatocyte line HepLi-4 could improve the Fischer index and serum indirect bilirubin level, and prolong the survival time in pigs with liver failure compared with the liver failure and sham BAL groups.[101]
Human adult liver stem cells which were successfully expanded in a rotary bioreactor perfusion system could be differentiated into mature hepatocyte-like cells. Thus, the rotary BAL incorporating adult liver progenitor cells can be recognized as a potential strategy for BAL.[125]The HepaRG cell line, a liver progenitor cell line, can be differentiated into hepatocyte clusters and it is surrounded by biliary epithelial-like cells at high density after exposure to DMSO, indicating that HepaRG cells are suitable for BAL application.[127]
Problems and prospects
Several cell types as cell sources of BAL have been previously used in clinical trials, including primary porcine hepatocytes,[23,27]primary human hepatocytes,[26]and the liver cell line C3A.[21,73]However, no BAL has been approved by the government for liver failure in clinic. Given the disadvantages of these three cell types, genetically engineered liver cell lines and immortalized human hepatocytes have been successfully used in the animal liver failure model in recent years,[101,194]indicating that they serve as the continuous cell source of metabolically active hepatocytes for BAL.
Aside from genetically engineered liver cells and immortalized human hepatocytes, stem cells (e.g., human ES cells, LPC, and iPS cells) with self-renewal and potential of differentiation into hepatocytes are also promising as sources for BAL. Importantly, mature hepatocyte-like cells from human adult liver stem cells could be expanded in a large quantity for BAL as suitable cell sources.[125]However, a number of problems need to be solved before these cells substitute the presently used cell sources in BAL, including optimization of differentiation protocols.[129,197]
Modified bioreactors mimicking the liver architecture should also be developed to provide a stable long-term culture microenvironment for the improvement of the function of hepatocytes from human liver donors and stem cells. Given the rapid advances in sources of hepatocytes, new-generation BAL with novel and effective liver cells may be achieved for the patients with liver failure in the near future.
Contributors:PXP wrote the article. LLJ proposed and revised the review. LLJ is the guarantor.
Funding:The work was supported by grants from the Chinese High-Tech Research & Development (863) Program (2011AA020104), Science Fund for Creative Research Groups of the National Natural Science Foundation of China (81121002), the Fundamental Research Funds for the Central Universities, and the Technology Group Project for Infectious Disease Control of Zhejiang Province (2009R50041).
Ethical approval:Not needed.
Competing interest:No benefits in any form have been received or will be received from a commercial party related directly or indirectly to the subject of this article.
1 Court FG, Wemyss-Holden SA, Dennison AR, Maddern GJ. Bioartificial liver support devices: historical perspectives. ANZ J Surg 2003;73:739-748.
2 Mas A, Rodés J. Fulminant hepatic failure. Lancet 1997;349: 1081-1085.
3 Michalopoulos GK, DeFrances MC. Liver regeneration. Science 1997;276:60-66.
4 Columbano A, Shinozuka H. Liver regeneration versus direct hyperplasia. FASEB J 1996;10:1118-1128.
5 Steer CJ. Liver regeneration. FASEB J 1995;9:1396-1400.
6 Du WB, Li LJ, Huang JR, Yang Q, Liu XL, Li J, et al. Effects of artificial liver support system on patients with acute or chronic liver failure. Transplant Proc 2005;37:4359-4364.
7 Laleman W, Wilmer A, Evenepoel P, Elst IV, Zeegers M, Zaman Z, et al. Effect of the molecular adsorbent recirculating system and Prometheus devices on systemic haemodynamics and vasoactive agents in patients with acuteon-chronic alcoholic liver failure. Crit Care 2006;10:R108.
8 Hassanein TI, Tofteng F, Brown RS Jr, McGuire B, Lynch P, Mehta R, et al. Randomized controlled study of extracorporeal albumin dialysis for hepatic encephalopathy in advanced cirrhosis. Hepatology 2007;46:1853-1862.
9 Carpentier B, Gautier A, Legallais C. Artificial and bioartificial liver devices: present and future. Gut 2009;58:1690-1702.
10 Allen JW, Hassanein T, Bhatia SN. Advances in bioartificial liver devices. Hepatology 2001;34:447-455.
11 Kumar A, Tripathi A, Jain S. Extracorporeal bioartificial liver for treating acute liver diseases. J Extra Corpor Technol 2011; 43:195-206.
12 Fisher RA, Strom SC. Human hepatocyte transplantation: worldwide results. Transplantation 2006;82:441-449.
13 Pietrosi G, Vizzini GB, Gruttadauria S, Gridelli B. Clinical applications of hepatocyte transplantation. World J Gastroenterol 2009;15:2074-2077.
14 Bilir BM, Guinette D, Karrer F, Kumpe DA, Krysl J, Stephens J, et al. Hepatocyte transplantation in acute liver failure. Liver Transpl 2000;6:32-40.
15 Sgroi A, Serre-Beinier V, Morel P, Bühler L. What clinical alternatives to whole liver transplantation? Current status of artificial devices and hepatocyte transplantation. Transplantation 2009;87:457-466.
16 Mitzner SR, Stange J, Klammt S, Risler T, Erley CM, Bader BD, et al. Improvement of hepatorenal syndrome with extracorporeal albumin dialysis MARS: results of a prospective, randomized, controlled clinical trial. Liver Transpl 2000;6:277-286.
17 Stadlbauer V, Krisper P, Aigner R, Haditsch B, Jung A, Lackner C, et al. Effect of extracorporeal liver support by MARS and Prometheus on serum cytokines in acute-onchronic liver failure. Crit Care 2006;10:R169.
18 Abe T, Kobata H, Hanba Y, Kitabata Y, Narukawa N, Hasegawa H, et al. Study of plasma exchange for liver failure: beneficial and harmful effects. Ther Apher Dial 2004;8:180-184.
19 Yoshiba M, Sekiyama K, Iwamura Y, Sugata F. Development of reliable artificial liver support (ALS)--plasma exchange in combination with hemodiafiltration using high-performance membranes. Dig Dis Sci 1993;38:469-476.
20 Inoue K, Kourin A, Watanabe T, Yamada M, Yasuda H, Yoshiba M. Plasma exchange in combination with onlinehemodiafiltration as a promising method for purifying the blood of fulminant hepatitis patients. Hepatol Res 2008;38: S46-51.
21 Ellis AJ, Hughes RD, Wendon JA, Dunne J, Langley PG, Kelly JH, et al. Pilot-controlled trial of the extracorporeal liver assist device in acute liver failure. Hepatology 1996;24:1446-1451.
22 Mazariegos GV, Kramer DJ, Lopez RC, Shakil AO, Rosenbloom AJ, DeVera M, et al. Safety observations in phase I clinical evaluation of the Excorp Medical Bioartificial Liver Support System after the first four patients. ASAIO J 2001;47:471-475.
23 van de Kerkhove MP, Di Florio E, Scuderi V, Mancini A, Belli A, Bracco A, et al. Phase I clinical trial with the AMC-bioartificial liver. Int J Artif Organs 2002;25:950-959.
24 Sauer IM, Kardassis D, Zeillinger K, Pascher A, Gruenwald A, Pless G, et al. Clinical extracorporeal hybrid liver support--phase I study with primary porcine liver cells. Xenotransplantation 2003;10:460-469.
25 Morsiani E, Pazzi P, Puviani AC, Brogli M, Valieri L, Gorini P, et al. Early experiences with a porcine hepatocyte-based bioartificial liver in acute hepatic failure patients. Int J Artif Organs 2002;25:192-202.
26 Sauer IM, Zeilinger K, Obermayer N, Pless G, Grünwald A, Pascher A, et al. Primary human liver cells as source for modular extracorporeal liver support--a preliminary report. Int J Artif Organs 2002;25:1001-1005.
27 Demetriou AA, Brown RS Jr, Busuttil RW, Fair J, McGuire BM, Rosenthal P, et al. Prospective, randomized, multicenter, controlled trial of a bioartificial liver in treating acute liver failure. Ann Surg 2004;239:660-670.
28 Qian Y, Lanjuan L, Jianrong H, Jun L, Hongcui C, Suzhen F, et al. Study of severe hepatitis treated with a hybrid artificial liver support system. Int J Artif Organs 2003;26:507-513.
29 Snykers S, De Kock J, Rogiers V, Vanhaecke T.In vitrodifferentiation of embryonic and adult stem cells into hepatocytes: state of the art. Stem Cells 2009;27:577-605.
30 Shiota G, Yasui T. Progress in stem cell biology in regenerative medicine for liver disease. Hepatol Res 2012;42:15-21.
31 Basma H, Soto-Gutiérrez A, Yannam GR, Liu L, Ito R, Yamamoto T, et al. Differentiation and transplantation of human embryonic stem cell-derived hepatocytes. Gastroenterology 2009;136:990-999.
32 Wu DC, Boyd AS, Wood KJ. Embryonic stem cell transplantation: potential applicability in cell replacement therapy and regenerative medicine. Front Biosci 2007;12:4525-4535.
33 Woo DH, Kim SK, Lim HJ, Heo J, Park HS, Kang GY, et al. Direct and indirect contribution of human embryonic stem cell-derived hepatocyte-like cells to liver repair in mice. Gastroenterology 2012;142:602-611.
34 Si-Tayeb K, Noto FK, Nagaoka M, Li J, Battle MA, Duris C, et al. Highly efficient generation of human hepatocytelike cells from induced pluripotent stem cells. Hepatology 2010;51:297-305.
35 Song Z, Cai J, Liu Y, Zhao D, Yong J, Duo S, et al. Efficient generation of hepatocyte-like cells from human induced pluripotent stem cells. Cell Res 2009;19:1233-1242.
36 Zhang W, Li W, Liu B, Wang P, Li W, Zhang H. Efficient generation of functional hepatocyte-like cells from human fetal hepatic progenitor cellsin vitro. J Cell Physiol 2012;227: 2051-2058.
37 Aurich H, Sgodda M, Kaltwasser P, Vetter M, Weise A, Liehr T, et al. Hepatocyte differentiation of mesenchymal stem cells from human adipose tissuein vitropromotes hepatic integrationin vivo. Gut 2009;58:570-581.
38 Dalgetty DM, Medine CN, Iredale JP, Hay DC. Progress and future challenges in stem cell-derived liver technologies. Am J Physiol Gastrointest Liver Physiol 2009;297:G241-248.
39 Abdallah BM, Kassem M. Human mesenchymal stem cells: from basic biology to clinical applications. Gene Ther 2008; 15:109-116.
40 Barry FP, Murphy JM. Mesenchymal stem cells: clinical applications and biological characterization. Int J Biochem Cell Biol 2004;36:568-584.
41 Yu CB, Pan XP, Li LJ. Progress in bioreactors of bioartificial livers. Hepatobiliary Pancreat Dis Int 2009;8:134-140.
42 Liu J, Pan J, Naik S, Santangini H, Trenkler D, Thompson N, et al. Characterization and evaluation of detoxification functions of a nontumorigenic immortalized porcine hepatocyte cell line (HepLiu). Cell Transplant 1999;8:219-232.
43 Meng FY, Chen ZS, Han M, Hu XP, He XX, Liu Y, et al. Porcine hepatocyte isolation and reversible immortalization mediated by retroviral transfer and site-specific recombination. World J Gastroenterol 2010;16:1660-1664.
44 Pan X, Du W, Yu X, Sheng G, Cao H, Yu C, et al. Establishment and characterization of immortalized porcine hepatocytes for the study of hepatocyte xenotransplantation. Transplant Proc 2010;42:1899-1906.
45 Specke V, Rubant S, Denner J. Productive infection of human primary cells and cell lines with porcine endogenous retroviruses. Virology 2001;285:177-180.
46 Martin U, Winkler ME, Id M, Radeke H, Arseniev L, Takeuchi Y, et al. Productive infection of primary human endothelial cells by pig endogenous retrovirus (PERV). Xenotransplantation 2000;7:138-142.
47 Martin U, Kiessig V, Blusch JH, Haverich A, von der Helm K, Herden T, et al. Expression of pig endogenous retrovirus by primary porcine endothelial cells and infection of human cells. Lancet 1998;352:692-694.
48 Irgang M, Sauer IM, Karlas A, Zeilinger K, Gerlach JC, Kurth R, et al. Porcine endogenous retroviruses: no infection in patients treated with a bioreactor based on porcine liver cells. J Clin Virol 2003;28:141-154.
49 Di Nicuolo G, van de Kerkhove MP, Hoekstra R, Beld MG, Amoroso P, Battisti S, et al. No evidence ofin vitroandin vivoporcine endogenous retrovirus infection after plasmapheresis through the AMC-bioartificial liver. Xenotransplantation 2005;12:286-292.
50 van de Kerkhove MP, Di Florio E, Scuderi V, Mancini A, Belli A, Bracco A, et al. Bridging a patient with acute liver failure to liver transplantation by the AMC-bioartificial liver. Cell Transplant 2003;12:563-568.
51 Di Nicuolo G, D'Alessandro A, Andria B, Scuderi V, Scognamiglio M, Tammaro A, et al. Long-term absence of porcine endogenous retrovirus infection in chronically immunosuppressed patients after treatment with the porcine cell-based Academic Medical Center bioartificial liver. Xenotransplantation 2010;17:431-439.
52 Frühauf JH, Mertsching H, Giri S, Frühauf NR, Bader A. Porcine endogenous retrovirus released by a bioartificial liver infects primary human cells. Liver Int 2009;29:1553-1561.
53 Yang Q, Liu F, Pan XP, Lv G, Zhang A, Yu CB, et al. Fluidizedbed bioartificial liver assist devices (BLADs) based on microencapsulated primary porcine hepatocytes have risk of porcine endogenous retroviruses transmission. Hepatol Int 2010;4:757-761.
54 Chamuleau RA, Poyck PP, van de Kerkhove MP. Bioartificial liver: its pros and cons. Ther Apher Dial 2006;10:168-174.
55 Baccarani U, Sanna A, Cariani A, Sainz-Barriga M, Adani GL, Zambito AM, et al. Isolation of human hepatocytes from livers rejected for liver transplantation on a national basis: results of a 2-year experience. Liver Transpl 2003;9:506-512.
56 Terry C, Mitry RR, Lehec SC, Muiesan P, Rela M, Heaton ND, et al. The effects of cryopreservation on human hepatocytes obtained from different sources of liver tissue. Cell Transplant 2005;14:585-594.
57 Hughes RD, Mitry RR, Dhawan A, Lehec SC, Girlanda R, Rela M, et al. Isolation of hepatocytes from livers from nonheart-beating donors for cell transplantation. Liver Transpl 2006;12:713-717.
58 Hang H, Shi X, Gu G, Wu Y, Ding Y. A simple isolation and cryopreservation method for adult human hepatocytes. Int J Artif Organs 2009;32:720-727.
59 Alexandrova K, Griesel C, Barthold M, Heuft HG, Ott M, Winkler M, et al. Large-scale isolation of human hepatocytes for therapeutic application. Cell Transplant 2005;14:845-853.
60 Page JL, Johnson MC, Olsavsky KM, Strom SC, Zarbl H, Omiecinski CJ. Gene expression profiling of extracellular matrix as an effector of human hepatocyte phenotype in primary cell culture. Toxicol Sci 2007;97:384-397.
61 Lang R, Stern MM, Smith L, Liu Y, Bharadwaj S, Liu G, et al. Three-dimensional culture of hepatocytes on porcine liver tissue-derived extracellular matrix. Biomaterials 2011;32: 7042-7052.
62 Török E, Lutgehetmann M, Bierwolf J, Melbeck S, Düllmann J, Nashan B, et al. Primary human hepatocytes on biodegradable poly(l-lactic acid) matrices: a promising model for improving transplantation efficiency with tissue engineering. Liver Transpl 2011;17:104-114.
63 Goral VN, Hsieh YC, Petzold ON, Clark JS, Yuen PK, Faris RA. Perfusion-based microfluidic device for threedimensional dynamic primary human hepatocyte cell culture in the absence of biological or synthetic matrices or coagulants. Lab Chip 2010;10:3380-3386.
64 Nishimura M, Hagi M, Ejiri Y, Kishimoto S, Horie T, Narimatsu S, et al. Secretion of albumin and induction of CYP1A2 and CYP3A4 in novel three-dimensional culture system for human hepatocytes using micro-space plate. Drug Metab Pharmacokinet 2010;25:236-242.
65 Bhatia SN, Balis UJ, Yarmush ML, Toner M. Effect of cellcell interactions in preservation of cellular phenotype: cocultivation of hepatocytes and nonparenchymal cells. FASEB J 1999;13:1883-1900.
66 Salerno S, Campana C, Morelli S, Drioli E, De Bartolo L. Human hepatocytes and endothelial cells in organotypic membrane systems. Biomaterials 2011;32:8848-8859.
67 Soto-Gutierrez A, Navarro-Alvarez N, Yagi H, Nahmias Y, Yarmush ML, Kobayashi N. Engineering of an hepatic organoid to develop liver assist devices. Cell Transplant 2010; 19:815-822.
68 Okamoto M, Ishida Y, Keogh A, Strain A. Evaluation of the function of primary human hepatocytes co-cultured with the human hepatic stellate cell (HSC) line LI90. Int J Artif Organs 1998;21:353-359.
69 Auth MK, Woitaschek D, Beste M, Schreiter T, Kim HS, Oppermann E, et al. Preservation of the synthetic and metabolic capacity of isolated human hepatocytes by coculture with human biliary epithelial cells. Liver Transpl 2005;11:410-419.
70 Gómez-Aristizábal A, Keating A, Davies JE. Mesenchymal stromal cells as supportive cells for hepatocytes. Mol Ther 2009;17:1504-1508.
71 Salerno S, Piscioneri A, Laera S, Morelli S, Favia P, Bader A, et al. Improved functions of human hepatocytes on NH3 plasma-grafted PEEK-WC-PU membranes. Biomaterials 2009;30:4348-4356.
72 De Bartolo L, Morelli S, Gallo MC, Campana C, Statti G, Rende M, et al. Effect of isoliquiritigenin on viability and differentiated functions of human hepatocytes maintained on PEEK-WC-polyurethane membranes. Biomaterials 2005; 26:6625-6634.
73 Millis JM, Cronin DC, Johnson R, Conjeevaram H, Conlin C, Trevino S, et al. Initial experience with the modified extracorporeal liver-assist device for patients with fulminant hepatic failure: system modifications and clinical impact. Transplantation 2002;74:1735-1746.
74 Ito A, Jitsunobu H, Kawabe Y, Kamihira M. Construction of heterotypic cell sheets by magnetic force-based 3-D coculture of HepG2 and NIH3T3 cells. J Biosci Bioeng 2007;104:371-378.
75 Ohno M, Motojima K, Okano T, Taniguchi A. Up-regulation of drug-metabolizing enzyme genes in layered co-culture of a human liver cell line and endothelial cells. Tissue Eng Part A 2008;14:1861-1869.
76 Ohno M, Motojima K, Okano T, Taniguchi A. Maturation of the extracellular matrix and cell adhesion molecules in layered co-cultures of HepG2 and endothelial cells. J Biochem 2009;145:591-597.
77 Ohno M, Motojima K, Okano T, Taniguchi A. Induction of drug-metabolizing enzymes by phenobarbital in layered coculture of a human liver cell line and endothelial cells. Biol Pharm Bull 2009;32:813-817.
78 Kinasiewicz A, Gautier A, Lewiska D, Smietanka A, Legallais C, Weryński A. Three-dimensional growth of human hepatoma C3A cells within alginate beads for fluidized bioartificial liver. Int J Artif Organs 2008;31:340-347.
79 Gautier A, Carpentier B, Dufresne M, Vu Dinh Q, Paullier P, Legallais C. Impact of alginate type and bead diameter on mass transfers and the metabolic activities of encapsulated C3A cells in bioartificial liver applications. Eur Cell Mater2011;21:94-106.
80 Laurent T, Murase D, Tsukioka S, Matsuura T, Nagamori S, Oda H. A novel human hepatoma cell line, FLC-4, exhibits highly enhanced liver differentiation functions through the three-dimensional cell shape. J Cell Physiol 2012;227:2898-2906.
81 Gotoh Y, Ishizuka Y, Matsuura T, Niimi S. Spheroid formation and expression of liver-specific functions of human hepatocellular carcinoma-derived FLC-4 cells cultured in lactose-silk fibroin conjugate sponges. Biomacromolecules 2011;12:1532-1539.
82 Enosawa S, Miyashita T, Suzuki S, Li XK, Tsunoda M, Amemiya H, et al. Long-term culture of glutamine synthetase-transfected HepG2 cells in circulatory flow bioreactor for development of a bioartificial liver. Cell Transplant 2000;9:711-715.
83 Tang NH, Wang XQ, Li XJ, Chen YL. Ammonia metabolism capacity of HepG2 cells with high expression of human glutamine synthetase. Hepatobiliary Pancreat Dis Int 2008;7: 621-627.
84 Omasa T, Kim K, Hiramatsu S, Katakura Y, Kishimoto M, Enosawa S, et al. Construction and evaluation of drugmetabolizing cell line for bioartificial liver support system. Biotechnol Prog 2005;21:161-167.
85 Wang N, Tsuruoka S, Yamamoto H, Enosawa S, Omasa T, Sata N, et al. The bioreactor with CYP3A4- and glutamine synthetase-introduced HepG2 cells: treatment of hepatic failure dog with diazepam overdosage. Artif Organs 2005;29: 681-684.
86 Terada S, Kumagai T, Yamamoto N, Ogawa A, Ishimura J, Fujita T, et al. Generation of a novel apoptosis-resistant hepatoma cell line. J Biosci Bioeng 2003;95:146-151.
87 Tang N, Wang Y, Wang X, Zhou L, Zhang F, Li X, et al. Stable overexpression of arginase I and ornithine transcarbamylase in HepG2 cells improves its ammonia detoxification. J Cell Biochem 2012;113:518-527.
88 Nibourg GA, Huisman MT, van der Hoeven TV, van Gulik TM, Chamuleau RA, Hoekstra R. Stable overexpression of pregnane X receptor in HepG2 cells increases its potential for bioartificial liver application. Liver Transpl 2010;16:1075-1085.
89 Naiki T, Nagaki M, Shidoji Y, Kojima H, Moriwaki H. Functional activity of human hepatoma cells transfected with adenovirus-mediated hepatocyte nuclear factor (HNF)-4 gene. Cell Transplant 2004;13:393-403.
90 Harimoto N, Taketomi A, Kitagawa D, Kuroda Y, Itoh S, Gion T, et al. The newly established human hepatocyte cell line: application for the bioartificial liver. J Hepatol 2005;42:557-564.
91 Li J, Li LJ, Cao HC, Sheng GP, Yu HY, Xu W, et al. Establishment of highly differentiated immortalized human hepatocyte line with simian virus 40 large tumor antigen for liver based cell therapy. ASAIO J 2005;51:262-268.
92 Tsuruga Y, Kiyono T, Matsushita M, Takahashi T, Kasai H, Matsumoto S, et al. Establishment of immortalized human hepatocytes by introduction of HPV16 E6/E7 and hTERT as cell sources for liver cell-based therapy. Cell Transplant 2008; 17:1083-1094.
93 Kobayashi N, Miyazaki M, Fukaya K, Inoue Y, Sakaguchi M, Noguchi H, et al. Treatment of surgically induced acute liver failure with transplantation of highly differentiated immortalized human hepatocytes. Cell Transplant 2000;9: 733-735.
94 Chen Y, Li J, Liu X, Zhao W, Wang Y, Wang X. Transplantation of immortalized human fetal hepatocytes prevents acute liver failure in 90% hepatectomized mice. Transplant Proc 2010;42:1907-1914.
95 Deurholt T, van Til NP, Chhatta AA, ten Bloemendaal L, Schwartlander R, Payne C, et al. Novel immortalized human fetal liver cell line, cBAL111, has the potential to differentiate into functional hepatocytes. BMC Biotechnol 2009;9:89.
96 Smalley M, Leiper K, Tootle R, McCloskey P, O'Hare MJ, Hodgson H. Immortalization of human hepatocytes by temperature-sensitive SV40 large-T antigen.In VitroCell Dev Biol Anim 2001;37:166-168.
97 Kobayashi N, Miyazaki M, Westerman KA, Noguchi H, Sakaguchi M, Totsugawa T, et al. Construction of a differentiated human hepatocyte cell line expressing the herpes simplex virus-thymidine kinase gene. ASAIO J 2001; 47:476-480.
98 Totsugawa T, Yong C, Rivas-Carrillo JD, Soto-Gutierrez A, Navarro-Alvarez N, Noguchi H, et al. Survival of liver failure pigs by transplantation of reversibly immortalized human hepatocytes with Tamoxifen-mediated self-recombination. J Hepatol 2007;47:74-82.
99 Nguyen TH, Mai G, Villiger P, Oberholzer J, Salmon P, Morel P, et al. Treatment of acetaminophen-induced acute liver failure in the mouse with conditionally immortalized human hepatocytes. J Hepatol 2005;43:1031-1037.
100 Kobayashi N, Fujiwara T, Westerman KA, Inoue Y, Sakaguchi M, Noguchi H, et al. Prevention of acute liver failure in rats with reversibly immortalized human hepatocytes. Science 2000;287:1258-1262.
101 Zhao L, Li J, Lv G, Zhang A, Zhou P, Yang Y, et al. Evaluation of a reversibly immortalized human hepatocyte line in bioartificial liver in pigs. Afr J Biotechnol 2012;11:4116-4126.
102 Yu CB, Lv GL, Pan XP, Chen YS, Cao HC, Zhang YM, et al.In vitrolarge-scale cultivation and evaluation of microencapsulated immortalized human hepatocytes (HepLL) in roller bottles. Int J Artif Organs 2009;32:272-281.
103 Watanabe T, Shibata N, Westerman KA, Okitsu T, Allain JE, Sakaguchi M, et al. Establishment of immortalized human hepatic stellate scavenger cells to develop bioartificial livers. Transplantation 2003;75:1873-1880.
104 Thomson JA, Itskovitz-Eldor J, Shapiro SS, Waknitz MA, Swiergiel JJ, Marshall VS, et al. Embryonic stem cell lines derived from human blastocysts. Science 1998;282:1145-1147.
105 Reubinoff BE, Pera MF, Fong CY, Trounson A, Bongso A. Embryonic stem cell lines from human blastocysts: somatic differentiationin vitro. Nat Biotechnol 2000;18:399-404.
106 Behbahan IS, Duan Y, Lam A, Khoobyari S, Ma X, Ahuja TP, et al. New approaches in the differentiation of human embryonic stem cells and induced pluripotent stem cells toward hepatocytes. Stem Cell Rev 2011;7:748-759.
107 Rambhatla L, Chiu CP, Kundu P, Peng Y, Carpenter MK. Generation of hepatocyte-like cells from human embryonic stem cells. Cell Transplant 2003;12:1-11.
108 Takayama K, Inamura M, Kawabata K, Katayama K, Higuchi M, Tashiro K, et al. Efficient generation of functional hepatocytes from human embryonic stem cells and inducedpluripotent stem cells by HNF4α transduction. Mol Ther 2012;20:127-137.
109 Duan Y, Ma X, Zou W, Wang C, Bahbahan IS, Ahuja TP, et al. Differentiation and characterization of metabolically functioning hepatocytes from human embryonic stem cells. Stem Cells 2010;28:674-686.
110 Hay DC, Zhao D, Fletcher J, Hewitt ZA, McLean D, Urruticoechea-Uriguen A, et al. Efficient differentiation of hepatocytes from human embryonic stem cells exhibiting markers recapitulating liver developmentin vivo. Stem Cells 2008;26:894-902.
111 Touboul T, Hannan NR, Corbineau S, Martinez A, Martinet C, Branchereau S, et al. Generation of functional hepatocytes from human embryonic stem cells under chemically defined conditions that recapitulate liver development. Hepatology 2010;51:1754-1765.
112 Nagamoto Y, Tashiro K, Takayama K, Ohashi K, Kawabata K, Sakurai F, et al. The promotion of hepatic maturation of human pluripotent stem cells in 3D co-culture using type I collagen and Swiss 3T3 cell sheets. Biomaterials 2012;33: 4526-4534.
113 Baharvand H, Hashemi SM, Kazemi Ashtiani S, Farrokhi A. Differentiation of human embryonic stem cells into hepatocytes in 2D and 3D culture systemsin vitro. Int J Dev Biol 2006;50:645-652.
114 Sharma R, Greenhough S, Medine CN, Hay DC. Threedimensional culture of human embryonic stem cell derived hepatic endoderm and its role in bioartificial liver construction. J Biomed Biotechnol 2010;2010:236147.
115 Miki T, Ring A, Gerlach J. Hepatic differentiation of human embryonic stem cells is promoted by three-dimensional dynamic perfusion culture conditions. Tissue Eng Part C Methods 2011;17:557-568.
116 Farzaneh Z, Pournasr B, Ebrahimi M, Aghdami N, Baharvand H. Enhanced functions of human embryonic stem cell-derived hepatocyte-like cells on three-dimensional nanofibrillar surfaces. Stem Cell Rev 2010;6:601-610.
117 Schmelzer E, Wauthier E, Reid LM. The phenotypes of pluripotent human hepatic progenitors. Stem Cells 2006;24: 1852-1858.
118 Fausto N. Liver regeneration and repair: hepatocytes, progenitor cells, and stem cells. Hepatology 2004;39:1477-1487.
119 Alison MR, Vig P, Russo F, Bigger BW, Amofah E, Themis M, et al. Hepatic stem cells: from inside and outside the liver? Cell Prolif 2004;37:1-21.
120 Malhi H, Irani AN, Gagandeep S, Gupta S. Isolation of human progenitor liver epithelial cells with extensive replication capacity and differentiation into mature hepatocytes. J Cell Sci 2002;115:2679-2688.
121 Wang P, Zhang H, Li W, Zhao Y, An W. Promoter-defined isolation and identification of hepatic progenitor cells from the human fetal liver. Histochem Cell Biol 2008;130:375-385.
122 Duret C, Gerbal-Chaloin S, Ramos J, Fabre JM, Jacquet E, Navarro F, et al. Isolation, characterization, and differentiation to hepatocyte-like cells of nonparenchymal epithelial cells from adult human liver. Stem Cells 2007;25: 1779-1790.
123 Herrera MB, Bruno S, Buttiglieri S, Tetta C, Gatti S, Deregibus MC, et al. Isolation and characterization of a stem cell population from adult human liver. Stem Cells 2006;24:2840-2850.
124 Stachelscheid H, Urbaniak T, Ring A, Spengler B, Gerlach JC, Zeilinger K. Isolation and characterization of adult human liver progenitors from ischemic liver tissue derived from therapeutic hepatectomies. Tissue Eng Part A 2009;15:1633-1643.
125 Fonsato V, Herrera MB, Buttiglieri S, Gatti S, Camussi G, Tetta C. Use of a rotary bioartificial liver in the differentiation of human liver stem cells. Tissue Eng Part C Methods 2010;16:123-132.
126 Parent R, Marion MJ, Furio L, Trépo C, Petit MA. Origin and characterization of a human bipotent liver progenitor cell line. Gastroenterology 2004;126:1147-1156.
127 Hoekstra R, Nibourg GA, van der Hoeven TV, Ackermans MT, Hakvoort TB, van Gulik TM, et al. The HepaRG cell line is suitable for bioartificial liver application. Int J Biochem Cell Biol 2011;43:1483-1489.
128 Zaehres H, Schöler HR. Induction of pluripotency: from mouse to human. Cell 2007;131:834-835.
129 Asgari S, Pournasr B, Salekdeh GH, Ghodsizadeh A, Ott M, Baharvand H. Induced pluripotent stem cells: a new era for hepatology. J Hepatol 2010;53:738-751.
130 Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T, Tomoda K, et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 2007;131: 861-872.
131 Park IH, Zhao R, West JA, Yabuuchi A, Huo H, Ince TA, et al. Reprogramming of human somatic cells to pluripotency with defined factors. Nature 2008;451:141-146.
132 Yu J, Vodyanik MA, Smuga-Otto K, Antosiewicz-Bourget J, Frane JL, Tian S, et al. Induced pluripotent stem cell lines derived from human somatic cells. Science 2007;318:1917-1920.
133 Maekawa M, Yamaguchi K, Nakamura T, Shibukawa R, Kodanaka I, Ichisaka T, et al. Direct reprogramming of somatic cells is promoted by maternal transcription factor Glis1. Nature 2011;474:225-229.
134 Inamura M, Kawabata K, Takayama K, Tashiro K, Sakurai F, Katayama K, et al. Efficient generation of hepatoblasts from human ES cells and iPS cells by transient overexpression of homeobox gene HEX. Mol Ther 2011;19:400-407.
135 Sullivan GJ, Hay DC, Park IH, Fletcher J, Hannoun Z, Payne CM, et al. Generation of functional human hepatic endoderm from human induced pluripotent stem cells. Hepatology 2010;51:329-335.
136 Chen YF, Tseng CY, Wang HW, Kuo HC, Yang VW, Lee OK. Rapid generation of mature hepatocyte-like cells from human induced pluripotent stem cells by an efficient threestep protocol. Hepatology 2012;55:1193-1203.
137 Takayama K, Inamura M, Kawabata K, Tashiro K, Katayama K, Sakurai F, et al. Efficient and directive generation of two distinct endoderm lineages from human ESCs and iPSCs by differentiation stage-specific SOX17 transduction. PLoS One 2011;6:e21780.
138 Asgari S, Moslem M, Bagheri-Lankarani K, Pournasr B, Miryounesi M, Baharvand H. Differentiation and transplantation of human induced pluripotent stem cellderived hepatocyte-like cells. Stem Cell Rev 2011.
139 Kern S, Eichler H, Stoeve J, Klüter H, Bieback K. Comparative analysis of mesenchymal stem cells from bone marrow, umbilical cord blood, or adipose tissue. Stem Cells2006;24:1294-1301.
140 Wagner W, Wein F, Seckinger A, Frankhauser M, Wirkner U, Krause U, et al. Comparative characteristics of mesenchymal stem cells from human bone marrow, adipose tissue, and umbilical cord blood. Exp Hematol 2005;33:1402-1416.
141 Ong SY, Dai H, Leong KW. Hepatic differentiation potential of commercially available human mesenchymal stem cells. Tissue Eng 2006;12:3477-3485.
142 Ayatollahi M, Soleimani M, Tabei SZ, Kabir Salmani M. Hepatogenic differentiation of mesenchymal stem cells induced by insulin like growth factor-I. World J Stem Cells 2011;3:113-121.
143 Pournasr B, Mohamadnejad M, Bagheri M, Aghdami N, Shahsavani M, Malekzadeh R, et al.In vitrodifferentiation of human bone marrow mesenchymal stem cells into hepatocyte-like cells. Arch Iran Med 2011;14:244-249.
144 Lin N, Lin J, Bo L, Weidong P, Chen S, Xu R. Differentiation of bone marrow-derived mesenchymal stem cells into hepatocyte-like cells in an alginate scaffold. Cell Prolif 2010;43:427-434.
145 Ishii K, Yoshida Y, Akechi Y, Sakabe T, Nishio R, Ikeda R, et al. Hepatic differentiation of human bone marrowderived mesenchymal stem cells by tetracycline-regulated hepatocyte nuclear factor 3beta. Hepatology 2008;48:597-606.
146 Kazemnejad S, Allameh A, Seoleimani M, Gharehbaghian A, Mohammadi Y, Amirizadeh N, et al. Functional hepatocytelike cells derived from human bone marrow mesenchymal stem cells on a novel 3-dimensional biocompatible nanofibrous scaffold. Int J Artif Organs 2008;31:500-507.
147 Ghaedi M, Soleimani M, Shabani I, Duan Y, Lotfi AS. Hepatic differentiation from human mesenchymal stem cells on a novel nanofiber scaffold. Cell Mol Biol Lett 2012; 17:89-106.
148 Kazemnejad S, Allameh A, Soleimani M, Gharehbaghian A, Mohammadi Y, Amirizadeh N, 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;24:278-287.
149 Stock P, Brückner S, Ebensing S, Hempel M, Dollinger MM, Christ B. The generation of hepatocytes from mesenchymal stem cells and engraftment into murine liver. Nat Protoc 2010;5:617-627.
150 Schäffler A, Büchler C. Concise review: adipose tissuederived stromal cells--basic and clinical implications for novel cell-based therapies. Stem Cells 2007;25:818-827.
151 Yarak S, Okamoto OK. Human adipose-derived stem cells: current challenges and clinical perspectives. An Bras Dermatol 2010;85:647-656.
152 Coradeghini R, Guida C, Scanarotti C, Sanguineti R, Bassi AM, Parodi A, et al. A comparative study of proliferation and hepatic differentiation of human adipose-derived stem cells. Cells Tissues Organs 2010;191:466-477.
153 Banas A, Teratani T, Yamamoto Y, Tokuhara M, Takeshita F, Quinn G, et al. Adipose tissue-derived mesenchymal stem cells as a source of human hepatocytes. Hepatology 2007;46: 219-228.
154 Taléns-Visconti R, Bonora A, Jover R, Mirabet V, Carbonell F, Castell JV, et al. Hepatogenic differentiation of human mesenchymal stem cells from adipose tissue in comparison with bone marrow mesenchymal stem cells. World J Gastroenterol 2006;12:5834-5845.
155 Lue J, Lin G, Ning H, Xiong A, Lin CS, Glenn JS. Transdifferentiation of adipose-derived stem cells into hepatocytes: a new approach. Liver Int 2010;30:913-922.
156 Seo MJ, Suh SY, Bae YC, Jung JS. Differentiation of human adipose stromal cells into hepatic lineagein vitroandin vivo. Biochem Biophys Res Commun 2005;328:258-264.
157 Wang M, Pei H, Zhang L, Guan L, Zhang R, Jia Y, et al. Hepatogenesis of adipose-derived stem cells on poly-lactideco-glycolide scaffolds:in vitroandin vivostudies. Tissue Eng Part C Methods 2010;16:1041-1050.
158 Matikainen T, Laine J. Placenta--an alternative source of stem cells. Toxicol Appl Pharmacol 2005;207:544-549.
159 Parolini O, Alviano F, Bagnara GP, Bilic G, Bühring HJ, Evangelista M, et al. Concise review: isolation and characterization of cells from human term placenta: outcome of the first international Workshop on Placenta Derived Stem Cells. Stem Cells 2008;26:300-311.
160 Terada S, Matsuura K, Enosawa S, Miki M, Hoshika A, Suzuki S, et al. Inducing proliferation of human amniotic epithelial (HAE) cells for cell therapy. Cell Transplant 2000;9:701-704.
161 Takashima S, Ise H, Zhao P, Akaike T, Nikaido T. Human amniotic epithelial cells possess hepatocyte-like characteristics and functions. Cell Struct Funct 2004;29:73-84.
162 Marongiu F, Gramignoli R, Dorko K, Miki T, Ranade AR, Paola Serra M, et al. Hepatic differentiation of amniotic epithelial cells. Hepatology 2011;53:1719-1729.
163 Kim J, Kang HM, Kim H, Kim MR, Kwon HC, Gye MC, et al. Ex vivo characteristics of human amniotic membranederived stem cells. Cloning Stem Cells 2007;9:581-594.
164 Tamagawa T, Oi S, Ishiwata I, Ishikawa H, Nakamura Y. Differentiation of mesenchymal cells derived from human amniotic membranes into hepatocyte-like cellsin vitro. Hum Cell 2007;20:77-84.
165 Miki T, Marongiu F, Ellis EC, Dorko K, Mitamura K, Ranade A, et al. Production of hepatocyte-like cells from human amnion. Methods Mol Biol 2009;481:155-168.
166 Chien CC, Yen BL, Lee FK, Lai TH, Chen YC, Chan SH, et al.In vitrodifferentiation of human placenta-derived multipotent cells into hepatocyte-like cells. Stem Cells 2006; 24:1759-1768.
167 Brooke G, Rossetti T, Pelekanos R, Ilic N, Murray P, Hancock S, et al. Manufacturing of human placenta-derived mesenchymal stem cells for clinical trials. Br J Haematol 2009;144:571-579.
168 Yu Y, Li K, Bao C, Liu T, Jin Y, Ren H, et al. Ex vitro expansion of human placenta-derived mesenchymal stem cells in stirred bioreactor. Appl Biochem Biotechnol 2009; 159:110-118.
169 Zhang D, Jiang M, Miao D. Transplanted human amniotic membrane-derived mesenchymal stem cells ameliorate carbon tetrachloride-induced liver cirrhosis in mouse. PLoS One 2011;6:e16789.
170 Manuelpillai U, Tchongue J, Lourensz D, Vaghjiani V, Samuel CS, Liu A, et al. Transplantation of human amnion epithelial cells reduces hepatic fibrosis in immunocompetent CCl4-treated mice. Cell Transplant 2010;19:1157-1168.
171 Cao H, Yang J, Yu J, Pan Q, Li J, Zhou P, et al. Therapeuticpotential of transplanted placental mesenchymal stem cells in treating Chinese miniature pigs with acute liver failure. BMC Med 2012;10:56.
172 Mihu CM, Mihu D, Costin N, Rus Ciucă D, Suşman S, Ciortea R. Isolation and characterization of stem cells from the placenta and the umbilical cord. Rom J Morphol Embryol 2008;49:441-446.
173 Lee OK, Kuo TK, Chen WM, Lee KD, Hsieh SL, Chen TH. Isolation of multipotent mesenchymal stem cells from umbilical cord blood. Blood 2004;103:1669-1675.
174 Lee KD, Kuo TK, Whang-Peng J, Chung YF, Lin CT, Chou SH, et al.In vitrohepatic differentiation of human mesenchymal stem cells. Hepatology 2004;40:1275-1284.
175 Zhao Q, Ren H, Li X, Chen Z, Zhang X, Gong W, et al. Differentiation of human umbilical cord mesenchymal stromal cells into low immunogenic hepatocyte-like cells. Cytotherapy 2009;11:414-426.
176 Kang XQ, Zang WJ, Bao LJ, Li DL, Xu XL, Yu XJ. Differentiating characterization of human umbilical cord blood-derived mesenchymal stem cellsin vitro. Cell Biol Int 2006;30:569-575.
177 Hong SH, Gang EJ, Jeong JA, Ahn C, Hwang SH, Yang IH, et al.In vitrodifferentiation of human umbilical cord bloodderived mesenchymal stem cells into hepatocyte-like cells. Biochem Biophys Res Commun 2005;330:1153-1161.
178 Yoshida Y, Shimomura T, Sakabe T, Ishii K, Gonda K, Matsuoka S, et al. A role of Wnt/beta-catenin signals in hepatic fate specification of human umbilical cord bloodderived mesenchymal stem cells. Am J Physiol Gastrointest Liver Physiol 2007;293:G1089-1098.
179 Kang XQ, Zang WJ, Bao LJ, Li DL, Song TS, Xu XL, et al. Fibroblast growth factor-4 and hepatocyte growth factor induce differentiation of human umbilical cord bloodderived mesenchymal stem cells into hepatocytes. World J Gastroenterol 2005;11:7461-7465.
180 Yu J, Cao H, Yang J, Pan Q, Ma J, Li J, et al.In vivohepatic differentiation of mesenchymal stem cells from human umbilical cord blood after transplantation into mice with liver injury. Biochem Biophys Res Commun 2012;422:539-545.
181 Yan Y, Xu W, Qian H, Si Y, Zhu W, Cao H, et al. Mesenchymal stem cells from human umbilical cords ameliorate mouse hepatic injuryin vivo. Liver Int 2009;29:356-365.
182 Jung KH, Shin HP, Lee S, Lim YJ, Hwang SH, Han H, et al. Effect of human umbilical cord blood-derived mesenchymal stem cells in a cirrhotic rat model. Liver Int 2009;29:898-909.
183 Shi LL, Liu FP, Wang DW. Transplantation of human umbilical cord blood mesenchymal stem cells improves survival rates in a rat model of acute hepatic necrosis. Am J Med Sci 2011;342:212-217.
184 Zhang S, Chen L, Liu T, Zhang B, Xiang D, Wang Z, et al. Human umbilical cord matrix stem cells efficiently rescue acute liver failure through paracrine effects rather than hepatic differentiation. Tissue Eng Part A 2012;18:1352-1364.
185 Schmelzer E, Mutig K, Schrade P, Bachmann S, Gerlach JC, Zeilinger K. Effect of human patient plasma ex vivo treatment on gene expression and progenitor cell activation of primary human liver cells in multi-compartment 3D perfusion bioreactors for extra-corporeal liver support. Biotechnol Bioeng 2009;103:817-827.
186 Mueller D, Tascher G, Müller-Vieira U, Knobeloch D, Nuessler AK, Zeilinger K, et al. In-depth physiological characterization of primary human hepatocytes in a 3D hollow-fiber bioreactor. J Tissue Eng Regen Med 2011;5: e207-218.
187 De Bartolo L, Salerno S, Curcio E, Piscioneri A, Rende M, Morelli S, et al. Human hepatocyte functions in a crossed hollow fiber membrane bioreactor. Biomaterials 2009;30: 2531-2543.
188 Gerlach JC, Zeilinger K, Grebe A, Puhl G, Pless G, Sauer I, et al. Recovery of preservation-injured primary human hepatocytes and nonparenchymal cells to tissuelike structures in large-scale bioreactors for liver support: an initial transmission electron microscopy study. J Invest Surg 2003;16:83-92.
189 Gerlach JC, Mutig K, Sauer IM, Schrade P, Efimova E, Mieder T, et al. Use of primary human liver cells originating from discarded grafts in a bioreactor for liver support therapy and the prospects of culturing adult liver stem cells in bioreactors: a morphologic study. Transplantation 2003;76:781-786.
190 Coward SM, Legallais C, David B, Thomas M, Foo Y, Mavri-Damelin D, et al. Alginate-encapsulated HepG2 cells in a fluidized bed bioreactor maintain function in human liver failure plasma. Artif Organs 2009;33:1117-1126.
191 David B, Dufresne M, Nagel MD, Legallais C.In vitroassessment of encapsulated C3A hepatocytes functions in a fluidized bed bioreactor. Biotechnol Prog 2004;20:1204-1212.
192 Rahman TM, Selden C, Khalil M, Diakanov I, Hodgson HJ. Alginate-encapsulated human hepatoblastoma cells in an extracorporeal perfusion system improve some systemic parameters of liver failure in a xenogeneic model. Artif Organs 2004;28:476-482.
193 Enosawa S, Miyashita T, Fujita Y, Suzuki S, Amemiya H, Omasa T, et al.In vivoestimation of bioartificial liver with recombinant HepG2 cells using pigs with ischemic liver failure. Cell Transplant 2001;10:429-433.
194 Enosawa S, Miyashita T, Saito T, Omasa T, Matsumura T. The significant improvement of survival times and pathological parameters by bioartificial liver with recombinant HepG2 in porcine liver failure model. Cell Transplant 2006;15:873-880.
195 Kanai H, Marushima H, Kimura N, Iwaki T, Saito M, Maehashi H, et al. Extracorporeal bioartificial liver using the radial-flow bioreactor in treatment of fatal experimental hepatic encephalopathy. Artif Organs 2007;31:148-151.
196 Poyck PP, van Wijk AC, van der Hoeven TV, de Waart DR, Chamuleau RA, van Gulik TM, et al. Evaluation of a new immortalized human fetal liver cell line (cBAL111) for application in bioartificial liver. J Hepatol 2008;48:266-275.
197 Jung Y, Bauer G, Nolta JA. Concise review: Induced pluripotent stem cell-derived mesenchymal stem cells: progress toward safe clinical products. Stem Cells 2012;30:42-47.
May 5, 2012
Accepted after revision August 30, 2012
Author Affiliations: State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China (Pan XP and Li LJ)
Lan-Juan Li, MD, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China (Tel/Fax: 86-571-87236759; Email: ljli@zju.edu.cn)
© 2012, Hepatobiliary Pancreat Dis Int. All rights reserved.
10.1016/S1499-3872(12)60230-6